1
|
Ma L, Wang F, Chen S, Wang S, Wang Z, Xia M, Li Y, Ma H, Shang J, Zhang J. Probable Novel APP Met671Leu Mutation in a Chinese Han Family with Early-Onset Alzheimer's Disease. Neuromolecular Med 2024; 26:6. [PMID: 38504005 DOI: 10.1007/s12017-023-08770-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/19/2023] [Indexed: 03/21/2024]
Abstract
Familial Alzheimer's disease (AD) is a rare disease caused by autosomal-dominant mutations. APP (encoding amyloid precursor protein), PSEN1 (encoding presenilin 1), and PSEN2 (encoding presenilin 2) are the most common genes cause dominant inherited AD. This study aimed to demonstrate a Chinese early-onset AD pedigree presenting as progressive memory impairment, apraxia, visual-spatial disorders, psychobehavioral disorders, and personality changes with a novel APP gene mutation. The family contains four patients, three carries and three normal family members. The proband underwent brain magnetic resonance imaging (MRI), 18F-fludeoxyglucose positron emission tomography (18F-FDG-PET), cerebrospinal fluid amyloid detection, 18F-florbetapir (AV-45) Positron Emission Computed Tomography (PET) imaging, whole-exome sequencing and Sanger sequencing. Brain MRI images showed brain atrophy, especially in the entorhinal cortex, temporal hippocampus, and lateral ventricle dilation. The FDG-PET showed hypometabolism in the frontotemporal, parietal, and hippocampal regions. 18F-florbetapir (AV-45) PET imaging showed cerebral cortex Aβ protein deposition. The cerebrospinal fluid amyloid protein test showed Aβ42/Aβ40 ratio decreases, pathological phosphor-tau level increases. Whole-exome sequencing detected a new missense mutation of codon 671 (M671L), which was a heterozygous A to T point mutation at position 2011 (c.2011A > T) in exon 16 of the amyloid precursor protein, resulting in the replacement of methionine to Leucine. The co-separation analysis was validated in this family. The mutation was found in 3 patients, 3 clinical normal members in the family, but not in the other 3 unaffected family members, 100 unrelated normal subjects, or 100 sporadic patients with AD. This mutation was probably pathogenic and novel in a Chinese Han family with early-onset AD.
Collapse
Affiliation(s)
- Limin Ma
- Department of Health Management Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fengyu Wang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuai Chen
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shenghui Wang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenzhen Wang
- Department of Radiology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingrong Xia
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yongli Li
- Department of Health Management Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huimin Ma
- Department of Health Management Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junkui Shang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Sidiropoulou GA, Metaxas A, Kourti M. Natural antioxidants that act against Alzheimer's disease through modulation of the NRF2 pathway: a focus on their molecular mechanisms of action. Front Endocrinol (Lausanne) 2023; 14:1217730. [PMID: 37465125 PMCID: PMC10351420 DOI: 10.3389/fendo.2023.1217730] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/01/2023] [Indexed: 07/20/2023] Open
Abstract
Characterized by a complex pathophysiology that includes the intraneuronal formation of neurofibrillary tangles and the extracellular deposition of β-amyloid plaques, Alzheimer's disease (AD) is a terminal neurodegenerative disease that causes dementia in older adults. Oxidative stress in the brain is considered as one of the contributing factors to the pathogenesis of AD, and thus, antioxidants have attracted much interest as potential therapeutic agents against the disorder. Natural antioxidants are typically characterized by low acute and chronic toxicity, which facilitates their potential therapeutic application. One important molecular target for the beneficial effects of natural antioxidants is the nuclear factor erythroid-derived 2-related factor 2 (NFE2L2/NRF2). NRF2 is a key transcription factor that orchestrates the cellular antioxidant response through regulating the expression of oxidative stress-related genes harboring the antioxidant response element (ARE) in their promoters. Indeed, in the case of excessive oxidative damage, NRF2 migrates to the nucleus and binds to ARE, activating the transcription of antioxidant protector genes. There is increasing evidence that NRF2 is implicated in AD pathology through dysfunction and altered localization, which renders it as a potential therapeutic target for AD. Thus, this review summarizes the most recent (2018-2023) advances on the NRF2-modulating activity of natural antioxidants observed in vitro and in AD animal models. This information will help elucidate the molecular mechanisms governing the antioxidant activity of such phytochemicals to highlight their therapeutic potential against common neurodegenerative diseases, such as AD.
