51
|
Elevating the Levels of Calcium Ions Exacerbate Alzheimer's Disease via Inducing the Production and Aggregation of β-Amyloid Protein and Phosphorylated Tau. Int J Mol Sci 2021; 22:ijms22115900. [PMID: 34072743 PMCID: PMC8198078 DOI: 10.3390/ijms22115900] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/08/2021] [Accepted: 05/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with a high incidence rate. The main pathological features of AD are β-amyloid plaques (APs), which are formed by β-amyloid protein (Aβ) deposition, and neurofibrillary tangles (NFTs), which are formed by the excessive phosphorylation of the tau protein. Although a series of studies have shown that the accumulation of metal ions, including calcium ions (Ca2+), can promote the formation of APs and NFTs, there is no systematic review of the mechanisms by which Ca2+ affects the development and progression of AD. In view of this, the current review summarizes the mechanisms by which Ca2+ is transported into and out of cells and organelles, such as the cell, endoplasmic reticulum, mitochondrial and lysosomal membranes to affect the balance of intracellular Ca2+ levels. In addition, dyshomeostasis of Ca2+ plays an important role in modulating the pathogenesis of AD by influencing the production and aggregation of Aβ peptides and tau protein phosphorylation and the ways that disrupting the metabolic balance of Ca2+ can affect the learning ability and memory of people with AD. In addition, the effects of these mechanisms on the synaptic plasticity are also discussed. Finally, the molecular network through which Ca2+ regulates the pathogenesis of AD is introduced, providing a theoretical basis for improving the clinical treatment of AD.
Collapse
|
52
|
|
53
|
Ruiz-Riquelme A, Mao A, Barghash MM, Lau HHC, Stuart E, Kovacs GG, Nilsson KPR, Fraser PE, Schmitt-Ulms G, Watts JC. Aβ43 aggregates exhibit enhanced prion-like seeding activity in mice. Acta Neuropathol Commun 2021; 9:83. [PMID: 33971978 PMCID: PMC8112054 DOI: 10.1186/s40478-021-01187-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
When injected into genetically modified mice, aggregates of the amyloid-β (Aβ) peptide from the brains of Alzheimer’s disease (AD) patients or transgenic AD mouse models seed cerebral Aβ deposition in a prion-like fashion. Within the brain, Aβ exists as a pool of distinct C-terminal variants with lengths ranging from 37 to 43 amino acids, yet the relative contribution of individual C-terminal Aβ variants to the seeding behavior of Aβ aggregates remains unknown. Here, we have investigated the relative seeding activities of Aβ aggregates composed exclusively of recombinant Aβ38, Aβ40, Aβ42, or Aβ43. Cerebral Aβ42 levels were not increased in AppNL−F knock-in mice injected with Aβ38 or Aβ40 aggregates and were only increased in a subset of mice injected with Aβ42 aggregates. In contrast, significant accumulation of Aβ42 was observed in the brains of all mice inoculated with Aβ43 aggregates, and the extent of Aβ42 induction was comparable to that in mice injected with brain-derived Aβ seeds. Mice inoculated with Aβ43 aggregates exhibited a distinct pattern of cerebral Aβ pathology compared to mice injected with brain-derived Aβ aggregates, suggesting that recombinant Aβ43 may polymerize into a unique strain. Our results indicate that aggregates containing longer Aβ C-terminal variants are more potent inducers of cerebral Aβ deposition and highlight the potential role of Aβ43 seeds as a crucial factor in the initial stages of Aβ pathology in AD.
Collapse
|
54
|
Zhou K, Wang Y, Bretonnel Cohen K, Kim JD, Ma X, Shen Z, Meng X, Xia J. Bridging heterogeneous mutation data to enhance disease gene discovery. Brief Bioinform 2021; 22:6224263. [PMID: 33847357 DOI: 10.1093/bib/bbab079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/01/2021] [Accepted: 02/19/2021] [Indexed: 12/18/2022] Open
Abstract
Bridging heterogeneous mutation data fills in the gap between various data categories and propels discovery of disease-related genes. It is known that genome-wide association study (GWAS) infers significant mutation associations that link genotype and phenotype. However, due to the differences of size and quality between GWAS studies, not all de facto vital variations are able to pass the multiple testing. In the meantime, mutation events widely reported in literature unveil typical functional biological process, including mutation types like gain of function and loss of function. To bring together the heterogeneous mutation data, we propose a 'Gene-Disease Association prediction by Mutation Data Bridging (GDAMDB)' pipeline with a statistic generative model. The model learns the distribution parameters of mutation associations and mutation types and recovers false-negative GWAS mutations that fail to pass significant test but represent supportive evidences of functional biological process in literature. Eventually, we applied GDAMDB in Alzheimer's disease (AD) and predicted 79 AD-associated genes. Besides, 12 of them from the original GWAS, 60 of them are supported to be AD-related by other GWAS or literature report, and rest of them are newly predicted genes. Our model is capable of enhancing the GWAS-based gene association discovery by well combining text mining results. The positive result indicates that bridging the heterogeneous mutation data is contributory for the novel disease-related gene discovery.
Collapse
Affiliation(s)
- Kaiyin Zhou
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| | - Yuxing Wang
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| | - Kevin Bretonnel Cohen
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| | - Jin-Dong Kim
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| | - Xiaohang Ma
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| | - Zhixue Shen
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| | - Xiangyu Meng
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| | - Jingbo Xia
- Hubei Key Lab of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, Hubei Province, P.R. China
| |
Collapse
|
55
|
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
|
56
|
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
|
57
|
Slc1a3-2A-CreERT2 mice reveal unique features of Bergmann glia and augment a growing collection of Cre drivers and effectors in the 129S4 genetic background. Sci Rep 2021; 11:5412. [PMID: 33686166 PMCID: PMC7940647 DOI: 10.1038/s41598-021-84887-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
Genetic variation is a primary determinant of phenotypic diversity. In laboratory mice, genetic variation can be a serious experimental confounder, and thus minimized through inbreeding. However, generalizations of results obtained with inbred strains must be made with caution, especially when working with complex phenotypes and disease models. Here we compared behavioral characteristics of C57Bl/6—the strain most widely used in biomedical research—with those of 129S4. In contrast to 129S4, C57Bl/6 demonstrated high within-strain and intra-litter behavioral hyperactivity. Although high consistency would be advantageous, the majority of disease models and transgenic tools are in C57Bl/6. We recently established six Cre driver lines and two Cre effector lines in 129S4. To augment this collection, we genetically engineered a Cre line to study astrocytes in 129S4. It was validated with two Cre effector lines: calcium indicator gCaMP5g-tdTomato and RiboTag—a tool widely used to study cell type-specific translatomes. These reporters are in different genomic loci, and in both the Cre was functional and astrocyte-specific. We found that calcium signals lasted longer and had a higher amplitude in cortical compared to hippocampal astrocytes, genes linked to a single neurodegenerative disease have highly divergent expression patterns, and that ribosome proteins are non-uniformly expressed across brain regions and cell types.
