201
|
Blood cell-produced amyloid-β induces cerebral Alzheimer-type pathologies and behavioral deficits. Mol Psychiatry 2021; 26:5568-5577. [PMID: 32681097 DOI: 10.1038/s41380-020-0842-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/04/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022]
Abstract
It is traditionally believed that cerebral amyloid-beta (Aβ) deposits are derived from the brain itself in Alzheimer's disease (AD). Peripheral cells such as blood cells also produce Aβ. The role of peripherally produced Aβ in the pathogenesis of AD remains unknown. In this study, we established a bone marrow transplantation model to investigate the contribution of blood cell-produced Aβ to AD pathogenesis. We found that bone marrow cells (BMCs) transplanted from APPswe/PS1dE9 transgenic mice into wild-type (Wt) mice at 3 months of age continuously expressed human Aβ in the blood, and caused AD phenotypes including Aβ plaques, cerebral amyloid angiopathy (CAA), tau hyperphosphorylation, neuronal degeneration, neuroinflammation, and behavioral deficits in the Wt recipient mice at 12 months after transplantation. Bone marrow reconstitution in APPswe/PS1dE9 mice with Wt-BMCs at 3 months of age reduced blood Aβ levels, and alleviated brain Aβ burden, neuronal degeneration, neuroinflammation, and behavioral deficits in the AD model mice at 12 months after transplantation. Our study demonstrated that blood cell-produced Aβ plays a significant role in AD pathogenesis, and the elimination of peripheral production of Aβ can decrease brain Aβ deposition and represents a novel therapeutic approach for AD.
Collapse
|
202
|
Potential Roles of Sestrin2 in Alzheimer's Disease: Antioxidation, Autophagy Promotion, and Beyond. Biomedicines 2021; 9:biomedicines9101308. [PMID: 34680426 PMCID: PMC8533411 DOI: 10.3390/biomedicines9101308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disease. It presents with progressive memory loss, worsens cognitive functions to the point of disability, and causes heavy socioeconomic burdens to patients, their families, and society as a whole. The underlying pathogenic mechanisms of AD are complex and may involve excitotoxicity, excessive generation of reactive oxygen species (ROS), aberrant cell cycle reentry, impaired mitochondrial function, and DNA damage. Up to now, there is no effective treatment available for AD, and it is therefore urgent to develop an effective therapeutic regimen for this devastating disease. Sestrin2, belonging to the sestrin family, can counteract oxidative stress, reduce activity of the mammalian/mechanistic target of rapamycin (mTOR), and improve cell survival. It may therefore play a crucial role in neurodegenerative diseases like AD. However, only limited studies of sestrin2 and AD have been conducted up to now. In this article, we discuss current experimental evidence to demonstrate the potential roles of sestrin2 in treating neurodegenerative diseases, focusing specifically on AD. Strategies for augmenting sestrin2 expression may strengthen neurons, adapting them to stressful conditions through counteracting oxidative stress, and may also adjust the autophagy process, these two effects together conferring neuronal resistance in cases of AD.
Collapse
|
203
|
Takata K, Ginhoux F, Shimohama S. Roles of microglia in Alzheimer's disease and impact of new findings on microglial heterogeneity as a target for therapeutic intervention. Biochem Pharmacol 2021; 192:114754. [PMID: 34480881 DOI: 10.1016/j.bcp.2021.114754] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/25/2022]
Abstract
Microglia are specialized macrophages that reside within the central nervous system and play key roles in brain immunity, development and homeostasis. Recent studies also revealed functions of microglia in neuroprotection and neuroinflammation, leading to the discovery that microglia are involved in several brain pathologies including Alzheimer's disease (AD). However, the beneficial and detrimental actions of this intriguing cell population can be challenging to dissect: the advent of single-cell and single-nucleus transcriptomic technologies has revolutionized our understanding of the heterogeneity of multiple cell types and is now being applied to the study of microglia in health and disease. Here, we review recent findings on microglial biology, focusing on insights from single cell transcriptomic studies and the heterogeneity that they reveal, and consider the impact of these findings on our understanding of AD. We also discuss how microglia might represent a next-generation therapeutic target for treatment of AD and other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Kazuyuki Takata
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Translational Immunology Institute, SingHealth/Duke-NUS, Academic Medical Centre, The Academia, Singapore 169856, Singapore
| | - Shun Shimohama
- Department of Neurology, Sapporo Medical University, School of Medicine, Sapporo 060-8543, Japan
| |
Collapse
|
204
|
Cuestas Torres DM, Cardenas FP. Synaptic plasticity in Alzheimer's disease and healthy aging. Rev Neurosci 2021; 31:245-268. [PMID: 32250284 DOI: 10.1515/revneuro-2019-0058] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
The strength and efficiency of synaptic connections are affected by the environment or the experience of the individual. This property, called synaptic plasticity, is directly related to memory and learning processes and has been modeled at the cellular level. These types of cellular memory and learning models include specific stimulation protocols that generate a long-term strengthening of the synapses, called long-term potentiation, or a weakening of the said long-term synapses, called long-term depression. Although, for decades, researchers have believed that the main cause of the cognitive deficit that characterizes Alzheimer's disease (AD) and aging was the loss of neurons, the hypothesis of an imbalance in the cellular and molecular mechanisms of synaptic plasticity underlying this deficit is currently widely accepted. An understanding of the molecular and cellular changes underlying the process of synaptic plasticity during the development of AD and aging will direct future studies to specific targets, resulting in the development of much more efficient and specific therapeutic strategies. In this review, we classify, discuss, and describe the main findings related to changes in the neurophysiological mechanisms of synaptic plasticity in excitatory synapses underlying AD and aging. In addition, we suggest possible mechanisms in which aging can become a high-risk factor for the development of AD and how its development could be prevented or slowed.
Collapse
Affiliation(s)
- Diana Marcela Cuestas Torres
- Departamento de Psicología and Departamento de Biología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| | - Fernando P Cardenas
- Departamento de Psicología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| |
Collapse
|
205
|
The potential roles of genetic factors in predicting ageing-related cognitive change and Alzheimer's disease. Ageing Res Rev 2021; 70:101402. [PMID: 34242808 DOI: 10.1016/j.arr.2021.101402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/22/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a complex neurological disorder of uncertain aetiology, although substantial research has been conducted to explore important factors related to risk of onset and progression. Both lifestyle (e.g., complex mental stimulation, vascular health) and genetic factors (e.g., APOE, BDNF, PICALM, CLU, APP, PSEN1, PSEN2, and other genes) have been associated with AD risk. Despite more than thirty years of genetic research, much of the heritability of AD is not explained by measured loci. This suggests that the missing heritability of AD might be potentially related to rare variants, gene-environment and gene-gene interactions, and potentially epigenetic modulators. Moreover, while ageing is the most substantial factor risk for AD, there are limited longitudinal studies examining the association of genetic factors with decline in cognitive function due to ageing and the preclinical stages of this condition. This review summarises findings from currently available research on the genetic factors of ageing-related cognitive change and AD and suggests some future research directions.
Collapse
|
206
|
Fuentes M, Schipke CG, Freiesleben SD, Klostermann A, Peters O. Presenilin 1 Gene Mutation (M139V) in a German Family with Early-Onset Alzheimer's Disease: A Case Report. Arch Clin Neuropsychol 2021; 37:521-530. [PMID: 34427587 DOI: 10.1093/arclin/acab070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE This study describes a 44-year-old German male with early-onset Alzheimer's disease as a result of a M139V presenilin 1 mutation. The patient has at least seven affected family members, spanning at least four generations. METHOD We performed a complete demographic, genetic, neuropsychological, neuropsychiatric, neuroradiological, and neuropathological characterizations of this patient. The findings were compared with previous reports of patients with the same mutation. Demographic, neuropsychological, neuropsychiatric, neuroradiological, and neuropathological data from several affected members of the patient's family were also addressed. RESULTS We describe similarities shared with other cases, including age at onset, rapid disease progression, severe deficits in arithmetic and visuo-constructive abilities with relative preservation of naming skills, and the presence of predominant frontal behavioral symptoms. Differences with respect to previously described cases, including the absence of positive neurological or radiological findings, psychotic symptoms, or a depressive disorder, are also identified and discussed. CONCLUSIONS Heterogeneity in symptoms between affected patients from the same or from different families suggests that individual, genetic, or epigenetic factors most likely modulate the phenotype of patients carrying the M139V mutation.
Collapse
Affiliation(s)
- Manuel Fuentes
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Carola G Schipke
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Silka Dawn Freiesleben
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Arne Klostermann
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Oliver Peters
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| |
Collapse
|
207
|
Azam S, Haque ME, Balakrishnan R, Kim IS, Choi DK. The Ageing Brain: Molecular and Cellular Basis of Neurodegeneration. Front Cell Dev Biol 2021; 9:683459. [PMID: 34485280 PMCID: PMC8414981 DOI: 10.3389/fcell.2021.683459] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Ageing is an inevitable event in the lifecycle of all organisms, characterized by progressive physiological deterioration and increased vulnerability to death. Ageing has also been described as the primary risk factor of most neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and frontotemporal lobar dementia (FTD). These neurodegenerative diseases occur more prevalently in the aged populations. Few effective treatments have been identified to treat these epidemic neurological crises. Neurodegenerative diseases are associated with enormous socioeconomic and personal costs. Here, the pathogenesis of AD, PD, and other neurodegenerative diseases has been presented, including a summary of their known associations with the biological hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, mitochondrial dysfunction, cellular senescence, deregulated nutrient sensing, stem cell exhaustion, and altered intercellular communications. Understanding the central biological mechanisms that underlie ageing is important for identifying novel therapeutic targets for neurodegenerative diseases. Potential therapeutic strategies, including the use of NAD+ precursors, mitophagy inducers, and inhibitors of cellular senescence, has also been discussed.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju-si, South Korea
| | - Md. Ezazul Haque
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju-si, South Korea
| | - Rengasamy Balakrishnan
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju-si, South Korea
| | - In-Su Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju-si, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju-si, South Korea
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju-si, South Korea
| |
Collapse
|
208
|
Bai B, Vanderwall D, Li Y, Wang X, Poudel S, Wang H, Dey KK, Chen PC, Yang K, Peng J. Proteomic landscape of Alzheimer's Disease: novel insights into pathogenesis and biomarker discovery. Mol Neurodegener 2021; 16:55. [PMID: 34384464 PMCID: PMC8359598 DOI: 10.1186/s13024-021-00474-z] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 07/18/2021] [Indexed: 12/15/2022] Open
Abstract
Mass spectrometry-based proteomics empowers deep profiling of proteome and protein posttranslational modifications (PTMs) in Alzheimer's disease (AD). Here we review the advances and limitations in historic and recent AD proteomic research. Complementary to genetic mapping, proteomic studies not only validate canonical amyloid and tau pathways, but also uncover novel components in broad protein networks, such as RNA splicing, development, immunity, membrane transport, lipid metabolism, synaptic function, and mitochondrial activity. Meta-analysis of seven deep datasets reveals 2,698 differentially expressed (DE) proteins in the landscape of AD brain proteome (n = 12,017 proteins/genes), covering 35 reported AD genes and risk loci. The DE proteins contain cellular markers enriched in neurons, microglia, astrocytes, oligodendrocytes, and epithelial cells, supporting the involvement of diverse cell types in AD pathology. We discuss the hypothesized protective or detrimental roles of selected DE proteins, emphasizing top proteins in "amyloidome" (all biomolecules in amyloid plaques) and disease progression. Comprehensive PTM analysis represents another layer of molecular events in AD. In particular, tau PTMs are correlated with disease stages and indicate the heterogeneity of individual AD patients. Moreover, the unprecedented proteomic coverage of biofluids, such as cerebrospinal fluid and serum, procures novel putative AD biomarkers through meta-analysis. Thus, proteomics-driven systems biology presents a new frontier to link genotype, proteotype, and phenotype, accelerating the development of improved AD models and treatment strategies.