Collapse
Affiliation(s)
- Grammatiki Alexandra Sidiropoulou
- Angiogenesis and Cancer Drug Discovery Group, Basic and Translational Cancer Research Centre, European University Cyprus, Nicosia, Cyprus
| | - Athanasios Metaxas
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Malamati Kourti
- Angiogenesis and Cancer Drug Discovery Group, Basic and Translational Cancer Research Centre, European University Cyprus, Nicosia, Cyprus
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
3
|
The Role of Aldose Reductase in Beta-Amyloid-Induced Microglia Activation. Int J Mol Sci 2022; 23:ijms232315088. [PMID: 36499422 PMCID: PMC9739496 DOI: 10.3390/ijms232315088] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The occurrence of Alzheimer's disease has been associated with the accumulation of beta-amyloid (β-amyloid) plaques. These plaques activate microglia to secrete inflammatory molecules, which damage neurons in the brain. Thus, understanding the underlying mechanism of microglia activation can provide a therapeutic strategy for alleviating microglia-induced neuroinflammation. The aldose reductase (AR) enzyme catalyzes the reduction of glucose to sorbitol in the polyol pathway. In addition to mediating diabetic complications in hyperglycemic environments, AR also helps regulate inflammation in microglia. However, little is known about the role of AR in β-amyloid-induced inflammation in microglia and subsequent neuronal death. In this study, we confirmed that AR inhibition attenuates increased β-amyloid-induced reactive oxygen species and tumor necrosis factor α secretion by suppressing ERK signaling in BV2 cells. In addition, we are the first to report that AR inhibition reduced the phagocytotic capability and cell migration of BV2 cells in response to β-amyloid. To further investigate the protective role of the AR inhibitor sorbinil in neurons, we co-cultured β-amyloid-induced microglia with stem cell-induced neurons. sorbinil ameliorated neuronal damage in both cells in the co-culture system. In summary, our findings reveal AR regulation of microglia activation as a novel therapeutic target for Alzheimer's disease.
Collapse
|
4
|
Shao W, Xiang S, Zhang Z, Huang K, Zhang J. Hyper-graph based sparse canonical correlation analysis for the diagnosis of Alzheimer's disease from multi-dimensional genomic data. Methods 2021; 189:86-94. [PMID: 32360353 DOI: 10.1016/j.ymeth.2020.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/30/2020] [Accepted: 04/23/2020] [Indexed: 10/24/2022] Open
Abstract
The effective and accurate diagnosis of Alzheimer's disease (AD), especially in the early stage (i.e., mild cognitive impairment (MCI)) remains a big challenge in AD research. So far, multiple biomarkers have been associated with AD diagnosis and progression. However, most of the existing research only utilized single modality data for diagnostic biomarker identification, which did not take the advantages of multi-modal data that provide comprehensive and complementary information at multiple levels into consideration. In this paper, we integrate multi-modal genomic data from postmortem AD brains (i.e., mRNA, miRNA and epigenomic data) and propose a hyper-graph based sparse canonical correlation analysis (HGSCCA) method to extract the most correlated multi-modal biomarkers associated with AD and MCI. Specifically, our model utilizes the sparse canonical correlation analysis framework (SCCA), which aims at finding the best linear projections for each input modality so that the strongest correlation within the selected features of multi-dimensional genomic data can be captured. In addition, with the consideration of high-order relationships among different subjects, we also introduce a hyper-graph-based regularization term that will lead to the selection of more discriminative biomarkers. To evaluate the effectiveness of the proposed method, we conduct the experiments on the well-known AD cohort study, The Religious Orders Study and Memory and Aging Project (ROSMAP) dataset, and the results show that our method can not only identify meaningful biomarkers for the diagnosis AD disease, but also achieve superior classification performance than the comparing methods.