Collapse
|
58
|
Hole KL, Williams RJ. Flavonoids as an Intervention for Alzheimer's Disease: Progress and Hurdles Towards Defining a Mechanism of Action. Brain Plast 2021; 6:167-192. [PMID: 33782649 PMCID: PMC7990465 DOI: 10.3233/bpl-200098] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Attempts to develop a disease modifying intervention for Alzheimer's disease (AD) through targeting amyloid β (Aβ) have so far been unsuccessful. There is, therefore, a need for novel therapeutics against alternative targets coupled with approaches which may be suitable for early and sustained use likely required for AD prevention. Numerous in vitro and in vivo studies have shown that flavonoids can act within processes and pathways relevant to AD, such as Aβ and tau pathology, increases in BDNF, inflammation, oxidative stress and neurogenesis. However, the therapeutic development of flavonoids has been hindered by an ongoing lack of clear mechanistic data that fully takes into consideration metabolism and bioavailability of flavonoids in vivo. With a focus on studies that incorporate these considerations into their experimental design, this review will evaluate the evidence for developing specific flavonoids as therapeutics for AD. Given the current lack of success of anti-Aβ targeting therapeutics, particular attention will be given to flavonoid-mediated regulation of tau phosphorylation and aggregation, where there is a comparable lack of study. Reflecting on this evidence, the obstacles that prevent therapeutic development of flavonoids will be examined. Finally, the significance of recent advances in flavonoid metabolomics, modifications and influence of the microbiome on the therapeutic capacity of flavonoids in AD are explored. By highlighting the potential of flavonoids to target multiple aspects of AD pathology, as well as considering the hurdles, this review aims to promote the efficient and effective identification of flavonoid-based approaches that have potential as therapeutic interventions for AD.
Collapse
Affiliation(s)
- Katriona L. Hole
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, UK
| | - Robert J. Williams
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, UK
| |
Collapse
|
59
|
Wolfe MS. Probing Mechanisms and Therapeutic Potential of γ-Secretase in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2021; 26:molecules26020388. [PMID: 33450968 PMCID: PMC7828430 DOI: 10.3390/molecules26020388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/02/2021] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
Abstract
The membrane-embedded γ-secretase complex carries out hydrolysis within the lipid bilayer in proteolyzing nearly 150 different membrane protein substrates. Among these substrates, the amyloid precursor protein (APP) has been the most studied, as generation of aggregation-prone amyloid β-protein (Aβ) is a defining feature of Alzheimer's disease (AD). Mutations in APP and in presenilin, the catalytic component of γ-secretase, cause familial AD, strong evidence for a pathogenic role of Aβ. Substrate-based chemical probes-synthetic peptides and peptidomimetics-have been critical to unraveling the complexity of γ-secretase, and small drug-like inhibitors and modulators of γ-secretase activity have been essential for exploring the potential of the protease as a therapeutic target for Alzheimer's disease. Such chemical probes and therapeutic prototypes will be reviewed here, with concluding commentary on the future directions in the study of this biologically important protease complex and the translation of basic findings into therapeutics.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, 1567 Irving Hill Road, GLH-2115, Lawrence, KS 66045, USA
| |
Collapse
|
60
|
Ayton S, Bush AI. β-amyloid: The known unknowns. Ageing Res Rev 2021; 65:101212. [PMID: 33188924 DOI: 10.1016/j.arr.2020.101212] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) stands out as a major disease without any form of preventative or disease modifying therapy. This is not for lack of trying. 33 phase 3 clinical trials of drugs targeting amyloid beta (Aβ) have failed to slow cognitive decline in AD. The field is at a cross-roads about whether to continue anti-Aβ therapy or more actively pursue alternative targets. With the burden of this disease to patients, families, and healthcare budgets growing yearly, the need for disease modifying AD therapies has become one of the highest priorities in all of medicine. While pathology, genetic and biochemical data offer a popular narrative for the causative role of Aβ, there are alternative explanations, and dissenting findings that, now more than ever, warrant thorough reanalysis. This review questions the major assumptions about Aβ on which therapies for AD were premised, and invites renewed interrogation into AD pathogenesis.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
61
|
Kapadia A, Sharma KK, Maurya IK, Singh V, Khullar M, Jain R. Structural and mechanistic insights into the inhibition of amyloid-β aggregation by Aβ 39-42 fragment derived synthetic peptides. Eur J Med Chem 2020; 212:113126. [PMID: 33395622 DOI: 10.1016/j.ejmech.2020.113126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/10/2020] [Accepted: 12/21/2020] [Indexed: 10/22/2022]
Abstract
The inhibition of amyloid-β (Aβ) aggregation is a promising approach towards therapeutic intervention for Alzheimer's disease (AD). Thirty eight tetrapeptides based upon Aβ39-42C-terminus fragment of the parent Aβ peptide were synthesized. The sequential replacement/modification employing unnatural amino acids imparted scaffold diversity, augmented activity, enhanced blood brain barrier permeability and offered proteolytic stability to the synthetic peptides. Several peptides exhibited promising protection against Aβ aggregation-mediated-neurotoxicity in PC-12 cells at doses ranged between 10 μM and 0.1 μM, further confirmed by the thioflavin-T fluorescence assay. CD study illustrate that these peptides restrict the β-sheet formation, and the non-appearance of Aβ42 fibrillar structures in the electron microscopy confirm the inhibition of Aβ42 aggregation. HRMS and ANS fluorescence spectroscopic analysis provided additional mechanistic insights. Two selected lead peptides 5 and 16 depicted enhanced blood-brain penetration and stability against serum and proteolytic enzyme. Structural insights into ligand-Aβ interactions on the monomeric and proto-fibrillar units of Aβ were computationally studied. Promising inhibitory potential and short sequence of the lead peptides offers new avenues for the advancement of peptide-derived therapeutics for AD.
Collapse
Affiliation(s)
- Akshay Kapadia
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar, Punjab, 160 062, India
| | - Krishna K Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar, Punjab, 160 062, India
| | - Indresh Kumar Maurya
- Department of Microbial Biotechnology, Punjab University, Sector 25, Chandigarh, 160 014, India
| | - Varinder Singh
- Post Graduate Institute of Medical Education and Research, Sector 11, Chandigarh, 160 014, India
| | - Madhu Khullar
- Post Graduate Institute of Medical Education and Research, Sector 11, Chandigarh, 160 014, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S Nagar, Punjab, 160 062, India.
| |
Collapse
|
62
|
Kumari R, Chaudhary A, Mani A. Casuarictin: A new herbal drug molecule for Alzheimer's disease as inhibitor of presenilin stabilization factor like protein. Heliyon 2020; 6:e05546. [PMID: 33294689 PMCID: PMC7689514 DOI: 10.1016/j.heliyon.2020.e05546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/12/2020] [Accepted: 11/16/2020] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder. In this disease neurodegeneration occurs due to deposition of aggregated amyloid-beta plaques and neurofibrillary tangles (hyperphosphorylated tau proteins). Present study focuses on interaction of different phytochemicals with presenilin stabilization factor like protein (PSFL). PSFL protein is known to stabilize Presenilin, which is mainly involved in intramembrane hydrolysis of selected type- I membrane proteins, including amyloid-beta precursor protein, and produces amyloid-beta protein. Amyloid-beta are small peptides comprising of 36–43 amino acids, which play a significant role in senile plaques formation in the brains of Alzheimer patients. Virtual screening and docking of phytochemicals with PSFL protein was done to find the potential inhibitor. Based on binding affinity, docked energy and molecular dynamics simulations, three phytochemicals namely Saponin, Casuarictin, and Enoxolone, were identified as potential inhibitors for the target protein.