Collapse
Affiliation(s)
- Bing Bai
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
- Current address: Center for Precision Medicine, Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Jiangsu 210008 Nanjing, China
| | - David Vanderwall
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| | - Yuxin Li
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
- Current address: Department of Biology, University of North Dakota, ND 58202 Grand Forks, USA
| | - Suresh Poudel
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| | - Hong Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| | - Kaushik Kumar Dey
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| | - Ping-Chung Chen
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| | - Ka Yang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 38105 Memphis, TN USA
| |
Collapse
|
209
|
Mao C, Li J, Dong L, Huang X, Lei D, Wang J, Chu S, Liu C, Peng B, Román GC, Cui L, Gao J. Clinical Phenotype and Mutation Spectrum of Alzheimer's Disease with Causative Genetic Mutation in a Chinese Cohort. Curr Alzheimer Res 2021; 18:265-272. [PMID: 34102969 PMCID: PMC8506917 DOI: 10.2174/1567205018666210608120339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/06/2021] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
Background Alzheimer’s disease with a causative genetic mutation (AD-CGM) is an uncommon form, characterized by a heterogeneous clinical phenotype and variations in the genotype of racial groups affected. Objective We aimed to systemically describe the phenotype variance and mutation spectrum in the large sample size of the Peking Union Medical College Hospital (PUMCH) cohort, Beijing, China. Methods Next-generation sequencing (NGS) was carried out in 1108 patients diagnosed with dementia. A total of 40 Han Chinese patients with three AD gene mutations were enrolled. A systemic review of all the patients was performed, including clinical history, neurocognitive assessment, brain magnetic resonance imaging, and cerebrospinal fluid (CSF) biomarkers. Results We studied the following gene mutation variants: 12 AβPP, 13 PSEN1, and 9 PSEN2, and 23 among them were novel. Most of them were early-onset, but PSEN1 mutation carriers had the youngest onset age. The commonest symptoms were similar to those of AD, including an amnestic syndrome, followed by psychiatric symptoms and movement disorder. On MRI, parietal and posterior temporal atrophy was prominent in PSEN1 and PSEN2 mutation carriers, while AβPP mutation carriers had more vascular changes. The CSF biomarkers profile was indistinguishable from sporadic AD. Conclusion We identified a small group of AD-CGM subjects representing 3.6% among more than 1000 demented patients in the PUMCH cohort. These subjects usually presented with early-onset
dementia and exhibited significant clinical and genetic heterogeneity. Identification required complete screening of genetic mutations using NGS. Although family history was usually present, we found non-familial cases of all three genetic mutations.
Collapse
Affiliation(s)
- Chenhui Mao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jie Li
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Liling Dong
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Xinying Huang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Dan Lei
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jie Wang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Shanshan Chu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Caiyan Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Bin Peng
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Gustavo C Román
- Stanley H. Appel Department of Neurology, Nantz National Alzheimer Center, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Jing Gao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
210
|
Pope CA, Wilkins HM, Swerdlow RH, Wolfe MS. Mutations in the Amyloid-β Protein Precursor Reduce Mitochondrial Function and Alter Gene Expression Independent of 42-Residue Amyloid-β Peptide. J Alzheimers Dis 2021; 83:1039-1049. [PMID: 34366346 DOI: 10.3233/jad-210366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Dominant missense mutations in the amyloid-β protein precursor (AβPP) cause early-onset familial Alzheimer's disease (FAD) and are associated with changes in the production or properties of the amyloid-β peptide (Aβ), particularly of the 42-residue variant (Aβ 42) that deposits in the Alzheimer's disease (AD) brain. Recent findings, however, show that FAD mutations in AβPP also lead to increased production of longer Aβ variants of 45-49 residues in length. OBJECTIVE We aimed to test neurotoxicity of Aβ 42 vis-á-vis longer variants, focusing specifically on mitochondrial function, as dysfunctional mitochondria are implicated in the pathogenesis of AD. METHODS We generated SH-SY5Y human neuroblastoma cells stably expressing AβPP mutations that lead to increased production of long Aβ peptides with or without Aβ 42. These AβPP-expressing cells were tested for oxygen consumption rates (OCR) under different conditions designed to interrogate mitochondrial function. These cell lines were also examined for expression of genes important for mitochondrial or neuronal structure and function. RESULTS The mutant AβPP-expressing cells showed decreased basal OCRs as well as decreased OCRs associated with mitochondrial ATP production, even more so in the absence of Aβ 42 production. Moreover, mutant AβPP-expressing cells producing longer forms of Aβ displayed altered expression of certain mitochondrial- and neuronal-associated genes, whether or not Aβ 42 was produced. CONCLUSION These findings suggest that mutant AβPP can cause mitochondrial dysfunction that is associated with long Aβ but not with Aβ 42.
Collapse
Affiliation(s)
- Chad A Pope
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
211
|
Mukhopadhyay S, Banerjee D. A Primer on the Evolution of Aducanumab: The First Antibody Approved for Treatment of Alzheimer's Disease. J Alzheimers Dis 2021; 83:1537-1552. [PMID: 34366359 DOI: 10.3233/jad-215065] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with global burden projected to triple by 2050. It incurs significant biopsychosocial burden worldwide with limited treatment options. Aducanumab is the first monoclonal antibody recently approved by the US-FDA for mild AD through the accelerated approval pathway. It is the first molecule to be approved for AD since 2003 and carries with it a therapeutic promise for the future. As the definition of AD has evolved from a pathological entity to a Clinico-biological construct over the years, the amyloid-β (Aβ) pathway has been increasingly implicated in its pathogenesis. The approval of Aducanumab is based on reduction of the Aβ load in the brain, which forms a surrogate marker for this pathway. The research populace has, however, been globally divided by skepticism and hope regarding this approval. Failure to meet clinical endpoints in the trials, alleged transparency issues, cost-effectiveness, potential adverse effects, need for regular monitoring, and critique of 'amyloid cascade hypothesis' itself are the main caveats concerning the antibody. With this controversy in background, this paper critically looks at antibody research in AD therapeutics, evidence, and evolution of Aducanumab as a drug and the potential clinical implications of its use in future. While the efficacy of this monoclonal antibody in AD stands as a test of time, based on the growing evidence it is vital to rethink and explore alternate pathways of pathogenesis (oxidative stress, neuroinflammation, cholesterol metabolism, vascular factors, etc.) as possible therapeutic targets that may help elucidate the enigma of this complex yet progressive and debilitating neurodegenerative disorder.
Collapse
Affiliation(s)
- Sanchari Mukhopadhyay
- Geriatric Unit, Department of Psychiatry, NationalInstitute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Debanjan Banerjee
- Geriatric Unit, Department of Psychiatry, NationalInstitute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
212
|
Liu C, Cheng ZY, Xia QP, Hu YH, Wang C, He L. GPR40 receptor agonist TAK-875 improves cognitive deficits and reduces β-amyloid production in APPswe/PS1dE9 mice. Psychopharmacology (Berl) 2021; 238:2133-2146. [PMID: 34173034 DOI: 10.1007/s00213-021-05837-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/22/2021] [Indexed: 02/03/2023]
Abstract
RATIONALE Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive cognitive dysfunction and memory impairment. G protein-coupled receptor 40 (GPR40) is expressed in brain in addition to periphery and is associated with cognitive function such as space orientation, memory, and learning. However, the effects and mechanisms of GPR40 agonist in improving the AD progression remain largely unknown. OBJECTIVES The present study aimed to investigate the therapeutic effects and mechanisms of a potent and selective GPR40 agonist TAK-875 on the APPswe/PS1dE9 mice. RESULTS The results showed that intracerebroventricular administration of TAK-875 significantly rescued cognitive deficits in APPswe/PS1dE9 mice, and these effects may be mediated by the regulation of phospholipase C/protein kinase C signaling pathway, which enhanced α-secretase ADAM10 activity, promoted amyloid precursor protein non-amyloidogenic processing pathway, and reduced β-amyloid production. CONCLUSIONS These results suggest that GPR40 may be a potential therapeutic target for AD, and GPR40 agonists may become promising AD drugs in the future.
Collapse
Affiliation(s)
- Chao Liu
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, Jiang Su Province, China
| | - Zhao-Yan Cheng
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, Jiang Su Province, China
| | - Qing-Peng Xia
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, Jiang Su Province, China
| | - Yu-Hui Hu
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, Jiang Su Province, China
| | - Chen Wang
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, Jiang Su Province, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, Jiang Su Province, China.
| |
Collapse
|
213
|
Smit T, Deshayes NAC, Borchelt DR, Kamphuis W, Middeldorp J, Hol EM. Reactive astrocytes as treatment targets in Alzheimer's disease-Systematic review of studies using the APPswePS1dE9 mouse model. Glia 2021; 69:1852-1881. [PMID: 33634529 PMCID: PMC8247905 DOI: 10.1002/glia.23981] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022]
Abstract
Astrocytes regulate synaptic communication and are essential for proper brain functioning. In Alzheimer's disease (AD) astrocytes become reactive, which is characterized by an increased expression of intermediate filament proteins and cellular hypertrophy. Reactive astrocytes are found in close association with amyloid-beta (Aβ) deposits. Synaptic communication and neuronal network function could be directly modulated by reactive astrocytes, potentially contributing to cognitive decline in AD. In this review, we focus on reactive astrocytes as treatment targets in AD in the APPswePS1dE9 AD mouse model, a widely used model to study amyloidosis and gliosis. We first give an overview of the model; that is, how it was generated, which cells express the transgenes, and the effect of its genetic background on Aβ pathology. Subsequently, to determine whether modifying reactive astrocytes in AD could influence pathogenesis and cognition, we review studies using this mouse model in which interventions were directly targeted at reactive astrocytes or had an indirect effect on reactive astrocytes. Overall, studies specifically targeting astrocytes to reduce astrogliosis showed beneficial effects on cognition, which indicates that targeting astrocytes should be included in developing novel therapies for AD.