Collapse
Affiliation(s)
- Wei Shao
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Shunian Xiang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, China; Department of Medical & Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Zuoyi Zhang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Regenstrief Institute, Indianapolis, IN 46202, USA
| | - Kun Huang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202 USA; Regenstrief Institute, Indianapolis, IN 46202, USA.
| | - Jie Zhang
- Department of Medical & Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
5
|
Flitsch LJ, Laupman KE, Brüstle O. Transcription Factor-Based Fate Specification and Forward Programming for Neural Regeneration. Front Cell Neurosci 2020; 14:121. [PMID: 32508594 PMCID: PMC7251072 DOI: 10.3389/fncel.2020.00121] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
Traditionally, in vitro generation of donor cells for brain repair has been dominated by the application of extrinsic growth factors and morphogens. Recent advances in cell engineering strategies such as reprogramming of somatic cells into induced pluripotent stem cells and direct cell fate conversion have impressively demonstrated the feasibility to manipulate cell identities by the overexpression of cell fate-determining transcription factors. These strategies are now increasingly implemented for transcription factor-guided differentiation of neural precursors and forward programming of pluripotent stem cells toward specific neural subtypes. This review covers major achievements, pros and cons, as well as future prospects of transcription factor-based cell fate specification and the applicability of these approaches for the generation of donor cells for brain repair.
Collapse
Affiliation(s)
- Lea J Flitsch
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Karen E Laupman
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, Germany
| |
Collapse
|
6
|
Dou KX, Zhang C, Tan CC, Xu W, Li JQ, Cao XP, Tan L, Yu JT. Genome-wide association study identifies CBFA2T3 affecting the rate of CSF Aβ 42 decline in non-demented elders. Aging (Albany NY) 2019; 11:5433-5444. [PMID: 31370031 PMCID: PMC6710044 DOI: 10.18632/aging.102125] [Citation(s) in RCA: 3] [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: 05/07/2019] [Accepted: 07/21/2019] [Indexed: 11/30/2022]
Abstract
Brain amyloid deposition is an early pathological event in Alzheimer's disease (AD), and abnormally low levels amyloid-β42 peptide (Aβ42) in cerebrospinal fluid (CSF) can be detected in preclinical AD. To identify the genetic determinants that regulate the rate of CSF Aβ42 decline among non-demented elders, we conducted a genome-wide association study involved 321 non-demented elders from Alzheimer's Disease Neuroimaging Initiative (ADNI) 1/GO/2 cohorts restricted to non-Hispanic Caucasians. A novel genome-wide significant association of higher annualized percent decline of CSF Aβ42 in the gene CBFA2T3 (CBFA2/RUNX1 translocation partner 3; rs13333659-T; p = 2.24 × 10-9) was identified. Besides displaying abnormal CSF Aβ42 levels, rs13333659-T carriers were more likely to exhibit a greater longitudinal cognitive decline (p = 0.029, β = 0.097) and hippocampal atrophy (p = 0.029, β = -0.160) in the non-demented elders, especially for the participants who were amyloid-positive at baseline. These findings suggest rs13333659 in CBFA2T3 as a risk locus to modulate the decline rate of CSF Aβ42 preceding the onset of clinical symptoms.
Collapse
Affiliation(s)
- Kai-Xin Dou
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Can Zhang
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Jie-Qiong Li
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | |
Collapse
|
7
|
Ashton NJ, Nevado-Holgado AJ, Barber IS, Lynham S, Gupta V, Chatterjee P, Goozee K, Hone E, Pedrini S, Blennow K, Schöll M, Zetterberg H, Ellis KA, Bush AI, Rowe CC, Villemagne VL, Ames D, Masters CL, Aarsland D, Powell J, Lovestone S, Martins R, Hye A. A plasma protein classifier for predicting amyloid burden for preclinical Alzheimer's disease. SCIENCE ADVANCES 2019; 5:eaau7220. [PMID: 30775436 PMCID: PMC6365111 DOI: 10.1126/sciadv.aau7220] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/19/2018] [Indexed: 05/03/2023]
Abstract
A blood-based assessment of preclinical disease would have huge potential in the enrichment of participants for Alzheimer's disease (AD) therapeutic trials. In this study, cognitively unimpaired individuals from the AIBL and KARVIAH cohorts were defined as Aβ negative or Aβ positive by positron emission tomography. Nontargeted proteomic analysis that incorporated peptide fractionation and high-resolution mass spectrometry quantified relative protein abundances in plasma samples from all participants. A protein classifier model was trained to predict Aβ-positive participants using feature selection and machine learning in AIBL and independently assessed in KARVIAH. A 12-feature model for predicting Aβ-positive participants was established and demonstrated high accuracy (testing area under the receiver operator characteristic curve = 0.891, sensitivity = 0.78, and specificity = 0.77). This extensive plasma proteomic study has unbiasedly highlighted putative and novel candidates for AD pathology that should be further validated with automated methodologies.