Collapse
Affiliation(s)
- Rupali Kumari
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Amit Chaudhary
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Ashutosh Mani
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| |
Collapse
|
63
|
Servizi S, Corrigan RR, Casadesus G. The Importance of Understanding Amylin Signaling Mechanisms for Therapeutic Development in the Treatment of Alzheimer's Disease. Curr Pharm Des 2020; 26:1345-1355. [PMID: 32188374 PMCID: PMC10088426 DOI: 10.2174/1381612826666200318151146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
Type II Diabetes (T2D) is a major risk factor for Alzheimer's Disease (AD). These two diseases share several pathological features, including amyloid accumulation, inflammation, oxidative stress, cell death and cognitive decline. The metabolic hormone amylin and amyloid-beta are both amyloids known to self-aggregate in T2D and AD, respectively, and are thought to be the main pathogenic entities in their respective diseases. Furthermore, studies suggest amylin's ability to seed amyloid-beta aggregation, the activation of common signaling cascades in the pancreas and the brain, and the ability of amyloid beta to signal through amylin receptors (AMYR), at least in vitro. However, paradoxically, non-aggregating forms of amylin such as pramlintide are given to treat T2D and functional and neuroprotective benefits of amylin and pramlintide administration have been reported in AD transgenic mice. These paradoxical results beget a deeper study of the complex nature of amylin's signaling through the several AMYR subtypes and other receptors associated with amylin effects to be able to fully understand its potential role in mediating AD development and/or prevention. The goal of this review is to provide such critical insight to begin to elucidate how the complex nature of this hormone's signaling may explain its equally complex relationship with T2D and mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Spencer Servizi
- School of Biomedical Sciences, Kent State University, Ohio, United States
| | - Rachel R Corrigan
- School of Biomedical Sciences, Kent State University, Ohio, United States
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Ohio, United States.,Department of Biological Sciences, Kent State University, Ohio, United States
| |
Collapse
|
64
|
Fulopova B, Stuart KE, Bennett W, Bindoff A, King AE, Vickers JC, Canty AJ. Regional differences in beta amyloid plaque deposition and variable response to midlife environmental enrichment in the cortex of APP/PS1 mice. J Comp Neurol 2020; 529:1849-1862. [PMID: 33104234 DOI: 10.1002/cne.25060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/21/2023]
Abstract
Environmentally enriched housing conditions can increase performance on cognitive tasks in APP/PS1 mice; however, the potential effects of environmental enrichment (EE) on disease modification in terms of pathological change are inconclusive. We hypothesized that previous contrasting findings may be attributable to regional differences in susceptibility to amyloid beta (Aβ) plaque deposition in cortical regions that are functionally associated with EE. We characterized fibrillar plaque deposition in 6, 12, and 18-22 months old APP/PS1 mice in the prefrontal (PFC), somatosensory (SS2), and primary motor cortex (M1). We found a significant increase in plaque load between 6 and 12 months in all regions. In animals over 12 months, only the PFC region continued to significantly accumulate plaques. Additionally, 12 months old animals subjected to 6 months of EE showed improved spatial navigation and had significantly fewer plaques in M1 and SS2, but not in the PFC. These findings suggest that the PFC region is selectively susceptible to Aβ deposition and less responsive to the attenuating effects of EE. In contrast, M1 and SS2 regions plateau with respect to Aβ deposition by 12 months of age and are susceptible to amyloid pathology modification by midlife EE.
Collapse
Affiliation(s)
- Barbora Fulopova
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley E Stuart
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - William Bennett
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Alison J Canty
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
65
|
Substrate recruitment by γ-secretase. Semin Cell Dev Biol 2020; 105:54-63. [DOI: 10.1016/j.semcdb.2020.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/27/2022]
|
66
|
Neuner SM, Tcw J, Goate AM. Genetic architecture of Alzheimer's disease. Neurobiol Dis 2020; 143:104976. [PMID: 32565066 PMCID: PMC7409822 DOI: 10.1016/j.nbd.2020.104976] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/30/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
Advances in genetic and genomic technologies over the last thirty years have greatly enhanced our knowledge concerning the genetic architecture of Alzheimer's disease (AD). Several genes including APP, PSEN1, PSEN2, and APOE have been shown to exhibit large effects on disease susceptibility, with the remaining risk loci having much smaller effects on AD risk. Notably, common genetic variants impacting AD are not randomly distributed across the genome. Instead, these variants are enriched within regulatory elements active in human myeloid cells, and to a lesser extent liver cells, implicating these cell and tissue types as critical to disease etiology. Integrative approaches are emerging as highly effective for identifying the specific target genes through which AD risk variants act and will likely yield important insights related to potential therapeutic targets in the coming years. In the future, additional consideration of sex- and ethnicity-specific contributions to risk as well as the contribution of complex gene-gene and gene-environment interactions will likely be necessary to further improve our understanding of AD genetic architecture.
Collapse
Affiliation(s)
- Sarah M Neuner
- Nash Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Julia Tcw
- Nash Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Alison M Goate
- Nash Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
67
|
Lautenschläger J, Wagner-Valladolid S, Stephens AD, Fernández-Villegas A, Hockings C, Mishra A, Manton JD, Fantham MJ, Lu M, Rees EJ, Kaminski CF, Kaminski Schierle GS. Intramitochondrial proteostasis is directly coupled to α-synuclein and amyloid β1-42 pathologies. J Biol Chem 2020; 295:10138-10152. [PMID: 32385113 PMCID: PMC7383368 DOI: 10.1074/jbc.ra119.011650] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 04/30/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial dysfunction has long been implicated in the neurodegenerative disorder Parkinson's disease (PD); however, it is unclear how mitochondrial impairment and α-synuclein pathology are coupled. Using specific mitochondrial inhibitors, EM analysis, and biochemical assays, we report here that intramitochondrial protein homeostasis plays a major role in α-synuclein aggregation. We found that interference with intramitochondrial proteases, such as HtrA2 and Lon protease, and mitochondrial protein import significantly aggravates α-synuclein seeding. In contrast, direct inhibition of mitochondrial complex I, an increase in intracellular calcium concentration, or formation of reactive oxygen species, all of which have been associated with mitochondrial stress, did not affect α-synuclein pathology. We further demonstrate that similar mechanisms are involved in amyloid-β 1-42 (Aβ42) aggregation. Our results suggest that, in addition to other protein quality control pathways, such as the ubiquitin-proteasome system, mitochondria per se can influence protein homeostasis of cytosolic aggregation-prone proteins. We propose that approaches that seek to maintain mitochondrial fitness, rather than target downstream mitochondrial dysfunction, may aid in the search for therapeutic strategies to manage PD and related neuropathologies.
Collapse
Affiliation(s)
- Janin Lautenschläger
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom ;
| | - Sara Wagner-Valladolid
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Amberley D Stephens
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Ana Fernández-Villegas
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Colin Hockings
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Ajay Mishra
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - James D Manton
- Quantitative Imaging Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Marcus J Fantham
- Laser Analytics Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Meng Lu
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Eric J Rees
- Quantitative Imaging Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Clemens F Kaminski
- Laser Analytics Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom
| | - Gabriele S Kaminski Schierle
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, United Kingdom ;
| |
Collapse
|
68
|
Suire CN, Abdul-Hay SO, Sahara T, Kang D, Brizuela MK, Saftig P, Dickson DW, Rosenberry TL, Leissring MA. Cathepsin D regulates cerebral Aβ42/40 ratios via differential degradation of Aβ42 and Aβ40. ALZHEIMERS RESEARCH & THERAPY 2020; 12:80. [PMID: 32631408 PMCID: PMC7339583 DOI: 10.1186/s13195-020-00649-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/24/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cathepsin D (CatD) is a lysosomal protease that degrades both the amyloid β-protein (Aβ) and the microtubule-associated protein, tau, and has been genetically linked to late-onset Alzheimer disease (AD). Here, we sought to examine the consequences of genetic deletion of CatD on Aβ proteostasis in vivo and to more completely characterize the degradation of Aβ42 and Aβ40 by CatD. METHODS We quantified Aβ degradation rates and levels of endogenous Aβ42 and Aβ40 in the brains of CatD-null (CatD-KO), heterozygous null (CatD-HET), and wild-type (WT) control mice. CatD-KO mice die by ~ 4 weeks of age, so tissues from younger mice, as well as embryonic neuronal cultures, were investigated. Enzymological assays and surface plasmon resonance were employed to quantify the kinetic parameters (KM, kcat) of CatD-mediated degradation of monomeric human Aβ42 vs. Aβ40, and the degradation of aggregated Aβ42 species was also characterized. Competitive inhibition assays were used to interrogate the relative inhibition of full-length human and mouse Aβ42 and Aβ40, as well as corresponding p3 fragments. RESULTS Genetic deletion of CatD resulted in 3- to 4-fold increases in insoluble, endogenous cerebral Aβ42 and Aβ40, exceeding the increases produced by deletion of an insulin-degrading enzyme, neprilysin or both, together with readily detectable intralysosomal deposits of endogenous Aβ42-all by 3 weeks of age. Quite significantly, CatD-KO mice exhibited ~ 30% increases in Aβ42/40 ratios, comparable to those induced by presenilin mutations. Mechanistically, the perturbed Aβ42/40 ratios were attributable to pronounced differences in the kinetics of degradation of Aβ42 vis-à-vis Aβ40. Specifically, Aβ42 shows a low-nanomolar affinity for CatD, along with an exceptionally slow turnover rate that, together, renders Aβ42 a highly potent competitive inhibitor of CatD. Notably, the marked differences in the processing of Aβ42 vs. Aβ40 also extend to p3 fragments ending at positions 42 vs. 40. CONCLUSIONS Our findings identify CatD as the principal intracellular Aβ-degrading protease identified to date, one that regulates Aβ42/40 ratios via differential degradation of Aβ42 vs. Aβ40. The finding that Aβ42 is a potent competitive inhibitor of CatD suggests a possible mechanistic link between elevations in Aβ42 and downstream pathological sequelae in AD.