Collapse
Affiliation(s)
- Tamar Smit
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
- Swammerdam Institute for Life SciencesCenter for Neuroscience, University of AmsterdamAmsterdamThe Netherlands
| | - Natasja A. C. Deshayes
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
- Swammerdam Institute for Life SciencesCenter for Neuroscience, University of AmsterdamAmsterdamThe Netherlands
| | - David R. Borchelt
- Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, Department of NeuroscienceUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Willem Kamphuis
- Netherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Jinte Middeldorp
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
- Department of ImmunobiologyBiomedical Primate Research CentreRijswijkThe Netherlands
| | - Elly M. Hol
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
214
|
King-Robson J, Wilson H, Politis M. Associations Between Amyloid and Tau Pathology, and Connectome Alterations, in Alzheimer's Disease and Mild Cognitive Impairment. J Alzheimers Dis 2021; 82:541-560. [PMID: 34057079 DOI: 10.3233/jad-201457] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The roles of amyloid-β and tau in the degenerative process of Alzheimer's disease (AD) remain uncertain. [18F]AV-45 and [18F]AV-1451 PET quantify amyloid-β and tau pathology, respectively, while diffusion tractography enables detection of their microstructural consequences. OBJECTIVE Examine the impact of amyloid-β and tau pathology on the structural connectome and cognition, in mild cognitive impairment (MCI) and AD. METHODS Combined [18F]AV-45 and [18F]AV-1451 PET, diffusion tractography, and cognitive assessment in 28 controls, 32 MCI, and 26 AD patients. RESULTS Hippocampal connectivity was reduced to the thalami, right lateral orbitofrontal, and right amygdala in MCI; alongside the insula, posterior cingulate, right entorhinal, and numerous cortical regions in AD (all p < 0.05). Hippocampal strength inversely correlated with [18F]AV-1451 SUVr in MCI (r = -0.55, p = 0.049) and AD (r = -0.57, p = 0.046), while reductions in hippocampal connectivity to ipsilateral brain regions correlated with increased [18F]AV-45 SUVr in those same regions in MCI (r = -0.33, p = 0.003) and AD (r = -0.31, p = 0.006). Cognitive scores correlated with connectivity of the right temporal pole in MCI (r = -0.60, p = 0.035) and left hippocampus in AD (r = 0.69, p = 0.024). Clinical Dementia Rating Scale scores correlated with [18F]AV-1451 SUVr in multiple areas reflecting Braak stages I-IV, including the right (r = 0.65, p = 0.004) entorhinal cortex in MCI; and Braak stages III-VI, including the right (r = 0.062, p = 0.009) parahippocampal gyrus in AD. CONCLUSION Reductions in hippocampal connectivity predominate in the AD connectome, correlating with hippocampal tau in MCI and AD, and with amyloid-β in the target regions of those connections. Cognitive scores correlate with microstructural changes and reflect the accumulation of tau pathology.
Collapse
Affiliation(s)
- Josh King-Robson
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Heather Wilson
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK
| | | |
Collapse
|
215
|
Li XY, Cui Y, Jing D, Xie K, Zhong X, Kong Y, Wang Y, Chu M, Wang C, Wu L. Novel PSEN1 and PSEN2 Mutations Identified in Sporadic Early-onset Alzheimer Disease and Posterior Cortical Atrophy. Alzheimer Dis Assoc Disord 2021; 35:208-213. [PMID: 33973882 PMCID: PMC8386585 DOI: 10.1097/wad.0000000000000438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 12/31/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND/PURPOSE Sporadic early-onset Alzheimer disease (sEOAD) and its visual variant, posterior cortical atrophy (PCA), have a disease onset at less than 65 years of age with no familial aggregation. The etiology and genetic basis of these diseases remain poorly understood. Our study aimed to identify additional mutations or variants associated with sEOAD and PCA and to further examine their genetic and phenotypic spectrums. METHODS We performed whole-exome sequencing and analyzed the clinical and neuroimaging features of mutation carriers with 29 patients having sEOAD and 25 having PCA. RESULTS Nine rare damaging variants were identified in 4 patients with sEOAD and 3 with PCA. A novel mutation (p.A136V) in PSEN1 was identified in a patient with sEOAD and a likely pathogenic variant (p.M239T) was identified for PSEN2 in a patient with PCA. In addition, 7 rare damaging variants were detected in other genes related to neurodegenerative diseases. The patient carrying the PSEN1 p.A136V mutation presented with typical clinical and imaging features of sEOAD, and the PCA patient with the PSEN2 p.M239T mutation presented with visuospatial impairment as the initial symptom. CONCLUSION Our study expands the PSEN1 mutation spectrum of sEOAD and highlights the importance of screening PSEN1 and/or PSEN2 mutations in PCA patients.
Collapse
|
216
|
Zhang T, Ma S, Lv J, Wang X, Afewerky HK, Li H, Lu Y. The emerging role of exosomes in Alzheimer's disease. Ageing Res Rev 2021; 68:101321. [PMID: 33727157 DOI: 10.1016/j.arr.2021.101321] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/20/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), manifested by memory loss and a decline in cognitive functions, is the most prevalent neurodegenerative disease accounting for 60-80 % of dementia cases. But, to-date, there is no effective treatment available to slow or stop the progression of AD. Exosomes are small extracellular vesicles that carry constituents, such as functional messenger RNAs, non-coding RNAs, proteins, lipids, DNA, and other bioactive substances of their source cells. In the brain, exosomes are likely to be sourced by almost all cell types and involve in cell communication to regulate cellular functions. The yet, accumulated evidence on the roles of exosomes and their constituents in the AD pathological process suggests their significance as additional biomarkers and therapeutic targets for AD. This review summarizes the current reported research findings on exosomes roles in the pathogenesis, diagnosis, and treatment of AD.
Collapse
|
217
|
Antonarakis SE. History of the methodology of disease gene identification. Am J Med Genet A 2021; 185:3266-3275. [PMID: 34159713 PMCID: PMC8596769 DOI: 10.1002/ajmg.a.62400] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 11/06/2022]
Abstract
The past 45 years have witnessed a triumph in the discovery of genes and genetic variation that cause Mendelian disorders due to high impact variants. Important discoveries and organized projects have provided the necessary tools and infrastructure for the identification of gene defects leading to thousands of monogenic phenotypes. This endeavor can be divided in three phases in which different laboratory strategies were employed for the discovery of disease-related genes: (i) the biochemical phase, (ii) the genetic linkage followed by positional cloning phase, and (iii) the sequence identification phase. However, much more work is needed to identify all the high impact genomic variation that substantially contributes to the phenotypic variation.
Collapse
Affiliation(s)
- Stylianos E Antonarakis
- University of Geneva Medical School, Geneva, Switzerland.,Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| |
Collapse
|
218
|
Zhu X, Zhang Y, Yang X, Hao C, Duan H. Gene Therapy for Neurodegenerative Disease: Clinical Potential and Directions. Front Mol Neurosci 2021; 14:618171. [PMID: 34194298 PMCID: PMC8236824 DOI: 10.3389/fnmol.2021.618171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/07/2021] [Indexed: 12/21/2022] Open
Abstract
The pathogenesis of neurodegenerative diseases (NDDs) is complex and diverse. Over the decades, our understanding of NDD has been limited to pathological features. However, recent advances in gene sequencing have facilitated elucidation of NDD at a deeper level. Gene editing techniques have uncovered new genetic links to phenotypes, promoted the development of novel treatment strategies and equipped researchers with further means to construct effective cell and animal models. The current review describes the history of evolution of gene editing tools, with the aim of improving overall understanding of this technology, and focuses on the four most common NDD disorders to demonstrate the potential future applications and research directions of gene editing.
Collapse
Affiliation(s)
- Xiaolin Zhu
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yu Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xin Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunyan Hao
- Department of Geriatrics, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hubin Duan
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, China.,Department of Neurosurgery, Lvliang People's Hospital, Lvliang, China
| |
Collapse
|
219
|
Jurcau A, Simion A. Oxidative Stress in the Pathogenesis of Alzheimer's Disease and Cerebrovascular Disease with Therapeutic Implications. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:94-108. [PMID: 32124703 DOI: 10.2174/1871527319666200303121016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 01/18/2020] [Accepted: 01/28/2020] [Indexed: 12/15/2022]
Abstract
The significant gain in life expectancy led to an increase in the incidence and prevalence of dementia. Although vascular risk factors have long and repeatedly been shown to increase the risk of Alzheimer's Disease (AD), translating these findings into effective preventive measures has failed. In addition, the finding that incident ischemic stroke approximately doubles the risk of a patient to develop AD has been recently reinforced. Current knowledge and pathogenetic hypotheses of AD are discussed. The implication of oxidative stress in the development of AD is reviewed, with special emphasis on its sudden burst in the setting of acute ischemic stroke and the possible link between this increase in oxidative stress and consequent cognitive impairment. Current knowledge and future directions in the prevention and treatment of AD are discussed outlining the hypothesis of a possible beneficial effect of antioxidant treatment in acute ischemic stroke in delaying the onset/progression of dementia.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Faculty of Medicine and Pharmacy, University of Oradea, 410154 Oradea, Romania.,Clinical Municipal Hospital "Dr. G Curteanu", Neurology Ward, Oradea, Romania
| | - Aurel Simion
- Faculty of Medicine and Pharmacy, University of Oradea, 410154 Oradea, Romania.,Clinical Municipal Hospital "Dr. G Curteanu", Neurological Rehabilitation Ward, Oradea, Romania
| |
Collapse
|
220
|
Yu W, Yu W, Yang Y, Lü Y. Exploring the Key Genes and Identification of Potential Diagnosis Biomarkers in Alzheimer's Disease Using Bioinformatics Analysis. Front Aging Neurosci 2021; 13:602781. [PMID: 34194312 PMCID: PMC8236887 DOI: 10.3389/fnagi.2021.602781] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 05/06/2021] [Indexed: 12/30/2022] Open
Abstract
Background Alzheimer’s disease (AD) is one of the major threats of the twenty-first century and lacks available therapy. Identification of novel molecular markers for diagnosis and treatment of AD is urgently demanded, and genetic biomarkers show potential prospects. Method We identify and intersected differentially expressed genes (DEGs) from five microarray datasets to detect consensus DEGs. Based on these DEGs, we conducted Gene Ontology (GO), performed the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, constructed a protein—protein interaction (PPI) network, and utilized Cytoscape to identify hub genes. The least absolute shrinkage and selection operator (LASSO) logistic regression was applied to identify potential diagnostic biomarkers. Gene set enrichment analysis (GSEA) was performed to investigate the biological functions of the key genes. Result We identified 608 consensus DEGs, several dysregulated pathways, and 18 hub genes. Sixteen hub genes dysregulated as AD progressed. The diagnostic model of 35 genes was constructed, which has a high area under the curve (AUC) value in both the validation dataset and combined dataset (AUC = 0.992 and AUC = 0.985, respectively). The model can also differentiate mild cognitive impairment and AD patients from controls in two blood datasets. Brain-derived neurotrophic factor (BDNF) and WW domain-containing transcription regulator protein 1 (WWTR1), which are associated with the Braak stage, Aβ 42 levels, and β-secretase activity, were identified as critical genes of AD. Conclusion Our study identified 16 hub genes correlated to the neuropathological stage and 35 potential biomarkers for the diagnosis of AD. WWTR1 were identified as candidate genes for future studies. This study deepens our understanding of the transcriptomic and functional features and provides new potential diagnostic biomarkers and therapeutic targets for AD.