Collapse
Affiliation(s)
- Nicholas J. Ashton
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Steven Lynham
- Proteomics Core Facility, James Black Centre, King’s College, London, UK
| | - Veer Gupta
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
- School of Medicine, Faculty of Health, Deakin University, 3220 VIC, Australia
| | - Pratishtha Chatterjee
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- KaRa Institute of Neurological Diseases, Macquarie Park, NSW, Australia
- Department of Biomedical Sciences, Macquarie University, 2109, NSW, Australia
| | - Kathryn Goozee
- KaRa Institute of Neurological Diseases, Macquarie Park, NSW, Australia
- Department of Biomedical Sciences, Macquarie University, 2109, NSW, Australia
- Clinical Research Department, Anglicare, Sydney, NSW, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, WA, Australia
| | - Eugene Hone
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
| | - Steve Pedrini
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kathryn A. Ellis
- Academic Unit for Psychiatry of Old Age, St. George’s Hospital, University of Melbourne, VIC, Australia
| | - Ashley I. Bush
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
- The Florey Institute, University of Melbourne, VIC, Australia
| | - Christopher C. Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Victor L. Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St. George’s Hospital, University of Melbourne, VIC, Australia
- National Ageing Research Institute, Parkville, VIC, Australia
| | | | - Dag Aarsland
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - John Powell
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | | | - Ralph Martins
- School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton South, VIC, Australia
- KaRa Institute of Neurological Diseases, Macquarie Park, NSW, Australia
- Department of Biomedical Sciences, Macquarie University, 2109, NSW, Australia
| | - Abdul Hye
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Corresponding author.
| |
Collapse
|
8
|
Coronel R, Palmer C, Bernabeu-Zornoza A, Monteagudo M, Rosca A, Zambrano A, Liste I. Physiological effects of amyloid precursor protein and its derivatives on neural stem cell biology and signaling pathways involved. Neural Regen Res 2019; 14:1661-1671. [PMID: 31169172 PMCID: PMC6585543 DOI: 10.4103/1673-5374.257511] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The pathological implication of amyloid precursor protein (APP) in Alzheimer's disease has been widely documented due to its involvement in the generation of amyloid-β peptide. However, the physiological functions of APP are still poorly understood. APP is considered a multimodal protein due to its role in a wide variety of processes, both in the embryo and in the adult brain. Specifically, APP seems to play a key role in the proliferation, differentiation and maturation of neural stem cells. In addition, APP can be processed through two canonical processing pathways, generating different functionally active fragments: soluble APP-α, soluble APP-β, amyloid-β peptide and the APP intracellular C-terminal domain. These fragments also appear to modulate various functions in neural stem cells, including the processes of proliferation, neurogenesis, gliogenesis or cell death. However, the molecular mechanisms involved in these effects are still unclear. In this review, we summarize the physiological functions of APP and its main proteolytic derivatives in neural stem cells, as well as the possible signaling pathways that could be implicated in these effects. The knowledge of these functions and signaling pathways involved in the onset or during the development of Alzheimer's disease is essential to advance the understanding of the pathogenesis of Alzheimer's disease, and in the search for potential therapeutic targets.