Collapse
Affiliation(s)
- Caitlin N Suire
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, USA
| | - Samer O Abdul-Hay
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Tomoko Sahara
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Dongcheul Kang
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Monica K Brizuela
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098, Kiel, Germany
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | | | - Malcolm A Leissring
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA. .,Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA.
| |
Collapse
|
69
|
Substrate-based chemical probes for Alzheimer’s γ-secretase. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02565-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
70
|
Kabir MT, Uddin MS, Setu JR, Ashraf GM, Bin-Jumah MN, Abdel-Daim MM. Exploring the Role of PSEN Mutations in the Pathogenesis of Alzheimer's Disease. Neurotox Res 2020; 38:833-849. [PMID: 32556937 DOI: 10.1007/s12640-020-00232-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Mutations of presenilin (PSEN) genes that encode presenilin proteins have been found as the vital causal factors for early-onset familial AD (FAD). AD pathological features such as memory loss, synaptic dysfunction, and formation of plaques have been successfully mimicked in the transgenic mouse models that coexpress FAD-related presenilin and amyloid precursor protein (APP) variants. γ-Secretase (GS) is an enzyme that plays roles in catalyzing intramembranous APP proteolysis to release pathogenic amyloid beta (Aβ). It has been found that presenilins can play a role as the GS's catalytic subunit. FAD-related mutations in presenilins can modify the site of GS cleavage in a way that can elevate the production of longer and highly fibrillogenic Aβ. Presenilins can interact with β-catenin to generate presenilin complexes. Aforesaid interactions have also been studied to observe the mutational and physiological activities in the catenin signal transduction pathway. Along with APP, GS can catalyze intramembrane proteolysis of various substrates that play a vital role in synaptic function. PSEN mutations can cause FAD with autosomal dominant inheritance and early onset of the disease. In this article, we have reviewed the current progress in the analysis of PSENs and the correlation of PSEN mutations and AD pathogenesis.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh. .,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | | | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11474, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
71
|
Biophysical studies of protein misfolding and aggregation in in vivo models of Alzheimer's and Parkinson's diseases. Q Rev Biophys 2020; 49:e22. [PMID: 32493529 DOI: 10.1017/s0033583520000025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodegenerative disorders, including Alzheimer's (AD) and Parkinson's diseases (PD), are characterised by the formation of aberrant assemblies of misfolded proteins. The discovery of disease-modifying drugs for these disorders is challenging, in part because we still have a limited understanding of their molecular origins. In this review, we discuss how biophysical approaches can help explain the formation of the aberrant conformational states of proteins whose neurotoxic effects underlie these diseases. We discuss in particular models based on the transgenic expression of amyloid-β (Aβ) and tau in AD, and α-synuclein in PD. Because biophysical methods have enabled an accurate quantification and a detailed understanding of the molecular mechanisms underlying protein misfolding and aggregation in vitro, we expect that the further development of these methods to probe directly the corresponding mechanisms in vivo will open effective routes for diagnostic and therapeutic interventions.
Collapse
|
72
|
Funamoto S, Tagami S, Okochi M, Morishima-Kawashima M. Successive cleavage of β-amyloid precursor protein by γ-secretase. Semin Cell Dev Biol 2020; 105:64-74. [PMID: 32354467 DOI: 10.1016/j.semcdb.2020.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
γ-Secretase is a multimeric aspartyl protease that cleaves the membrane-spanning region of the β-carboxyl terminal fragment (βCTF) generated from β-amyloid precursor protein. γ-Secretase defines the generated molecular species of amyloid β-protein (Aβ), a critical molecule in the pathogenesis of Alzheimer's disease (AD). Many therapeutic trials for AD have targeted γ-secretase. However, in contrast to the great efforts in drug discovery, the enzymatic features and cleavage mechanism of γ-secretase are poorly understood. Here we review our protein-chemical analyses of the cleavage products generated from βCTF by γ-secretase, which revealed that Aβ was produced by γ-secretase through successive cleavages of βCTF, mainly at three-residue intervals. Two representative product lines were identified. ε-Cleavages occur first at Leu49-Val50 and Thr48-Leu49 of βCTF (in accordance with Aβ numbering). Longer generated Aβs, Aβ49 and Aβ48, are precursors to the majority of Aβ40 and Aβ42, concomitantly releasing the tripeptides, ITL, VIV, and IAT; and VIT and TVI, respectively. A portion of Aβ42 is processed further to Aβ38, releasing a tetrapeptide, VVIA. The presence of additional multiple minor pathways may reflect labile cleavage activities derived from the conformational flexibility of γ-secretase through molecular interactions. Because these peptide byproducts are not secreted and remain within the cells, they may serve as an indicator that reflects γ-secretase activity more directly than secreted Aβ.
Collapse
Affiliation(s)
- Satoru Funamoto
- Department of Neuropathology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Shinji Tagami
- Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masayasu Okochi
- Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Maho Morishima-Kawashima
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
73
|
Vyas Y, Montgomery JM, Cheyne JE. Hippocampal Deficits in Amyloid-β-Related Rodent Models of Alzheimer's Disease. Front Neurosci 2020; 14:266. [PMID: 32317913 PMCID: PMC7154147 DOI: 10.3389/fnins.2020.00266] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/09/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is the most common cause of dementia. Symptoms of AD include memory loss, disorientation, mood and behavior changes, confusion, unfounded suspicions, and eventually, difficulty speaking, swallowing, and walking. These symptoms are caused by neuronal degeneration and cell loss that begins in the hippocampus, and later in disease progression spreading to the rest of the brain. While there are some medications that alleviate initial symptoms, there are currently no treatments that stop disease progression. Hippocampal deficits in amyloid-β-related rodent models of AD have revealed synaptic, behavioral and circuit-level defects. These changes in synaptic function, plasticity, neuronal excitability, brain connectivity, and excitation/inhibition imbalance all have profound effects on circuit function, which in turn could exacerbate disease progression. Despite, the wealth of studies on AD pathology we don't yet have a complete understanding of hippocampal deficits in AD. With the increasing development of in vivo recording techniques in awake and freely moving animals, future studies will extend our current knowledge of the mechanisms underpinning how hippocampal function is altered in AD, and aid in progression of treatment strategies that prevent and/or delay AD symptoms.