Collapse
Affiliation(s)
- Wuhan Yu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weihua Yu
- Institutes of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Yan Yang
- State Key Laboratory of Power Transmission Equipment and System Security and New Technology, College of Electrical Engineering, Chongqing University, Chongqing, China
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
221
|
Tortelli R, Seripa D, Zecca C, Dell’Abate MT, Bisceglia P, Barulli MR, De Blasi R, Logroscino G. A New Presenilin 1 (Psen1) Mutation (p.Cys263Trp) as a Cause of Both Early and Late-Onset Alzheimer's Disease in a Large Italian Family. Int J Mol Sci 2021; 22:ijms22126215. [PMID: 34207526 PMCID: PMC8230263 DOI: 10.3390/ijms22126215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
Mutations in the PSEN1 gene are the most common cause of autosomal dominant Alzheimer's disease, and are characterized by a high phenotype variability. This study describes a five-generation family, with a prevalent late-onset of the disease and a high frequency of depression, in which a new missense mutation (c.789T > G, p.Cys263Trp) in exon 8 of the PSEN1 gene was found. Only the proband presented an early onset at the age of 45 with attention deficit, followed by spatial disorientation, psychiatric symptoms and parkinsonian signs. The other two cases had a late onset of the disease and a typical presentation with memory loss. Both were characterized by a high level of anxiety and depression. The disease course was different with signs of Lewy body dementia for the proband's mother, and pyramidal involvement and a shorter disease duration for the proband's maternal aunt. The other eight cases with late-onset dementia and three cases with a long history of depression have been reported in the family pedigree, underlying the high phenotype variability of PSEN1 mutations.
Collapse
Affiliation(s)
- Rosanna Tortelli
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
- Correspondence: (R.T.); (G.L.); Tel.: +39-0833773904 (R.T. & G.L.); Fax: +39-08831830670 (R.T. & G.L.)
| | - Davide Seripa
- Complex Unit of Geriatrics, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (D.S.); (P.B.)
- Laboratory for Advanced Hematological Diagnostics, Department of Hematology and Stem Cell Transplant, “Vito Fazzi” Hospital, 73100 Lecce, Italy
| | - Chiara Zecca
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
| | - Maria Teresa Dell’Abate
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
| | - Paola Bisceglia
- Complex Unit of Geriatrics, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (D.S.); (P.B.)
| | - Maria Rosaria Barulli
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
| | - Roberto De Blasi
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari “Aldo Moro”—A.O. Pia Fondazione Cardinale G. Panico, 73039 Tricase, Italy; (C.Z.); (M.T.D.); (M.R.B.); (R.D.B.)
- Department of Radiology, “Pia Fondazione Cardinale G. Panico”, 73039 Tricase, Italy
| | - Giancarlo Logroscino
- Department of Radiology, “Pia Fondazione Cardinale G. Panico”, 73039 Tricase, Italy
- Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, 70124 Bari, Italy
- Correspondence: (R.T.); (G.L.); Tel.: +39-0833773904 (R.T. & G.L.); Fax: +39-08831830670 (R.T. & G.L.)
| |
Collapse
|
222
|
Müller L, Kirschstein T, Köhling R, Kuhla A, Teipel S. Neuronal Hyperexcitability in APPSWE/PS1dE9 Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2021; 81:855-869. [PMID: 33843674 DOI: 10.3233/jad-201540] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transgenic mouse models serve a better understanding of Alzheimer's disease (AD) pathogenesis and its consequences on neuronal function. Well-known and broadly used AD models are APPswe/PS1dE9 mice, which are able to reproduce features of amyloid-β (Aβ) plaque formations as well as neuronal dysfunction as reflected in electrophysiological recordings of neuronal hyperexcitability. The most prominent findings include abnormal synaptic function and synaptic reorganization as well as changes in membrane threshold and spontaneous neuronal firing activities leading to generalized excitation-inhibition imbalances in larger neuronal circuits and networks. Importantly, these findings in APPswe/PS1dE9 mice are at least partly consistent with results of electrophysiological studies in humans with sporadic AD. This underscores the potential to transfer mechanistic insights into amyloid related neuronal dysfunction from animal models to humans. This is of high relevance for targeted downstream interventions into neuronal hyperexcitability, for example based on repurposing of existing antiepileptic drugs, as well as the use of combinations of imaging and electrophysiological readouts to monitor effects of upstream interventions into amyloid build-up and processing on neuronal function in animal models and human studies. This article gives an overview on the pathogenic and methodological basis for recording of neuronal hyperexcitability in AD mouse models and on key findings in APPswe/PS1dE9 mice. We point at several instances to the translational perspective into clinical intervention and observation studies in humans. We particularly focus on bi-directional relations between hyperexcitability and cerebral amyloidosis, including build-up as well as clearance of amyloid, possibly related to sleep and so called glymphatic system function.
Collapse
Affiliation(s)
- Luisa Müller
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock and Greifswald, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| |
Collapse
|
223
|
Poddar MK, Banerjee S, Chakraborty A, Dutta D. Metabolic disorder in Alzheimer's disease. Metab Brain Dis 2021; 36:781-813. [PMID: 33638805 DOI: 10.1007/s11011-021-00673-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD), a well known aging-induced neurodegenerative disease is related to amyloid proteinopathy. This proteinopathy occurs due to abnormalities in protein folding, structure and thereby its function in cells. The root cause of such kind of proteinopathy and its related neurodegeneration is a disorder in metabolism, rather metabolomics of the major as well as minor nutrients. Metabolomics is the most relevant "omics" platform that offers a great potential for the diagnosis and prognosis of neurodegenerative diseases as an individual's metabolome. In recent years, the research on such kinds of neurodegenerative diseases, especially aging-related disorders is broadened its scope towards metabolic function. Different neurotransmitter metabolisms are also involved with AD and its associated neurodegeneration. The genetic and epigenetic backgrounds are also noteworthy. In this review, the physiological changes of AD in relation to its corresponding biochemical, genetic and epigenetic involvements including its (AD) therapeutic aspects are discussed.
Collapse
Affiliation(s)
- Mrinal K Poddar
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India.
| | - Soumyabrata Banerjee
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Departrment of Psychology, Neuroscience Program, Field Neurosciences Institute Research Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Apala Chakraborty
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
| | - Debasmita Dutta
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, 58102, USA
| |
Collapse
|
224
|
|
225
|
Limited Substrate Specificity of PS/γ-Secretase Is Supported by Novel Multiplexed FRET Analysis in Live Cells. BIOSENSORS-BASEL 2021; 11:bios11060169. [PMID: 34073182 PMCID: PMC8228125 DOI: 10.3390/bios11060169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/01/2022]
Abstract
Presenilin (PS)/γ-secretase is an aspartyl protease that processes a wide range of transmembrane proteins such as the amyloid precursor protein (APP) and Notch1, playing essential roles in normal biological events and diseases. However, whether there is a substrate preference for PS/γ-secretase processing in cells is not fully understood. Structural studies of PS/γ-secretase enfolding a fragment of APP or Notch1 showed that the two substrates engage the protease in broadly similar ways, suggesting the limited substrate specificity of PS/γ-secretase. In the present study, we developed a new multiplexed imaging platform that, for the first time, allowed us to quantitatively monitor how PS/γ-secretase processes two different substrates (e.g., APP vs. Notch1) in the same cell. In this assay, we utilized the recently reported, spectrally compatible visible and near-infrared (NIR)-range Förster resonance energy transfer (FRET) biosensors that permit quantitative recording of PS/γ-secretase activity in live cells. Here, we show that, overall, PS/γ-secretase similarly cleaves Notch1 N100, wild-type APP C99, and familial Alzheimer’s disease (FAD)-linked APP C99 mutants in Chinese hamster ovary (CHO) cells, which further supports the limited PS/γ-secretase substrate specificity. On the other hand, a cell-by-cell basis analysis demonstrates a certain degree of variability in substrate recognition and processing by PS/γ-secretase among different cells. Our new multiplexed FRET assay could be a useful tool to better understand how PS/γ-secretase processes its multiple substrates in normal and disease conditions in live, intact cells.
Collapse
|
226
|
Podvin S, Jones A, Liu Q, Aulston B, Mosier C, Ames J, Winston C, Lietz CB, Jiang Z, O’Donoghue AJ, Ikezu T, Rissman RA, Yuan SH, Hook V. Mutant Presenilin 1 Dysregulates Exosomal Proteome Cargo Produced by Human-Induced Pluripotent Stem Cell Neurons. ACS OMEGA 2021; 6:13033-13056. [PMID: 34056454 PMCID: PMC8158845 DOI: 10.1021/acsomega.1c00660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/16/2021] [Indexed: 05/28/2023]
Abstract
The accumulation and propagation of hyperphosphorylated tau (p-Tau) is a neuropathological hallmark occurring with neurodegeneration of Alzheimer's disease (AD). Extracellular vesicles, exosomes, have been shown to initiate tau propagation in the brain. Notably, exosomes from human-induced pluripotent stem cell (iPSC) neurons expressing the AD familial A246E mutant form of presenilin 1 (mPS1) are capable of inducing tau deposits in the mouse brain after in vivo injection. To gain insights into the exosome proteome cargo that participates in propagating tau pathology, this study conducted proteomic analysis of exosomes produced by human iPSC neurons expressing A246E mPS1. Significantly, mPS1 altered the profile of exosome cargo proteins to result in (1) proteins present only in mPS1 exosomes and not in controls, (2) the absence of proteins in the mPS1 exosomes which were present only in controls, and (3) shared proteins which were upregulated or downregulated in the mPS1 exosomes compared to controls. These results show that mPS1 dysregulates the proteome cargo of exosomes to result in the acquisition of proteins involved in the extracellular matrix and protease functions, deletion of proteins involved in RNA and protein translation systems along with proteasome and related functions, combined with the upregulation and downregulation of shared proteins, including the upregulation of amyloid precursor protein. Notably, mPS1 neuron-derived exosomes displayed altered profiles of protein phosphatases and kinases involved in regulating the status of p-tau. The dysregulation of exosome cargo proteins by mPS1 may be associated with the ability of mPS1 neuron-derived exosomes to propagate tau pathology.
Collapse
Affiliation(s)
- Sonia Podvin
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Alexander Jones
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Qing Liu
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Brent Aulston
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Charles Mosier
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Janneca Ames
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Charisse Winston
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Christopher B. Lietz
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Zhenze Jiang
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Anthony J. O’Donoghue
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Tsuneya Ikezu
- Department
of Pharmacology and Experimental Therapeutics, Department of Neurology,
Alzheimer’s Disease Research Center, Boston University, School of Medicine, Boston 02118, Massachusetts, United States
| | - Robert A. Rissman
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
- Veterans
Affairs San Diego Healthcare System,
La Jolla, San Diego 92161, California, United States
| | - Shauna H. Yuan
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Vivian Hook
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, San Diego 92093, California, United States
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| |
Collapse
|
227
|
Modesti L, Danese A, Angela Maria Vitto V, Ramaccini D, Aguiari G, Gafà R, Lanza G, Giorgi C, Pinton P. Mitochondrial Ca 2+ Signaling in Health, Disease and Therapy. Cells 2021; 10:cells10061317. [PMID: 34070562 PMCID: PMC8230075 DOI: 10.3390/cells10061317] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
The divalent cation calcium (Ca2+) is considered one of the main second messengers inside cells and acts as the most prominent signal in a plethora of biological processes. Its homeostasis is guaranteed by an intricate and complex system of channels, pumps, and exchangers. In this context, by regulating cellular Ca2+ levels, mitochondria control both the uptake and release of Ca2+. Therefore, at the mitochondrial level, Ca2+ plays a dual role, participating in both vital physiological processes (ATP production and regulation of mitochondrial metabolism) and pathophysiological processes (cell death, cancer progression and metastasis). Hence, it is not surprising that alterations in mitochondrial Ca2+ (mCa2+) pathways or mutations in Ca2+ transporters affect the activities and functions of the entire cell. Indeed, it is widely recognized that dysregulation of mCa2+ signaling leads to various pathological scenarios, including cancer, neurological defects and cardiovascular diseases (CVDs). This review summarizes the current knowledge on the regulation of mCa2+ homeostasis, the related mechanisms and the significance of this regulation in physiology and human diseases. We also highlight strategies aimed at remedying mCa2+ dysregulation as promising therapeutical approaches.