Collapse
Affiliation(s)
- Raquel Coronel
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Charlotte Palmer
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Adela Bernabeu-Zornoza
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - María Monteagudo
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Andreea Rosca
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Alberto Zambrano
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| |
Collapse
|
9
|
Neuronal and Glial Differentiation of Human Neural Stem Cells Is Regulated by Amyloid Precursor Protein (APP) Levels. Mol Neurobiol 2018; 56:1248-1261. [DOI: 10.1007/s12035-018-1167-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/30/2018] [Indexed: 12/31/2022]
|
10
|
Southam KA, Stennard F, Pavez C, Small DH. Knockout of Amyloid β Protein Precursor (APP) Expression Alters Synaptogenesis, Neurite Branching and Axonal Morphology of Hippocampal Neurons. Neurochem Res 2018; 44:1346-1355. [PMID: 29572646 DOI: 10.1007/s11064-018-2512-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/15/2018] [Accepted: 03/20/2018] [Indexed: 12/23/2022]
Abstract
The function of the β-A4 amyloid protein precursor (APP) of Alzheimer's disease (AD) remains unclear. APP has a number of putative roles in neuronal differentiation, survival, synaptogenesis and cell adhesion. In this study, we examined the development of axons, dendrites and synapses in cultures of hippocampus neutrons derived from APP knockout (KO) mice. We report that loss of APP function reduces the branching of cultured hippocampal neurons, resulting in reduced synapse formation. Using a compartmentalised culture approach, we found reduced axonal outgrowth in cultured hippocampal neurons and we also identified abnormal growth characteristics of isolated hippocampal neuron axons. Although APP has previously been suggested to play an important role in promoting cell adhesion, we surprisingly found that APPKO hippocampal neurons adhered more strongly to a poly-L-lysine substrate and their neurites displayed an increased density of focal adhesion puncta. The findings suggest that the function of APP has an important role in both dendritic and axonal growth and that endogenous APP may regulate substrate adhesion of hippocampal neurons. The results may explain neuronal and synaptic morphological abnormalities in APPKO mice and the presence of abnormal APP expression in dystrophic neurites around amyloid deposits in AD.
Collapse
Affiliation(s)
- Katherine A Southam
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia. .,Faculty of Health, School of Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia.
| | - Fiona Stennard
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Cassandra Pavez
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - David H Small
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| |
Collapse
|
11
|
Role of Amyloid Precursor Protein (APP) and Its Derivatives in the Biology and Cell Fate Specification of Neural Stem Cells. Mol Neurobiol 2018; 55:7107-7117. [DOI: 10.1007/s12035-018-0914-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/18/2018] [Indexed: 01/31/2023]
|
12
|
Wang X, Zhou X, Li G, Zhang Y, Wu Y, Song W. Modifications and Trafficking of APP in the Pathogenesis of Alzheimer's Disease. Front Mol Neurosci 2017; 10:294. [PMID: 28966576 PMCID: PMC5605621 DOI: 10.3389/fnmol.2017.00294] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/31/2017] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder, is the leading cause of dementia. Neuritic plaque, one of the major characteristics of AD neuropathology, mainly consists of amyloid β (Aβ) protein. Aβ is derived from amyloid precursor protein (APP) by sequential cleavages of β- and γ-secretase. Although APP upregulation can promote AD pathogenesis by facilitating Aβ production, growing evidence indicates that aberrant post-translational modifications and trafficking of APP play a pivotal role in AD pathogenesis by dysregulating APP processing and Aβ generation. In this report, we reviewed the current knowledge of APP modifications and trafficking as well as their role in APP processing. More importantly, we discussed the effect of aberrant APP modifications and trafficking on Aβ generation and the underlying mechanisms, which may provide novel strategies for drug development in AD.