Collapse
Affiliation(s)
| | - Johanna M. Montgomery
- Department of Physiology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Juliette E. Cheyne
- Department of Physiology, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
74
|
Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21041327. [PMID: 32079110 PMCID: PMC7072986 DOI: 10.3390/ijms21041327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by extracellular amyloid plaques composed of the β-amyloid peptides and intracellular neurofibrillary tangles and associates with progressive declines in memory and cognition. Several genes play important roles and regulate enzymes that produce a pathological accumulation of β-amyloid in the brain, such as gamma secretase (γ-secretase). Induced pluripotent stem cells from patients with Alzheimer’s disease with different underlying genetic mechanisms may help model different phenotypes of Alzheimer’s disease and facilitate personalized drug screening platforms for the identification of small molecules. We also discuss recent developments by γ-secretase inhibitors and modulators in the treatment of AD. In addition, small-molecule drugs isolated from Chinese herbal medicines have been shown effective in treating Alzheimer’s disease. We propose a mechanism of small-molecule drugs in treating Alzheimer’s disease. Combining therapy with different small-molecule drugs may increase the chance of symptomatic treatment. A customized strategy tailored to individuals and in combination with therapy may be a more suitable treatment option for Alzheimer’s disease in the future.
Collapse
|
75
|
Abstract
With the imminent needs of rapid, accurate, simple point-of-care systems for global healthcare industry, electrochemical biosensors have been widely developed owing to their cost-effectiveness and simple instrumentation. However, typical electrochemical biosensors for direct analysis of proteins in the human biological sample still suffer from complex biosensor fabrication, lack of general method, limited sensitivity, and matrix-caused biofouling effect. To resolve these challenges, we developed a general electrochemical sensing strategy based on a designed steric hindrance effect on an antibody surface layer. This strategy utilizes the interaction pattern of protein-G and immunoglobulin G (Fc and Fab regions), providing a steric hindrance effect during the target capturing process. The provided steric hindrance effect minimizes the matrix effect-caused fouling surface and altered the path of electron transfer, delivering a low-fouling and high-sensitivity detection of protein in complex matrices. Also, an enzyme-based horseradish peroxidase/hydroquinone/H2O2 transduction system can also be applied to the system, demonstrating the versatility of this sensing strategy for general electrochemical sensing applications. We demonstrated this platform through the detection of Tau protein and programming death ligand 1 with a subpico molar detection limit within 10 min, satisfying the clinical point-of-care requirements for rapid turnaround time and ultrasensitivity.
Collapse
|
76
|
Galla L, Redolfi N, Pozzan T, Pizzo P, Greotti E. Intracellular Calcium Dysregulation by the Alzheimer's Disease-Linked Protein Presenilin 2. Int J Mol Sci 2020; 21:E770. [PMID: 31991578 PMCID: PMC7037278 DOI: 10.3390/ijms21030770] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Even though most AD cases are sporadic, a small percentage is familial due to autosomal dominant mutations in amyloid precursor protein (APP), presenilin-1 (PSEN1), and presenilin-2 (PSEN2) genes. AD mutations contribute to the generation of toxic amyloid β (Aβ) peptides and the formation of cerebral plaques, leading to the formulation of the amyloid cascade hypothesis for AD pathogenesis. Many drugs have been developed to inhibit this pathway but all these approaches currently failed, raising the need to find additional pathogenic mechanisms. Alterations in cellular calcium (Ca2+) signaling have also been reported as causative of neurodegeneration. Interestingly, Aβ peptides, mutated presenilin-1 (PS1), and presenilin-2 (PS2) variously lead to modifications in Ca2+ homeostasis. In this contribution, we focus on PS2, summarizing how AD-linked PS2 mutants alter multiple Ca2+ pathways and the functional consequences of this Ca2+ dysregulation in AD pathogenesis.
Collapse
Affiliation(s)
- Luisa Galla
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Nelly Redolfi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), 35131 Padua, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (L.G.); (N.R.); (T.P.); (E.G.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| |
Collapse
|
77
|
Wolfe MS. Unraveling the complexity of γ-secretase. Semin Cell Dev Biol 2020; 105:3-11. [PMID: 31980377 PMCID: PMC7371508 DOI: 10.1016/j.semcdb.2020.01.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/26/2019] [Accepted: 01/15/2020] [Indexed: 01/07/2023]
Abstract
γ-Secretase was initially defined as a proteolytic activity that cleaves within the transmembrane of the amyloid precursor protein (APP) to produce the amyloid β-peptide of Alzheimer's disease. The discovery of mutations in APP and the presenilins associated with familial Alzheimer's disease and their effects on APP processing dovetailed with pharmacological studies on γ-secretase, leading to the revelation that presenilins are unprecedented membrane-embedded aspartyl proteases. Other members of what became known as the γ-secretase complex were subsequently identified. In parallel with these advances, connections between presenilins and Notch receptors essential to metazoan development became evident, resulting in the concurrent realization that γ-secretase also carries out intramembrane proteolysis of Notch as part of its signaling mechanism. Substantial progress has been made toward elucidating how γ-secretase carries out complex processing of transmembrane domains, how it goes awry in familial Alzheimer's disease, the scope of its substrates, and the atomic details of its structure. Critical questions remain for future study, toward further unraveling the complexity of this unique membrane-embedded proteolytic machine and its roles in biology and disease.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
78
|
Trambauer J, Rodríguez Sarmiento RM, Fukumori A, Feederle R, Baumann K, Steiner H. Aβ43-producing PS1 FAD mutants cause altered substrate interactions and respond to γ-secretase modulation. EMBO Rep 2020; 21:e47996. [PMID: 31762188 PMCID: PMC6945062 DOI: 10.15252/embr.201947996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
Abnormal generation of neurotoxic amyloid-β peptide (Aβ) 42/43 species due to mutations in the catalytic presenilin 1 (PS1) subunit of γ-secretase is the major cause of familial Alzheimer's disease (FAD). Deeper mechanistic insight on the generation of Aβ43 is still lacking, and it is unclear whether γ-secretase modulators (GSMs) can reduce the levels of this Aβ species. By comparing several types of Aβ43-generating FAD mutants, we observe that very high levels of Aβ43 are often produced when presenilin function is severely impaired. Altered interactions of C99, the precursor of Aβ, are found for all mutants and are independent of their particular effect on Aβ production. Furthermore, unlike previously described GSMs, the novel compound RO7019009 can effectively lower Aβ43 production of all mutants. Finally, substrate-binding competition experiments suggest that RO7019009 acts mechanistically after initial C99 binding. We conclude that altered C99 interactions are a common feature of diverse types of PS1 FAD mutants and that also patients with Aβ43-generating FAD mutations could in principle be treated by GSMs.