Collapse
Affiliation(s)
- Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Alberto Danese
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Daniela Ramaccini
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Roberta Gafà
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Giovanni Lanza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
- Correspondence: ; Tel.: +39-0532-455802
| |
Collapse
|
228
|
Prendecki M, Kowalska M, Toton E, Kozubski W. Genetic Editing and Pharmacogenetics in Current And Future Therapy Of Neurocognitive Disorders. Curr Alzheimer Res 2021; 17:238-258. [PMID: 32321403 DOI: 10.2174/1567205017666200422152440] [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: 04/05/2019] [Revised: 02/05/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022]
Abstract
Dementia is an important issue in western societies, and in the following years, this problem will also rise in the developing regions, such as Africa and Asia. The most common types of dementia in adults are Alzheimer's Disease (AD), Dementia with Lewy Bodies (DLB), Frontotemporal Dementia (FTD) and Vascular Dementia (VaD), of which, AD accounts for more than half of the cases. The most prominent symptom of AD is cognitive impairment, currently treated with four drugs: Donepezil, rivastigmine, and galantamine, enhancing cholinergic transmission; as well as memantine, protecting neurons against glutamate excitotoxicity. Despite ongoing efforts, no new drugs in the treatment of AD have been registered for the last ten years, thus multiple studies have been conducted on genetic factors affecting the efficacy of antidementia pharmacotherapy. The researchers investigate the effects of variants in multiple genes, such as ABCB1, ACE, CHAT, CHRNA7, CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP3A5, CYP3A7, NR1I2, NR1I3, POR, PPAR, RXR, SLC22A1/2/5, SLC47A1, UGT1A6, UGT1A9 and UGT2B7, associated with numerous pathways: the development of pathological proteins, formation and metabolism of acetylcholine, transport, metabolism and excretion of antidementia drugs and transcription factors regulating the expression of genes responsible for metabolism and transport of drugs. The most promising results have been demonstrated for APOE E4, dementia risk variant, BCHE-K, reduced butyrylcholinesterase activity variant, and CYP2D6 UM, ultrarapid hepatic metabolism. Further studies investigate the possibilities of the development of emerging drugs or genetic editing by CRISPR/Cas9 for causative treatment. In conclusion, the pharmacogenetic studies on dementia diseases may improve the efficacy of pharmacotherapy in some patients with beneficial genetic variants, at the same time, identifying the carriers of unfavorable alleles, the potential group of novel approaches to the treatment and prevention of dementia.
Collapse
Affiliation(s)
- Michal Prendecki
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marta Kowalska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Ewa Toton
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
229
|
Zhang Z, Yue P, Lu T, Wang Y, Wei Y, Wei X. Role of lysosomes in physiological activities, diseases, and therapy. J Hematol Oncol 2021; 14:79. [PMID: 33990205 PMCID: PMC8120021 DOI: 10.1186/s13045-021-01087-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Long known as digestive organelles, lysosomes have now emerged as multifaceted centers responsible for degradation, nutrient sensing, and immunity. Growing evidence also implicates role of lysosome-related mechanisms in pathologic process. In this review, we discuss physiological function of lysosomes and, more importantly, how the homeostasis of lysosomes is disrupted in several diseases, including atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, lysosomal storage disorders, and malignant tumors. In atherosclerosis and Gaucher disease, dysfunction of lysosomes changes cytokine secretion from macrophages, partially through inflammasome activation. In neurodegenerative diseases, defect autophagy facilitates accumulation of toxic protein and dysfunctional organelles leading to neuron death. Lysosomal dysfunction has been demonstrated in pathology of pancreatitis. Abnormal autophagy activation or inhibition has been revealed in autoimmune disorders. In tumor microenvironment, malignant phenotypes, including tumorigenesis, growth regulation, invasion, drug resistance, and radiotherapy resistance, of tumor cells and behaviors of tumor-associated macrophages, fibroblasts, dendritic cells, and T cells are also mediated by lysosomes. Based on these findings, a series of therapeutic methods targeting lysosomal proteins and processes have been developed from bench to bedside. In a word, present researches corroborate lysosomes to be pivotal organelles for understanding pathology of atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, and lysosomal storage disorders, and malignant tumors and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Ziqi Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Pengfei Yue
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Tianqi Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| |
Collapse
|
230
|
Abstract
Somatic mutations arise postzygotically, producing genetic differences between cells in an organism. Well established as a driver of cancer, somatic mutations also exist in nonneoplastic cells, including in the brain. Technological advances in nucleic acid sequencing have enabled recent break-throughs that illuminate the roles of somatic mutations in aging and degenerative diseases of the brain. Somatic mutations accumulate during aging in human neurons, a process termed genosenium. A number of recent studies have examined somatic mutations in Alzheimer’s disease (AD), primarily from the perspective of genes causing familial AD. We have also gained new information on genome-wide mutations, providing insights into the cellular events driving somatic mutation and cellular dysfunction. This review highlights recent concepts, methods, and findings in the progress to understand the role of brain somatic mutation in aging and AD.
Collapse
Affiliation(s)
- Michael B Miller
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Neuropathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA; .,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Hannah C Reed
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Allegheny College, Meadville, Pennsylvania 16335, USA;
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
231
|
Seita Y, Morimura T, Watanabe N, Iwatani C, Tsuchiya H, Nakamura S, Suzuki T, Yanagisawa D, Tsukiyama T, Nakaya M, Okamura E, Muto M, Ema M, Nishimura M, Tooyama I. Generation of Transgenic Cynomolgus Monkeys Overexpressing the Gene for Amyloid-β Precursor Protein. J Alzheimers Dis 2021; 75:45-60. [PMID: 32250299 PMCID: PMC7306892 DOI: 10.3233/jad-191081] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia and understanding its pathogenesis should lead to improved therapeutic and diagnostic methods. Although several groups have developed transgenic mouse models overexpressing the human amyloid-β precursor protein (APP) gene with AD mutations, with and without presenilin mutations, as well as APP gene knock-in mouse models, these animals display amyloid pathology but do not show neurofibrillary tangles or neuronal loss. This presumably is due to differences between the etiology of the aged-related human disease and the mouse models. Here we report the generation of two transgenic cynomolgus monkeys overexpressing the human gene for APP with Swedish, Artic, and Iberian mutations, and demonstrated expression of gene tagged green fluorescent protein marker in the placenta, amnion, hair follicles, and peripheral blood. We believe that these nonhuman primate models will be very useful to study the pathogenesis of dementia and AD. However, generated Tg monkeys still have some limitations. We employed the CAG promoter, which will promote gene expression in a non-tissue specific manner. Moreover, we used transgenic models but not knock-in models. Thus, the inserted transgene destroys endogenous gene(s) and may affect the phenotype(s). Nevertheless, it will be of great interest to determine whether these Tg monkeys will develop tauopathy and neurodegeneration similar to human AD.
Collapse
Affiliation(s)
- Yasunari Seita
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Toshifumi Morimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Naoki Watanabe
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Chizuru Iwatani
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Hideaki Tsuchiya
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Shinichiro Nakamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita12-Nishi6, Kita-ku, Sapporo, Japan
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Tomoyuki Tsukiyama
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Masataka Nakaya
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Eiichi Okamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Masanaga Muto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Masaki Nishimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, Japan
| |
Collapse
|
232
|
Importance of GWAS in finding un-targeted genetic association of sporadic Alzheimer’s disease. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00130-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
233
|
Zhang J, Zhao ZJ, Fu X, Niu H, Hu C, Dong Y, Cui MZ, Zhang F, Zeng L, Xu X. Proapoptotic Mitochondrial Carrier Homolog Protein PSAP Mediates Death Receptor 6 Induced Apoptosis. J Alzheimers Dis 2021; 74:1097-1106. [PMID: 32144986 DOI: 10.3233/jad-191086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Presenilin-associated protein (PSAP) was originally identified as a mitochondrial proapoptotic protein. To further explore the apoptotic pathway that involves PSAP, our yeast two-hybrid screen revealed that PSAP interacts with a death receptor, DR6. DR6 is a relatively less common member of the death receptor family and has been shown to mediate the neurotoxicity of amyloid-β, mutant SOD1, and prion proteins and has also been implicated in the regulation of immune cell proliferation and differentiation. Our previous study showed that DR6 induces apoptosis via a unique mitochondria-dependent pathway different from the conventional death receptor-mediated extrinsic apoptotic pathways. Thus, the interaction of DR6 with PSAP established a direct molecular link between DR6 and mitochondrial apoptotic pathway. We investigated the possible role of PSAP in DR6-induced apoptosis. Interestingly, it was discovered that knockdown of PSAP strongly inhibited DR6-induced apoptosis. To further elucidate the mechanism by which PSAP mediates DR6-induced mitochondria-dependent apoptosis, our data demonstrated that knockdown of PSAP blocked DR6-induced Bax translocation and cytochrome c release from the mitochondria. Moreover, it was found that both PSAP and DR6 form complexes with Bax, but at different subcellular locations. The DR6-Bax complex was detected in the cytosolic fraction while the PSAP-Bax complex was detected in the mitochondrial fraction. The observation that knockdown of DR6 significantly reduced the amount of PSAP-Bax complex detected in mitochondria suggests a possibility that DR6-bound Bax is transferred to PSAP upon interaction with PSAP at the mitochondria, leading to cytochrome c release and eventually apoptosis.