Collapse
Affiliation(s)
- Xin Wang
- Department of Psychiatry, Jining Medical UniversityJining, China.,Shandong Key Laboratory of Behavioral Medicine, Jining Medical UniversityJining, China
| | - Xuan Zhou
- Department of Psychiatry, Jining Medical UniversityJining, China.,Shandong Key Laboratory of Behavioral Medicine, Jining Medical UniversityJining, China
| | - Gongying Li
- Department of Psychiatry, Jining Medical UniversityJining, China.,Shandong Key Laboratory of Behavioral Medicine, Jining Medical UniversityJining, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical UniversityJining, China
| | - Yun Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British ColumbiaVancouver, BC, Canada
| | - Yili Wu
- Department of Psychiatry, Jining Medical UniversityJining, China.,Shandong Key Laboratory of Behavioral Medicine, Jining Medical UniversityJining, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical UniversityJining, China.,Townsend Family Laboratories, Department of Psychiatry, The University of British ColumbiaVancouver, BC, Canada
| | - Weihong Song
- Townsend Family Laboratories, Department of Psychiatry, The University of British ColumbiaVancouver, BC, Canada
| |
Collapse
|
13
|
Brzdak P, Nowak D, Wiera G, Mozrzymas JW. Multifaceted Roles of Metzincins in CNS Physiology and Pathology: From Synaptic Plasticity and Cognition to Neurodegenerative Disorders. Front Cell Neurosci 2017; 11:178. [PMID: 28713245 PMCID: PMC5491558 DOI: 10.3389/fncel.2017.00178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/12/2017] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) and membrane proteolysis play a key role in structural and functional synaptic plasticity associated with development and learning. A growing body of evidence underscores the multifaceted role of members of the metzincin superfamily, including metalloproteinases (MMPs), A Disintegrin and Metalloproteinases (ADAMs), A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTSs) and astacins in physiological and pathological processes in the central nervous system (CNS). The expression and activity of metzincins are strictly controlled at different levels (e.g., through the regulation of translation, limited activation in the extracellular space, the binding of endogenous inhibitors and interactions with other proteins). Thus, unsurprising is that the dysregulation of proteolytic activity, especially the greater expression and activation of metzincins, is associated with neurodegenerative disorders that are considered synaptopathies, especially Alzheimer's disease (AD). We review current knowledge of the functions of metzincins in the development of AD, mainly the proteolytic processing of amyloid precursor protein, the degradation of amyloid β (Aβ) peptide and several pathways for Aβ clearance across brain barriers (i.e., blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB)) that contain specific receptors that mediate the uptake of Aβ peptide. Controlling the proteolytic activity of metzincins in Aβ-induced pathological changes in AD patients' brains may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Patrycja Brzdak
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Daria Nowak
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Grzegorz Wiera
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| |
Collapse
|
14
|
Gomazkov OA. Correction of neurogenesis in the adult brain: Selection of therapeutic targets. NEUROCHEM J+ 2017. [DOI: 10.1134/s181971241604005x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
15
|
Wang S, Bolós M, Clark R, Cullen CL, Southam KA, Foa L, Dickson TC, Young KM. Amyloid β precursor protein regulates neuron survival and maturation in the adult mouse brain. Mol Cell Neurosci 2016; 77:21-33. [DOI: 10.1016/j.mcn.2016.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 08/12/2016] [Accepted: 09/19/2016] [Indexed: 01/08/2023] Open
|
16
|
β-Amyloid precursor protein-b is essential for Mauthner cell development in the zebrafish in a Notch-dependent manner. Dev Biol 2016; 413:26-38. [PMID: 26994945 DOI: 10.1016/j.ydbio.2016.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/10/2016] [Indexed: 02/05/2023]
Abstract
Amyloid precursor protein (APP) is a transmembrane glycoprotein that has been the subject of intense research because of its implication in Alzheimer's disease. However, the physiological function of APP in the development and maintenance of the central nervous system remains largely unknown. We have previously shown that the APP homologue in zebrafish (Danio rerio), Appb, is required for motor neuron patterning and formation. Here we study the function of Appb during neurogenesis in the zebrafish hindbrain. Partial knockdown of Appb using antisense morpholino oligonucleotides blocked the formation of the Mauthner neurons, uni- or bilaterally, with an aberrant behavior as a consequence of this cellular change. The Appb morphants had decreased neurogenesis, increased notch signaling and notch1a expression at the expense of deltaA/D expression. The Mauthner cell development could be restored either by a general decrease in Notch signaling through γ-secretase inhibition or by a partial knock down of Notch1a. Together, this demonstrates the importance of Appb in neurogenesis and for the first time shows the essential requirement of Appb in the formation of a specific cell type, the Mauthner cell, in the hindbrain during development. Our results suggest that Appb-regulated neurogenesis is mediated through balancing the Notch1a signaling pathway and provide new insights into the development of the Mauthner cell.
Collapse
|