Collapse
Affiliation(s)
- Johannes Trambauer
- Biomedical Center (BMC), Metabolic BiochemistryLudwig‐Maximilians‐UniversityMunichGermany
| | | | - Akio Fukumori
- Department of Aging NeurobiologyNational Center for Geriatrics and GerontologyObuJapan
- Department of Mental Health PromotionOsaka University Graduate School of MedicineToyonakaJapan
| | - Regina Feederle
- Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Center MunichGerman Research Center for Environmental HealthNeuherbergGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Karlheinz Baumann
- Roche Pharma Research and Early DevelopmentRoche Innovation Center Basel, F. Hoffmann‐La Roche Ltd.BaselSwitzerland
| | - Harald Steiner
- Biomedical Center (BMC), Metabolic BiochemistryLudwig‐Maximilians‐UniversityMunichGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| |
Collapse
|
79
|
Park J, Lai MKP, Arumugam TV, Jo DG. O-GlcNAcylation as a Therapeutic Target for Alzheimer's Disease. Neuromolecular Med 2020; 22:171-193. [PMID: 31894464 DOI: 10.1007/s12017-019-08584-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and the number of elderly patients suffering from AD has been steadily increasing. Despite worldwide efforts to cope with this disease, little progress has been achieved with regard to identification of effective therapeutics. Thus, active research focusing on identification of new therapeutic targets of AD is ongoing. Among the new targets, post-translational modifications which modify the properties of mature proteins have gained attention. O-GlcNAcylation, a type of PTM that attaches O-linked β-N-acetylglucosamine (O-GlcNAc) to a protein, is being sought as a new target to treat AD pathologies. O-GlcNAcylation has been known to modify the two important components of AD pathological hallmarks, amyloid precursor protein, and tau protein. In addition, elevating O-GlcNAcylation levels in AD animal models has been shown to be effective in alleviating AD-associated pathology. Although studies investigating the precise mechanism of reversal of AD pathologies by targeting O-GlcNAcylation are not yet complete, it is clearly important to examine O-GlcNAcylation regulation as a target of AD therapeutics. This review highlights the mechanisms of O-GlcNAcylation and its role as a potential therapeutic target under physiological and pathological AD conditions.
Collapse
Affiliation(s)
- Jinsu Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
- Department of Health Science and Technology, Sungkyunkwan University, Seoul, 06351, Korea
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea.
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore, 117593, Singapore.
- Department of Physiology, Anatomy & Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia.
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea.
- Department of Health Science and Technology, Sungkyunkwan University, Seoul, 06351, Korea.
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
80
|
Wang C, Holtzman DM. Bidirectional relationship between sleep and Alzheimer's disease: role of amyloid, tau, and other factors. Neuropsychopharmacology 2020; 45:104-120. [PMID: 31408876 PMCID: PMC6879647 DOI: 10.1038/s41386-019-0478-5] [Citation(s) in RCA: 338] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/28/2019] [Accepted: 08/02/2019] [Indexed: 01/04/2023]
Abstract
As we age, we experience changes in our nighttime sleep and daytime wakefulness. Individuals afflicted with Alzheimer's disease (AD) can develop sleep problems even before memory and other cognitive deficits are reported. As the disease progresses and cognitive changes ensue, sleep disturbances become even more debilitating. Thus, it is imperative to gain a better understanding of the relationship between sleep and AD pathogenesis. We postulate a bidirectional relationship between sleep and the neuropathological hallmarks of AD; in particular, the accumulation of amyloid-β (Aβ) and tau. Our research group has shown that extracellular levels of both Aβ and tau fluctuate during the normal sleep-wake cycle. Disturbed sleep and increased wakefulness acutely lead to increased Aβ production and decreased Aβ clearance, whereas Aβ aggregation and deposition is enhanced by chronic increased wakefulness in animal models. Once Aβ accumulates, there is evidence in both mice and humans that this results in disturbed sleep. New findings from our group reveal that acute sleep deprivation increases levels of tau in mouse brain interstitial fluid (ISF) and human cerebrospinal fluid (CSF) and chronic sleep deprivation accelerates the spread of tau protein aggregates in neural networks. Finally, recent evidence also suggests that accumulation of tau aggregates in the brain correlates with decreased nonrapid eye movement (NREM) sleep slow wave activity. In this review, we first provide a brief overview of the AD and sleep literature and then highlight recent advances in the understanding of the relationship between sleep and AD pathogenesis. Importantly, the effects of the bidirectional relationship between the sleep-wake cycle and tau have not been previously discussed in other reviews on this topic. Lastly, we provide possible directions for future studies on the role of sleep in AD.
Collapse
Affiliation(s)
- Chanung Wang
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
81
|
Tiscione SA, Vivas O, Ginsburg KS, Bers DM, Ory DS, Santana LF, Dixon RE, Dickson EJ. Disease-associated mutations in Niemann-Pick type C1 alter ER calcium signaling and neuronal plasticity. J Cell Biol 2019; 218:4141-4156. [PMID: 31601621 PMCID: PMC6891088 DOI: 10.1083/jcb.201903018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 08/12/2019] [Accepted: 09/06/2019] [Indexed: 11/22/2022] Open
Abstract
Niemann-Pick type C1 (NPC1) protein is essential for the transport of externally derived cholesterol from lysosomes to other organelles. Deficiency of NPC1 underlies the progressive NPC1 neurodegenerative disorder. Currently, there are no curative therapies for this fatal disease. Given the Ca2+ hypothesis of neurodegeneration, which posits that altered Ca2+ dynamics contribute to neuropathology, we tested if disease mutations in NPC1 alter Ca2+ signaling and neuronal plasticity. We determine that NPC1 inhibition or disease mutations potentiate store-operated Ca2+ entry (SOCE) due to a presenilin 1 (PSEN1)-dependent reduction in ER Ca2+ levels alongside elevated expression of the molecular SOCE components ORAI1 and STIM1. Associated with this dysfunctional Ca2+ signaling is destabilization of neuronal dendritic spines. Knockdown of PSEN1 or inhibition of the SREBP pathway restores Ca2+ homeostasis, corrects differential protein expression, reduces cholesterol accumulation, and rescues spine density. These findings highlight lysosomes as a crucial signaling platform responsible for tuning ER Ca2+ signaling, SOCE, and synaptic architecture in health and disease.
Collapse
Affiliation(s)
- Scott A Tiscione
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Oscar Vivas
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | | | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA
| | - Daniel S Ory
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Luis F Santana
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Rose E Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| |
Collapse
|
82
|
Vöglein J, Willem M, Trambauer J, Schönecker S, Dieterich M, Biskup S, Giudici C, Utz K, Oberstein T, Brendel M, Rominger A, Danek A, Steiner H, Haass C, Levin J. Identification of a rare presenilin 1 single amino acid deletion mutation (F175del) with unusual amyloid-β processing effects. Neurobiol Aging 2019; 84:241.e5-241.e11. [DOI: 10.1016/j.neurobiolaging.2019.08.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/22/2019] [Accepted: 08/31/2019] [Indexed: 10/26/2022]
|
83
|
Chan HH, Koh RY, Lim CL, Leong CO. Receptor-Interacting Protein Kinase 1 (RIPK1) as a Potential Therapeutic Target: An Overview of Its Possible Role in the Pathogenesis of Alzheimer's Disease. Curr Alzheimer Res 2019; 16:907-918. [PMID: 31642777 DOI: 10.2174/1567205016666191023102422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/02/2019] [Accepted: 09/06/2019] [Indexed: 01/03/2023]
Abstract
Alzheimer's Disease (AD) is an age-dependent neurodegenerative disorder, the most common type of dementia that is clinically characterized by the presence of beta-amyloid (Aβ) extracellularly and intraneuronal tau protein tangles that eventually leads to the onset of memory and cognition impairment, development of psychiatric symptoms and behavioral disorders that affect basic daily activities. Current treatment approved by the U.S Food and Drug Administration (FDA) for AD is mainly focused on the symptoms but not on the pathogenesis of the disease. Recently, receptor-interacting protein kinase 1 (RIPK1) has been identified as a key component in the pathogenesis of AD through necroptosis. Furthermore, genetic and pharmacological suppression of RIPK1 has been shown to revert the phenotype of AD and its mediating pathway is yet to be deciphered. This review is aimed to provide an overview of the pathogenesis and current treatment of AD with the involvement of autophagy as well as providing a novel insight into RIPK1 in reverting the progression of AD, probably through an autophagy machinery.