Collapse
Affiliation(s)
- Jingtian Zhang
- Edmond H. Fischer Signal Transduction Laboratory, College of Life Sciences, Jilin University, Changchun, China
| | - Zhizhuang Joe Zhao
- Edmond H. Fischer Signal Transduction Laboratory, College of Life Sciences, Jilin University, Changchun, China.,Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xueqi Fu
- Edmond H. Fischer Signal Transduction Laboratory, College of Life Sciences, Jilin University, Changchun, China
| | - Han Niu
- Edmond H. Fischer Signal Transduction Laboratory, College of Life Sciences, Jilin University, Changchun, China
| | - Chen Hu
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yunzhou Dong
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mei-Zhen Cui
- Department of Biology, College of Arts and Sciences, University of Texas of the Permian Basin, Odessa, TX, USA
| | - Fuqiang Zhang
- Scientific Research Centre of China-Japan Union Hospital, Jilin University, Changchun, China
| | - Linlin Zeng
- Edmond H. Fischer Signal Transduction Laboratory, College of Life Sciences, Jilin University, Changchun, China
| | - Xuemin Xu
- Department of Biology, College of Arts and Sciences, University of Texas of the Permian Basin, Odessa, TX, USA
| |
Collapse
|
234
|
Mold M, Linhart C, Gómez-Ramírez J, Villegas-Lanau A, Exley C. Aluminum and Amyloid-β in Familial Alzheimer's Disease. J Alzheimers Dis 2021; 73:1627-1635. [PMID: 31958088 DOI: 10.3233/jad-191140] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Genetic predispositions associated with metabolism of the amyloid-β protein precursor underlie familial Alzheimer's disease; a form of dementia characterized by early disease onset and elevated levels of cortical amyloid-β. Human exposure to aluminum is linked to the etiology of Alzheimer's disease and recent research measured a high content of aluminum in brain tissue in familial Alzheimer's disease. To elaborate upon this finding, we have obtained brain tissues from a Colombian cohort of donors with familial Alzheimer's disease. We have used established methods to measure the aluminum content of these tissues and we have compared the data with a recently measured dataset for control brain tissues. We report significantly higher levels of aluminum in brain tissues in donors with familial Alzheimer's disease than in control tissues from donors without neurological impairment or neurodegeneration. We have used aluminum-specific fluorescence microscopy along with complementary imaging for amyloid-β to demonstrate a very high degree of co-localization of these two risk factors in brain tissue in familial Alzheimer's disease. Aluminum and amyloid-β were co-located in senile plaques as well as vasculature, the latter resembling cerebral amyloid angiopathy. Aluminum was also found separately from amyloid-β in intracellular compartments including glia and neuronal axons. The research has identified an arguably unique association between high brain aluminum content and amyloid-β and allows postulation that genetic predispositions defining familial Alzheimer's disease underlie this relationship.
Collapse
Affiliation(s)
- Matthew Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| | - Caroline Linhart
- Institute of Pharmacy/Pharmacognosy, University of Innsbruck, Innsbruck, Austria
| | - Johana Gómez-Ramírez
- Grupo de Neurociencias de Antioquia, Sede de Investigación Universitaria SIU, Medellín, Colombia
| | - Andrés Villegas-Lanau
- Grupo de Neurociencias de Antioquia, Sede de Investigación Universitaria SIU, Medellín, Colombia
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire, United Kingdom
| |
Collapse
|
235
|
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: 101] [Impact Index Per Article: 25.3] [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
|
236
|
Shukla R, Singh TR. High-throughput screening of natural compounds and inhibition of a major therapeutic target HsGSK-3β for Alzheimer's disease using computational approaches. J Genet Eng Biotechnol 2021; 19:61. [PMID: 33945025 PMCID: PMC8096881 DOI: 10.1186/s43141-021-00163-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/15/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Alzheimer's disease is a leading neurodegenerative disease worldwide and is the 6th leading cause of death in the USA. AD is a very complex disease and the drugs available in the market cannot fully cure it. The glycogen synthase kinase 3 beta plays a major role in the hyperphosphorylation of tau protein which forms the neurofibrillary tangles which is a major hallmark of AD. In this study, we have used a series of computational approaches to find novel inhibitors against GSK-3β to reduce the TAU hyperphosphorylation. RESULTS We have retrieved a set of compounds (n=167,741) and screened against GSK-3β in four sequential steps. The resulting analysis of virtual screening suggested that 404 compounds show good binding affinity and can be employed for pharmacokinetic analysis. From here, we have selected 20 compounds those were good in terms of pharmacokinetic parameters. All these compounds were re-docked by using Autodock Vina followed by Autodock. Four best compounds were employed for MDS and here predicted RMSD, RMSF, Rg, hydrogen bonds, SASA, PCA, and binding-free energy. From all these analyses, we have concluded that out of 167,741 compounds, the ZINC15968620, ZINC15968622, and ZINC70707119 can act as lead compounds against HsGSK-3β to reduce the hyperphosphorylation. CONCLUSION The study suggested three compounds (ZINC15968620, ZINC15968622, and ZINC70707119) have great potential to be a drug candidate and can be tested using in vitro and in vivo experiments for further characterization and applications.
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, H.P., 173234, India
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, H.P., 173234, India.
| |
Collapse
|
237
|
Habekost M, Qvist P, Denham M, Holm IE, Jørgensen AL. Directly Reprogrammed Neurons Express MAPT and APP Splice Variants Pertinent to Ageing and Neurodegeneration. Mol Neurobiol 2021; 58:2075-2087. [PMID: 33415685 PMCID: PMC8018937 DOI: 10.1007/s12035-020-02258-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Neurons produced by reprogramming of other cell types are used to study cellular mechanisms of age-related neurodegenerative diseases. To model Alzheimer's disease and other tauopathies, it is essential that alternative splicing of the MAPT transcript in these neurons produces the relevant tau isoforms. Human neurons derived from induced pluripotent stem cells, however, express tau isoform compositions characteristic of foetal neurons rather than of adult neurons unless cultured in vitro for extended time periods. In this study, we characterised the dynamics of the MAPT and APP alternative splicing during a developmental time-course of porcine and murine cerebral cortices. We found age-dependent and species-specific isoform composition of MAPT, including 3R and 4R isoforms in the porcine adult brain similar to that of the adult human brain. We converted adult and embryonic fibroblasts directly into induced neurons and found similar developmental patterns of isoform composition, notably, the 3R and 4R isoforms relevant to the pathogenesis of Alzheimer's disease. Also, we observed cell-type-specific isoform expression of APP transcripts during the conversion. The approach was further used to generate induced neurons from transgenic pigs carrying Alzheimer's disease-causing mutations. We show that such neurons authentically model the first crucial steps in AD pathogenesis.
Collapse
Affiliation(s)
- Mette Habekost
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark.
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark.
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- iPSYCH, Lundbeck Foundation Initiative for Integrative Psychiatric Research, 8000C, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, 8000C, Aarhus, Denmark
| | - Mark Denham
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Ida E Holm
- Department of Pathology, Randers Hospital, 8930, Randers, Denmark
- Department of Clinical Medicine, Aarhus University, 8000C, Aarhus, Denmark
| | | |
Collapse
|
238
|
Harwood JC, Leonenko G, Sims R, Escott-Price V, Williams J, Holmans P. Defining functional variants associated with Alzheimer's disease in the induced immune response. Brain Commun 2021; 3:fcab083. [PMID: 33959712 PMCID: PMC8087896 DOI: 10.1093/braincomms/fcab083] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Defining the mechanisms involved in the aetiology of Alzheimer's disease from genome-wide association studies alone is challenging since Alzheimer's disease is polygenic and most genetic variants are non-coding. Non-coding Alzheimer's disease risk variants can influence gene expression by affecting miRNA binding and those located within enhancers and within CTCF sites may influence gene expression through alterations in chromatin states. In addition, their function can be cell-type specific. They can function specifically in microglial enhancers thus affecting gene expression in the brain. Hence, transcriptome-wide association studies have been applied to test the genetic association between disease risk and cell-/tissue-specific gene expression. Many Alzheimer's disease-associated loci are involved in the pathways of the innate immune system. Both microglia, the primary immune cells of the brain, and monocytes which can infiltrate the brain and differentiate into activated macrophages, have roles in neuroinflammation and β-amyloid clearance through phagocytosis. In monocytes the function of regulatory variants can be context-specific after immune stimulation. To dissect the variants associated with Alzheimer's disease in the context of monocytes, we utilized data from naïve monocytes and following immune stimulation in vitro, in combination with genome-wide association studies of Alzheimer's disease in transcriptome-wide association studies. Of the nine genes with statistically independent transcriptome-wide association signals, seven are located in known Alzheimer's disease risk loci: BIN1, PTK2B, SPI1, MS4A4A, MS4A6E, APOE and PVR. The transcriptome-wide association signal for MS4A6E, PTK2B and PVR and the direction of effect replicated in an independent genome-wide association studies. Our analysis identified two novel candidate genes for Alzheimer's disease risk, LACTB2 and PLIN2/ADRP. LACTB2 replicated in a transcriptome-wide association study using independent expression weights. LACTB2 and PLIN2/ADRP are involved in mitochondrial function and lipid metabolism, respectively. Comparison of transcriptome-wide association study results from monocytes, whole blood and brain showed that the signal for PTK2B is specific to blood and MS4A6E is specific to LPS stimulated monocytes.
Collapse
Affiliation(s)
- Janet C Harwood
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Ganna Leonenko
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Valentina Escott-Price
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Julie Williams
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Peter Holmans
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
239
|
Crist AM, Hinkle KM, Wang X, Moloney CM, Matchett BJ, Labuzan SA, Frankenhauser I, Azu NO, Liesinger AM, Lesser ER, Serie DJ, Quicksall ZS, Patel TA, Carnwath TP, DeTure M, Tang X, Petersen RC, Duara R, Graff-Radford NR, Allen M, Carrasquillo MM, Li H, Ross OA, Ertekin-Taner N, Dickson DW, Asmann YW, Carter RE, Murray ME. Transcriptomic analysis to identify genes associated with selective hippocampal vulnerability in Alzheimer's disease. Nat Commun 2021; 12:2311. [PMID: 33875655 PMCID: PMC8055900 DOI: 10.1038/s41467-021-22399-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 03/03/2021] [Indexed: 12/14/2022] Open
Abstract
Selective vulnerability of different brain regions is seen in many neurodegenerative disorders. The hippocampus and cortex are selectively vulnerable in Alzheimer's disease (AD), however the degree of involvement of the different brain regions differs among patients. We classified corticolimbic patterns of neurofibrillary tangles in postmortem tissue to capture extreme and representative phenotypes. We combined bulk RNA sequencing with digital pathology to examine hippocampal vulnerability in AD. We identified hippocampal gene expression changes associated with hippocampal vulnerability and used machine learning to identify genes that were associated with AD neuropathology, including SERPINA5, RYBP, SLC38A2, FEM1B, and PYDC1. Further histologic and biochemical analyses suggested SERPINA5 expression is associated with tau expression in the brain. Our study highlights the importance of embracing heterogeneity of the human brain in disease to identify disease-relevant gene expression.
Collapse
Affiliation(s)
- Angela M Crist
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Kelly M Hinkle
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Xue Wang
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | - Isabelle Frankenhauser
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Paracelsus Medical Private University, Salzburg, Austria
| | - Nkem O Azu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Elizabeth R Lesser
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Daniel J Serie
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Tulsi A Patel
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Troy P Carnwath
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Xiaojia Tang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Ranjan Duara
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
| | | | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Rickey E Carter
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
240
|
Li X, Tsolis KC, Koper MJ, Ronisz A, Ospitalieri S, von Arnim CAF, Vandenberghe R, Tousseyn T, Scheuerle A, Economou A, Carpentier S, Otto M, Thal DR. Sequence of proteome profiles in preclinical and symptomatic Alzheimer's disease. Alzheimers Dement 2021; 17:946-958. [PMID: 33871169 DOI: 10.1002/alz.12345] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022]
Abstract
Proteome profile changes in Alzheimer's disease (AD) brains have been reported. However, it is unclear whether they represent a continuous process, or whether there is a sequential involvement of distinct proteins. To address this question, we used mass spectrometry. We analyzed soluble, dispersible, sodium dodecyl sulfate, and formic acid fractions of neocortex homogenates (mainly Brodmann area 17-19) from 18 pathologically diagnosed preclinical AD, 17 symptomatic AD, and 18 cases without signs of neurodegeneration. By doing so, we identified four groups of AD-related proteins being changed in levels in preclinical and symptomatic AD cases: early-responding, late-responding, gradually-changing, and fraction-shifting proteins. Gene ontology analysis of these proteins and all known AD-risk/causative genes identified vesicle endocytosis and the secretory pathway-related processes as an early-involved AD component. In conclusion, our findings suggest that subtle changes involving the secretory pathway and endocytosis precede severe proteome changes in symptomatic AD as part of the preclinical phase of AD. The respective early-responding proteins may also contribute to synaptic vesicle cycle alterations in symptomatic AD.