Collapse
Affiliation(s)
- Hong Hao Chan
- School of Postgraduate Studies and Research, International Medical University, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Chooi Ling Lim
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Chee Onn Leong
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
84
|
Sushma, Mondal AC. Role of GPCR signaling and calcium dysregulation in Alzheimer's disease. Mol Cell Neurosci 2019; 101:103414. [PMID: 31655116 DOI: 10.1016/j.mcn.2019.103414] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), a late onset neurodegenerative disorder is characterized by the loss of memory, disordered cognitive function, caused by accumulation of amyloid-β (Aβ) peptide and neurofibrillary tangles (NFTs) in the neocortex and hippocampal brain area. Extensive research has been done on the findings of the disease etiology or pathological causes of aggregation of Aβ and hyperphosphorylation of tau protein without much promising results. Recently, calcium dysregulation has been reported to play an important role in the pathophysiology of AD. Calcium ion acts as one of the major secondary messengers, regulates many signaling pathways involved in cell survival, proliferation, differentiation, transcription and apoptosis. Calcium signaling is one of the major signaling pathways involved in the formation of memory, generation of energy and other physiological functions. It also can modulate function of many proteins upon binding. Dysregulation in calcium homeostasis leads to many physiological changes leading to neurodegenerative diseases including AD. In AD, GPCRs generate secondary messengers which regulate calcium homeostasis inside the cell and is reported to be disturbed in the pathological condition. Calcium channels and receptors present on the plasma membrane and intracellular organelle maintain calcium homeostasis through different signaling mechanisms. In this review, we have summarized the different calcium channels and receptors involved in calcium dysregulation which in turn play a critical role in the pathogenesis of AD. Understanding the role of calcium channels and GPCRs to maintain calcium homeostasis is an attempt to develop effective AD treatments.
Collapse
Affiliation(s)
- Sushma
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India.
| |
Collapse
|
85
|
Lautenschäger J, Kaminski Schierle GS. Mitochondrial degradation of amyloidogenic proteins - A new perspective for neurodegenerative diseases. Prog Neurobiol 2019; 181:101660. [PMID: 31301323 DOI: 10.1016/j.pneurobio.2019.101660] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 06/02/2019] [Accepted: 07/04/2019] [Indexed: 01/09/2023]
Abstract
This perspective article outlines mechanisms of mitochondrial import and protein degradation and how these have been linked to alpha-synuclein and Amyloid beta (Aβ) homeostasis. Our aim is to underpin and stimulate the debate on the recent conception of mitochondria as protein degrading organelles, which suggests that mitochondria are more directly involved in neurodegenerative diseases than previously assumed.
Collapse
Affiliation(s)
- Janin Lautenschäger
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK.
| | - Gabriele S Kaminski Schierle
- Molecular Neuroscience Group, Department of Chemical Engineering and Biotechnology, University of Cambridge, West Cambridge Site, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| |
Collapse
|
86
|
Haass C, Levin J. [Did Alzheimer research fail entirely? : Failure of amyloid-based clinical studies]. DER NERVENARZT 2019; 90:884-890. [PMID: 31243509 DOI: 10.1007/s00115-019-0751-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Numerous amyloid-based clinical studies have recently failed. Does this mean that the mechanisms of Alzheimer's disease have to be reinvestigated and that amyloid is not the trigger of the disease? Strong genetic evidence from familial Alzheimer's disease contradicts this fatalistic opinion. Mutations in all genes associated with familial Alzheimer's disease affect amyloid metabolism and aggregation. Moreover, a protective mutation reduces amyloid production by 20-30% throughout the lifetime. Clinical studies rather failed because secretase inhibitors block cleavage of numerous other physiologically important substrates of secretases. Moreover, the disease is initiated decades before symptoms occur. Successful treatment attempts with anti-amyloid medication based on other prototype amyloidoses are described. Finally, new therapeutic target molecules expressed in microglia cells are discussed.
Collapse
Affiliation(s)
- Christian Haass
- Lehrstuhl für Stoffwechselbiochemie, Biomedizinisches Centrum (BMC), Fakultät für Medizin, Ludwig-Maximilians-Universität München, Feodor-Lynen Straße 17, 81377, München, Deutschland. .,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) München, München, Deutschland. .,Münchner Cluster für Systemneurologie (SyNergy), München, Deutschland.
| | - Johannes Levin
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) München, München, Deutschland.,Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Deutschland
| |
Collapse
|
87
|
Mengel D, Liu L, Yamamoto R, Zülow S, Deuschl C, Hermann DM, Zerr I, Selkoe DJ, Dodel R. A novel V272D presenilin mutation associated with logopenia, disorientation, and apraxia in an autosomal-dominant Alzheimer's disease family. Neurobiol Aging 2019; 85:154.e5-154.e7. [PMID: 31500908 DOI: 10.1016/j.neurobiolaging.2019.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/26/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022]
Abstract
In the present study, a novel mutation in the presenilin 1 gene was discovered in an Iraq-native patient with early-onset Alzheimer's disease, who presented with speech impairment and memory decline at age 46 years. Magnetic resonance imaging showed a frontotemporal atrophy. Sanger sequencing identified a heterozygous T to A transversion at position 815 (c.815T>A) in the presenilin 1 gene (PSEN1), resulting in a novel missense mutation at codon 272 from valine to aspartate (V272D). We tested this PSEN1 mutation in vitro and found V272D resulted in an altered Aβ42/40 ratio.
Collapse
Affiliation(s)
- David Mengel
- Chair of Geriatric Medicine, University Essen, and Geriatric Centre Haus Berge, Contilia Group, Essen, Germany; Department of Neurology, Philipps-University Marburg, Marburg, Germany; Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lei Liu
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Raina Yamamoto
- Medizinisches Versorgungszentrum Dr. Eberhard & Partner, Dortmund, Germany
| | - Stefan Zülow
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Cornelius Deuschl
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Inga Zerr
- Department of Neurology, University of Göttingen, Göttingen, Germany
| | - Dennis J Selkoe
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Richard Dodel
- Chair of Geriatric Medicine, University Essen, and Geriatric Centre Haus Berge, Contilia Group, Essen, Germany; Department of Neurology, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
88
|
Fišar Z, Hansíková H, Křížová J, Jirák R, Kitzlerová E, Zvěřová M, Hroudová J, Wenchich L, Zeman J, Raboch J. Activities of mitochondrial respiratory chain complexes in platelets of patients with Alzheimer's disease and depressive disorder. Mitochondrion 2019; 48:67-77. [PMID: 31377247 DOI: 10.1016/j.mito.2019.07.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/03/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022]
Abstract
We analyzed activities of complex I, II, III, and IV, and citrate synthase (CS) in patients with major depressive disorder (MDD) or Alzheimer's disease (AD) presenting with or without depression. Associations of these parameters with disease or disease severity were observed in both AD and MDD; however, mean values of mitochondrial parameters were significantly altered in AD but not in MDD. Potential mitochondrial dysfunction in MDD seems not to be caused by disturbed activity of CS or respiratory complexes. In AD, a decrease in the activity of CS and complex IV may cause mitochondrial dysfunction, whereas an increase in activities of other mitochondrial complexes or their ratios to CS may be an adaptive response. The data indicate that comorbid depression in AD is associated with increased complex II activity. The mitochondrial parameters measured can be included in the panel of biomarkers of AD.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Hana Hansíková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Jana Křížová
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Eva Kitzlerová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Martina Zvěřová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic; Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, Praha 2 128 00, Prague 2, Czech Republic.