Collapse
Affiliation(s)
- Xiaohang Li
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven (University of Leuven), Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Konstantinos C Tsolis
- Laboratory of Molecular Bacteriology, Rega Institute, Department of Microbiology and Immunology, KU Leuven (University of Leuven), Leuven, Belgium
| | - Marta J Koper
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven (University of Leuven), Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.,Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven (University of Leuven), Leuven, Belgium.,Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Alicja Ronisz
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven (University of Leuven), Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Simona Ospitalieri
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven (University of Leuven), Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Christine A F von Arnim
- Department of Neurology, University of Ulm, Ulm, Germany.,Department of Geriatrics, University Medical Center Göttingen, Göttingen, Germany
| | - Rik Vandenberghe
- Department of Neurology, UZ Leuven (University Hospitals Leuven), Leuven, Belgium.,Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven (University of Leuven), Leuven, Belgium
| | - Thomas Tousseyn
- Department of Pathology, UZ Leuven (University Hospitals Leuven), Leuven, Belgium
| | | | - Anastassios Economou
- Laboratory of Molecular Bacteriology, Rega Institute, Department of Microbiology and Immunology, KU Leuven (University of Leuven), Leuven, Belgium
| | - Sebastien Carpentier
- BIOMED facility for SYstems BIOlogy based MAss spectrometry, KU Leuven (University of Leuven), Leuven, Belgium
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven (University of Leuven), Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.,Department of Pathology, UZ Leuven (University Hospitals Leuven), Leuven, Belgium
| |
Collapse
|
241
|
Mold MJ, O’Farrell A, Morris B, Exley C. Aluminum and Tau in Neurofibrillary Tangles in Familial Alzheimer's Disease. J Alzheimers Dis Rep 2021; 5:283-294. [PMID: 34113785 PMCID: PMC8150251 DOI: 10.3233/adr-210011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Familial Alzheimer's disease (fAD) is driven by genetic predispositions affecting the expression and metabolism of the amyloid-β protein precursor. Aluminum is a non-essential yet biologically-reactive metal implicated in the etiology of AD. Recent research has identified aluminum intricately and unequivocally associated with amyloid-β in senile plaques and, more tentatively, co-deposited with neuropil-like threads in the brains of a Colombian cohort of donors with fAD. OBJECTIVE Herein, we have assessed the co-localization of aluminum to immunolabelled phosphorylated tau to probe the potential preferential binding of aluminum to senile plaques or neurofibrillary tangles in the same Colombian kindred. METHODS Herein, we have performed phosphorylated tau-specific immunolabelling followed by aluminum-specific fluorescence microscopy of the identical brain tissue sections via a sequential labelling method. RESULTS Aluminum was co-localized with immunoreactive phosphorylated tau in the brains of donors with fAD. While aluminum was predominantly co-located to neurofibrillary tangles in the temporal cortex, aluminum was more frequently co-deposited with cortical senile plaques. CONCLUSION These data suggest that the co-deposition of aluminum with amyloid-β precedes that with neurofibrillary tangles. Extracellularly deposited amyloid-β may also be more immediately available to bind aluminum versus intracellular aggregates of tau. Therapeutic approaches to reduce tau have demonstrated the amelioration of its synergistic interactions with amyloid-β, ultimately reducing tau pathology and reducing neuronal loss. These data support the intricate associations of aluminum in the neuropathology of fAD, of which its subsequent reduction may further therapeutic benefits observed in ongoing clinical trials in vivo.
Collapse
Affiliation(s)
- Matthew John Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, UK
| | - Adam O’Farrell
- School of Life Sciences, Huxley Building, Keele University, Keele, Staffordshire, UK
| | - Benjamin Morris
- School of Life Sciences, Huxley Building, Keele University, Keele, Staffordshire, UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, UK
| |
Collapse
|
242
|
CRISPR-activated patient fibroblasts for modeling of familial Alzheimer's disease. Neurosci Res 2021; 172:7-12. [PMID: 33819561 DOI: 10.1016/j.neures.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/07/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
Analyzing an appropriate disease model system is important to conduct disease research. Analyzing cells obtained from patient tissues could not only help elucidate the pathological mechanisms and to develop novel therapy but also lead to personalized medicine in the future. However, it is generally difficult to collect and culture neuronal cells from patients suffering from neurodegenerative disorders. Skin fibroblasts are easier to collect than neurons but may not show the expected pathology when disease-relevant genes are not sufficiently expressed. In this article, I describe an in vitro model system that enables the facile analysis of neurological disease mechanisms in patient fibroblast cultures by CRISPR transcriptional activation of endogenous disease-relevant genes. This system introduces an additional platform to analyze neurodegenerative disorders.
Collapse
|
243
|
Cheng Z, Shang Y, Xu X, Dong Z, Zhang Y, Du Z, Lu X, Zhang T. Presenilin 1 mutation likely contributes to U1 small nuclear RNA dysregulation and Alzheimer's disease-like symptoms. Neurobiol Aging 2021; 100:1-10. [PMID: 33450722 DOI: 10.1016/j.neurobiolaging.2020.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022]
Abstract
Previous studies showed that U1 small nuclear RNA (snRNA) was selectively enriched in the brain of individuals with familial Alzheimer's disease (AD), resulting in widespread changes in RNA splicing. Our study further reported that presenilin-1 (PSEN1) induced an increase in U1 snRNA expression, accompanied by changed amyloid precursor protein expression, β-amyloid level, and cell death in SH-SY5Y cells. However, the effect of U1 snRNA overexpression on learning and memory is still unclear. In the present study, we found that neuronal U1 snRNA overexpression could generate U1 snRNA aggregates in the nuclear, accompanied by the widespread alteration of RNA splicing, resulting in the impairments of synaptic plasticity and spatial memory. In addition, more U1 snRNAs is bound to the intron binding sites accompanied by an increased intracellular U1 snRNA level. This suggests that U1 snRNA overexpression regulates RNA splicing and gene expression in neurons by manipulating the recruitment of the U1 snRNA to the nascent transcripts. Using in situ hybridization staining of human central nervous system-type neurons, we identified nuclear aggregates of U1 snRNA in neurons by upregulating the U1 snRNA level. Quantitative polymerase chain reaction analysis showed U1 snRNA accumulation in the insoluble fraction of neurons with PSEN1 mutation neurons rather than other types of U snRNAs. These results show an independent function of U1 snRNA in regulating RNA splicing, suggesting that aberrant RNA processing may mediate neurodegeneration induced by PSEN1 mutation.
Collapse
Affiliation(s)
- Zhi Cheng
- College of Life Sciences & State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, PR China
| | - Yingchun Shang
- College of Life Sciences & State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, PR China
| | - Xinxin Xu
- College of Life Sciences & State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, PR China
| | - Zhiqiang Dong
- College of Life Sciences & State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, PR China
| | - Yongwang Zhang
- College of Pharmacy, Nankai University, Tianjin, PR China
| | - Zhanqiang Du
- College of Life Sciences & State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, PR China
| | - Xinyi Lu
- College of Life Sciences & State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, PR China; College of Pharmacy, Nankai University, Tianjin, PR China
| | - Tao Zhang
- College of Life Sciences & State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, PR China.
| |
Collapse
|
244
|
Duran-Aniotz C, Moreno-Gonzalez I, Gamez N, Perez-Urrutia N, Vegas-Gomez L, Soto C, Morales R. Amyloid pathology arrangements in Alzheimer's disease brains modulate in vivo seeding capability. Acta Neuropathol Commun 2021; 9:56. [PMID: 33785065 PMCID: PMC8008576 DOI: 10.1186/s40478-021-01155-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/14/2021] [Indexed: 12/23/2022] Open
Abstract
Amyloid-β (Aβ) misfolding is one of the hallmark pathological features of Alzheimer's disease (AD). AD can manifest with diverse symptomatology including variable rates of cognitive decline, duration of clinical disease, and other detrimental changes. Several reports suggest that conformational diversity in misfolded Aβ is a leading factor for clinical variability in AD, analogous to what it has been described for prion strains in prion diseases. Notably, prion strains generate diverse patterns of misfolded protein deposition in the brains of affected individuals. Here, we tested the in vivo prion-like transmission features of four AD brains displaying particular patterns of amyloidosis. AD brains induced different phenotypes in recipient mice, as evaluated by their specific seeding activity, as well as the total amount of Aβ deposited surrounding vascular structures and the reactivity of amyloid pathology to thioflavin S. Our results support the notion that AD-subtypes are encoded in disease-associated Aβ. Further research exploring whether AD include a spectrum of different clinical conditions or syndromes may pave the way to personalized diagnosis and treatments.
Collapse
Affiliation(s)
- Claudia Duran-Aniotz
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Universidad de los Andes, Facultad de Medicina, Av. San Carlos de Apoquindo, 2200, Las Condes, Santiago, Chile
| | - Ines Moreno-Gonzalez
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Department of Cell Biology, Faculty of Sciences, University of Malaga-IBIMA, 29010, Malaga, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Centro Integrativo de Biologia Y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Nazaret Gamez
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Department of Cell Biology, Faculty of Sciences, University of Malaga-IBIMA, 29010, Malaga, Spain
| | - Nelson Perez-Urrutia
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
| | - Laura Vegas-Gomez
- Department of Cell Biology, Faculty of Sciences, University of Malaga-IBIMA, 29010, Malaga, Spain
| | - Claudio Soto
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Universidad de los Andes, Facultad de Medicina, Av. San Carlos de Apoquindo, 2200, Las Condes, Santiago, Chile
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA.
- Centro Integrativo de Biologia Y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile.
| |
Collapse
|
245
|
Unzeta M, Hernàndez-Guillamon M, Sun P, Solé M. SSAO/VAP-1 in Cerebrovascular Disorders: A Potential Therapeutic Target for Stroke and Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22073365. [PMID: 33805974 PMCID: PMC8036996 DOI: 10.3390/ijms22073365] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/24/2022] Open
Abstract
The semicarbazide-sensitive amine oxidase (SSAO), also known as vascular adhesion protein-1 (VAP-1) or primary amine oxidase (PrAO), is a deaminating enzyme highly expressed in vessels that generates harmful products as a result of its enzymatic activity. As a multifunctional enzyme, it is also involved in inflammation through its ability to bind and promote the transmigration of circulating leukocytes into inflamed tissues. Inflammation is present in different systemic and cerebral diseases, including stroke and Alzheimer’s disease (AD). These pathologies show important affectations on cerebral vessels, together with increased SSAO levels. This review summarizes the main roles of SSAO/VAP-1 in human physiology and pathophysiology and discusses the mechanisms by which it can affect the onset and progression of both stroke and AD. As there is an evident interrelationship between stroke and AD, basically through the vascular system dysfunction, the possibility that SSAO/VAP-1 could be involved in the transition between these two pathologies is suggested. Hence, its inhibition is proposed to be an interesting therapeutical approach to the brain damage induced in these both cerebral pathologies.
Collapse
Affiliation(s)
- Mercedes Unzeta
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences, Universitat Auònoma de Barcelona, 08193 Barcelona, Spain;
| | - Mar Hernàndez-Guillamon
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
- Correspondence: ; Tel.: +34-934-896-766
| | - Ping Sun
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| |
Collapse
|
246
|
Carpanini SM, Harwood JC, Baker E, Torvell M, The GERAD1 Consortium, Sims R, Williams J, Morgan BP. The Impact of Complement Genes on the Risk of Late-Onset Alzheimer's Disease. Genes (Basel) 2021; 12:443. [PMID: 33804666 PMCID: PMC8003605 DOI: 10.3390/genes12030443] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 12/27/2022] Open
Abstract
Late-onset Alzheimer's disease (LOAD), the most common cause of dementia, and a huge global health challenge, is a neurodegenerative disease of uncertain aetiology. To deliver effective diagnostics and therapeutics, understanding the molecular basis of the disease is essential. Contemporary large genome-wide association studies (GWAS) have identified over seventy novel genetic susceptibility loci for LOAD. Most are implicated in microglial or inflammatory pathways, bringing inflammation to the fore as a candidate pathological pathway. Among the most significant GWAS hits are three complement genes: CLU, encoding the fluid-phase complement inhibitor clusterin; CR1 encoding complement receptor 1 (CR1); and recently, C1S encoding the complement enzyme C1s. Complement activation is a critical driver of inflammation; changes in complement genes may impact risk by altering the inflammatory status in the brain. To assess complement gene association with LOAD risk, we manually created a comprehensive complement gene list and tested these in gene-set analysis with LOAD summary statistics. We confirmed associations of CLU and CR1 genes with LOAD but showed no significant associations for the complement gene-set when excluding CLU and CR1. No significant association with other complement genes, including C1S, was seen in the IGAP dataset; however, these may emerge from larger datasets.
Collapse
Affiliation(s)
- Sarah M. Carpanini
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff, CF24 4HQ, UK; (S.M.C.); (E.B.); (M.T.); (J.W.)
- Division of Infection and Immunity, School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | - Janet C. Harwood
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK; (J.C.H.); (R.S.)
| | - Emily Baker
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff, CF24 4HQ, UK; (S.M.C.); (E.B.); (M.T.); (J.W.)
| | - Megan Torvell
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff, CF24 4HQ, UK; (S.M.C.); (E.B.); (M.T.); (J.W.)
- Division of Infection and Immunity, School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | | | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK; (J.C.H.); (R.S.)
| | - Julie Williams
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff, CF24 4HQ, UK; (S.M.C.); (E.B.); (M.T.); (J.W.)
| | - B. Paul Morgan
- UK Dementia Research Institute at Cardiff University, School of Medicine, Cardiff, CF24 4HQ, UK; (S.M.C.); (E.B.); (M.T.); (J.W.)
- Division of Infection and Immunity, School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
247
|
Siddappaji KK, Gopal S. Molecular mechanisms in Alzheimer's disease and the impact of physical exercise with advancements in therapeutic approaches. AIMS Neurosci 2021; 8:357-389. [PMID: 34183987 PMCID: PMC8222772 DOI: 10.3934/neuroscience.2021020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/16/2021] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common, severe neurodegenerative brain disorder characterized by the accumulation of amyloid-beta plaques, neurofibrillary tangles in the brain causing neural disintegration, synaptic dysfunction, and neuronal death leading to dementia. Although many US-FDA-approved drugs like Donepezil, Rivastigmine, Galantamine are available in the market, their consumption reduces only the symptoms of the disease but fails in potency to cure the disease. This disease affects many individuals with aging. Combating the disease tends to be very expensive. This review focuses on biochemical mechanisms in the neuron both at normal and AD state with relevance to the tau hypothesis, amyloid hypothesis, the risk factors influencing dementia, oxidative stress, and neuroinflammation altogether integrated with neurodegeneration. A brief survey is carried out on available biomarkers in the diagnosis of the disease, drugs used for the treatment, and the challenges in approaching therapeutic targets in inhibiting the disease pathologies. This review conjointly assesses the demerits with the inefficiency of drugs to reach targets, their side effects, and toxicity. Optimistically, this review directs on the advantageous strategies in using nanotechnology-based drug delivery systems to cross the blood-brain barrier for improving the efficacy of drugs combined with a novel neuronal stem cell therapy approach. Determinately, this review aims at the natural, non-therapeutic healing impact of physical exercise on different model organisms and the effect of safe neuromodulation treatments using repetitive Transcranial Magnetic Stimulation (rTMS), transcranial Electrical Stimulation (tES) in humans to control the disease pathologies prominent in enhancing the synaptic function.
Collapse
Affiliation(s)
| | - Shubha Gopal
- Department of Studies in Microbiology, University of Mysore, Mysuru, 570006, Karnataka, India
| |
Collapse
|
248
|
López-Ortiz S, Pinto-Fraga J, Valenzuela PL, Martín-Hernández J, Seisdedos MM, García-López O, Toschi N, Di Giuliano F, Garaci F, Mercuri NB, Nisticò R, Emanuele E, Lista S, Lucia A, Santos-Lozano A. Physical Exercise and Alzheimer's Disease: Effects on Pathophysiological Molecular Pathways of the Disease. Int J Mol Sci 2021; 22:ijms22062897. [PMID: 33809300 PMCID: PMC7999827 DOI: 10.3390/ijms22062897] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD), the most common form of neurodegenerative dementia in adults worldwide, is a multifactorial and heterogeneous disorder characterized by the interaction of genetic and epigenetic factors and the dysregulation of numerous intracellular signaling and cellular/molecular pathways. The introduction of the systems biology framework is revolutionizing the study of complex diseases by allowing the identification and integration of cellular/molecular pathways and networks of interaction. Here, we reviewed the relationship between physical activity and the next pathophysiological processes involved in the risk of developing AD, based on some crucial molecular pathways and biological process dysregulated in AD: (1) Immune system and inflammation; (2) Endothelial function and cerebrovascular insufficiency; (3) Apoptosis and cell death; (4) Intercellular communication; (5) Metabolism, oxidative stress and neurotoxicity; (6) DNA damage and repair; (7) Cytoskeleton and membrane proteins; (8) Synaptic plasticity. Moreover, we highlighted the increasingly relevant role played by advanced neuroimaging technologies, including structural/functional magnetic resonance imaging, diffusion tensor imaging, and arterial spin labelling, in exploring the link between AD and physical exercise. Regular physical exercise seems to have a protective effect against AD by inhibiting different pathophysiological molecular pathways implicated in AD.
Collapse
Affiliation(s)
- Susana López-Ortiz
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Jose Pinto-Fraga
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Pedro L. Valenzuela
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
| | - Juan Martín-Hernández
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - María M. Seisdedos
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Oscar García-López
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.T.); (F.G.)
- Department of Radiology, “Athinoula A. Martinos” Center for Biomedical Imaging, Boston, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Francesca Di Giuliano
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.T.); (F.G.)
- Casa di Cura “San Raffaele Cassino”, 03043 Cassino, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy;
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00161 Rome, Italy;
- School of Pharmacy, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Simone Lista
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
- School of Pharmacy, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
- Research Institute of the Hospital 12 de Octubre (“imas12”), 28041 Madrid, Spain
- Centro de Investigación Biomeédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), 28029 Madrid, Spain
- Correspondence:
| | - Alejandro Santos-Lozano
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
- Research Institute of the Hospital 12 de Octubre (“imas12”), 28041 Madrid, Spain
| |
Collapse
|
249
|
Raybould R, Sims R. Searching the Dark Genome for Alzheimer's Disease Risk Variants. Brain Sci 2021; 11:332. [PMID: 33800766 PMCID: PMC7999247 DOI: 10.3390/brainsci11030332] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Sporadic Alzheimer's disease (AD) is a complex genetic disease, and the leading cause of dementia worldwide. Over the past 3 decades, extensive pioneering research has discovered more than 70 common and rare genetic risk variants. These discoveries have contributed massively to our understanding of the pathogenesis of AD but approximately half of the heritability for AD remains unaccounted for. There are regions of the genome that are not assayed by mainstream genotype and sequencing technology. These regions, known as the Dark Genome, often harbour large structural DNA variants that are likely relevant to disease risk. Here, we describe the dark genome and review current technological and bioinformatics advances that will enable researchers to shed light on these hidden regions of the genome. We highlight the potential importance of the hidden genome in complex disease and how these strategies will assist in identifying the missing heritability of AD. Identification of novel protein-coding structural variation that increases risk of AD will open new avenues for translational research and new drug targets that have the potential for clinical benefit to delay or even prevent clinical symptoms of disease.
Collapse
Affiliation(s)
- Rachel Raybould
- UK Dementia Research Institute in Cardiff, Haydn Ellis Building, Cardiff University, Wales CF24 4HQ, UK
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Wales CF24 4HQ, UK
| |
Collapse
|
250
|
Flexible and Accurate Substrate Processing with Distinct Presenilin/γ-Secretases in Human Cortical Neurons. eNeuro 2021; 8:ENEURO.0500-20.2021. [PMID: 33608391 PMCID: PMC7932187 DOI: 10.1523/eneuro.0500-20.2021] [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] [Received: 11/21/2020] [Revised: 01/27/2021] [Accepted: 01/30/2021] [Indexed: 01/10/2023] Open
Abstract
Mutations in the presenilin genes (PS1, PS2) have been linked to the majority of familial Alzheimer’s disease (AD). Although great efforts have been made to investigate pathogenic PS mutations, which ultimately cause an increase in the toxic form of β-amyloid (Aβ), the intrinsic physiological functions of PS in human neurons remain to be determined. In this study, to investigate the physiological roles of PS in human neurons, we generated PS1 conditional knock-out (KO) induced pluripotent stem cells (iPSCs), in which PS1 can be selectively abrogated under Cre transduction with or without additional PS2 KO. We showed that iPSC-derived neural progenitor cells (NPCs) do not confer a maintenance ability in the absence of both PS1 and PS2, showing the essential role of PS in Notch signaling. We then generated PS-null human cortical neurons, where PS1 was intact until full neuronal differentiation occurred. Aβ40 production was reduced exclusively in human PS1/PS2-null neurons along with a concomitant accumulation of amyloid β precursor protein (APP)-C-terminal fragments CTFs, whereas Aβ42 was decreased in neurons devoid of PS2. Unlike previous studies in mice, in which APP cleavage is largely attributable to PS1, γ-secretase activity seemed to be comparable between PS1 and PS2. In contrast, cleavage of another substrate, N-cadherin, was impaired only in neurons devoid of PS1. Moreover, PS2/γ-secretase exists largely in late endosomes/lysosomes, as measured by specific antibody against the γ-secretase complex, in which Aβ42 species are supposedly produced. Using this novel stem cell-based platform, we assessed important physiological PS1/PS2 functions in mature human neurons, the dysfunction of which could underlie AD pathogenesis.
Collapse
|