| | - László Wenchich
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Jiří Zeman
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| |
Collapse
|
89
|
Gandhi J, Antonelli AC, Afridi A, Vatsia S, Joshi G, Romanov V, Murray IVJ, Khan SA. Protein misfolding and aggregation in neurodegenerative diseases: a review of pathogeneses, novel detection strategies, and potential therapeutics. Rev Neurosci 2019; 30:339-358. [PMID: 30742586 DOI: 10.1515/revneuro-2016-0035] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022]
Abstract
Protein folding is a complex, multisystem process characterized by heavy molecular and cellular footprints. Chaperone machinery enables proper protein folding and stable conformation. Other pathways concomitant with the protein folding process include transcription, translation, post-translational modifications, degradation through the ubiquitin-proteasome system, and autophagy. As such, the folding process can go awry in several different ways. The pathogenic basis behind most neurodegenerative diseases is that the disruption of protein homeostasis (i.e. proteostasis) at any level will eventually lead to protein misfolding. Misfolded proteins often aggregate and accumulate to trigger neurotoxicity through cellular stress pathways and consequently cause neurodegenerative diseases. The manifestation of a disease is usually dependent on the specific brain region that the neurotoxicity affects. Neurodegenerative diseases are age-associated, and their incidence is expected to rise as humans continue to live longer and pursue a greater life expectancy. We presently review the sequelae of protein misfolding and aggregation, as well as the role of these phenomena in several neurodegenerative diseases including Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, Parkinson's disease, transmissible spongiform encephalopathies, and spinocerebellar ataxia. Strategies for treatment and therapy are also conferred with respect to impairing, inhibiting, or reversing protein misfolding.
Collapse
Affiliation(s)
- Jason Gandhi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA.,Medical Student Research Institute, St. George's University School of Medicine, Grenada, West Indies
| | - Anthony C Antonelli
- Department of Pathology, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA
| | - Adil Afridi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA
| | - Sohrab Vatsia
- Department of Cardiothoracic Surgery, Lenox Hill Hospital, 130 East 77th Street, New York, NY 10075, USA
| | - Gunjan Joshi
- Department of Internal Medicine, Stony Brook Southampton Hospital, 240 Meeting House Lane, Southampton, NY 11968, USA
| | - Victor Romanov
- Department of Urology, Health Sciences Center T9-040, Stony Brook University School of Medicine, 101 Nicolls Road, Stony Brook, NY 11794-8093, USA
| | - Ian V J Murray
- Department of Physiology and Neuroscience, St. George's University School of Medicine, Grenada, West Indies
| | - Sardar Ali Khan
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, 101 Nicolls Road, Health Sciences Center, Stony Brook, NY 11794-8434, USA.,Department of Urology, Health Sciences Center T9-040, Stony Brook University School of Medicine, 101 Nicolls Road, Stony Brook, NY 11794-8093, USA
| |
Collapse
|
90
|
Substrate recognition and processing by γ-secretase. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1862:183016. [PMID: 31295475 PMCID: PMC6899174 DOI: 10.1016/j.bbamem.2019.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/13/2019] [Accepted: 07/02/2019] [Indexed: 12/22/2022]
Abstract
The γ-secretase complex is composed of four membrane protein subunits, including presenilin as the catalytic component with aspartyl protease activity. The enzyme cleaves within the transmembrane domain of >70 different type I integral membrane proteins and has been dubbed "the proteasome of the membrane". The most studied substrates include the Notch family of receptors, involved in cell differentiation, and the amyloid precursor protein (APP), involved in the pathogenesis of Alzheimer's disease. A central mechanistic question is how γ-secretase recognizes helical transmembrane substrates and carries out processive proteolysis. Recent findings addressing substrate recognition and processing will be discussed, including the role of protease subunit nicastrin as a gatekeeper, the effects of Alzheimer-causing mutations in presenilin on processive proteolysis of APP, and evidence that three pockets in the active site (S1', S2', and S3') determine carboxypeptidase cleavage of substrate in intervals of three residues. This article is part of a Special Issue entitled: Molecular biophysics of membranes and membrane proteins.
Collapse
|
91
|
Abstract
γ-Secretase is a membrane-embedded protease complex, with presenilin as the catalytic component containing two transmembrane aspartates in the active site. With more than 90 known substrates, the γ-secretase complex is considered "the proteasome of the membrane", with central roles in biology and medicine. The protease carries out hydrolysis within the lipid bilayer to cleave the transmembrane domain of the substrate multiple times before releasing secreted products. For many years, elucidation of γ-secretase structure and function largely relied on small-molecule probes and mutagenesis. Recently, however, advances in cryo-electron microscopy have led to the first detailed structures of the protease complex. Two new reports of structures of γ-secretase bound to membrane protein substrates provide great insight into the nature of substrate recognition and how Alzheimer's disease-causing mutations in presenilin might alter substrate binding and processing. These new structures offer a powerful platform for elucidating enzyme mechanisms, deciphering effects of disease-causing mutations, and advancing Alzheimer's disease drug discovery.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry , University of Kansas , Lawrence , Kansas 66045 , United States
| |
Collapse
|
92
|
Liu S, Ando F, Fujita Y, Liu J, Maeda T, Shen X, Kikuchi K, Matsumoto A, Yokomori M, Tanabe-Fujimura C, Shimokata H, Michikawa M, Komano H, Zou K. A clinical dose of angiotensin-converting enzyme (ACE) inhibitor and heterozygous ACE deletion exacerbate Alzheimer's disease pathology in mice. J Biol Chem 2019; 294:9760-9770. [PMID: 31072831 DOI: 10.1074/jbc.ra118.006420] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 05/07/2019] [Indexed: 12/23/2022] Open
Abstract
Inhibition of angiotensin-converting enzyme (ACE) is a strategy used worldwide for managing hypertension. In addition to converting angiotensin I to angiotensin II, ACE also converts neurotoxic β-amyloid protein 42 (Aβ42) to Aβ40. Because of its neurotoxicity, Aβ42 is believed to play a causative role in the development of Alzheimer's disease (AD), whereas Aβ40 has neuroprotective effects against Aβ42 aggregation and also against metal-induced oxidative damage. Whether ACE inhibition enhances Aβ42 aggregation or impairs human cognitive ability are very important issues for preventing AD onset and for optimal hypertension management. In an 8-year longitudinal study, we found here that the mean intelligence quotient of male, but not female, hypertensive patients taking ACE inhibitors declined more rapidly than that of others taking no ACE inhibitors. Moreover, the sera of all AD patients exhibited a decrease in Aβ42-to-Aβ40-converting activity compared with sera from age-matched healthy individuals. Using human amyloid precursor protein transgenic mice, we found that a clinical dose of an ACE inhibitor was sufficient to increase brain amyloid deposition. We also generated human amyloid precursor protein/ACE+/- mice and found that a decrease in ACE levels promoted Aβ42 deposition and increased the number of apoptotic neurons. These results suggest that inhibition of ACE activity is a risk factor for impaired human cognition and for triggering AD onset.
Collapse
Affiliation(s)
- Shuyu Liu
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan.,the Liaoning Provincial Key Laboratory of Behavior and Cognitive Neuroscience, Shenyang Medical College, Shenyang 110034, China
| | - Fujiko Ando
- the Faculty of Health and Medical Sciences, Aichi Shukutoku University, Nagakute 480-1146, Japan
| | - Yu Fujita
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Junjun Liu
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Tomoji Maeda
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Xuefeng Shen
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Kota Kikuchi
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Aoi Matsumoto
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Mirai Yokomori
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Chiaki Tanabe-Fujimura
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan
| | - Hiroshi Shimokata
- the Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin 470-0196, Japan, and
| | - Makoto Michikawa
- the Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Hiroto Komano
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan,
| | - Kun Zou
- From the Department of Neuroscience, School of Pharmacy, Iwate Medical University, Yahaba 028-3694, Japan, .,the Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| |
Collapse
|
93
|
Fišar Z, Jirák R, Zvěřová M, Setnička V, Habartová L, Hroudová J, Vaníčková Z, Raboch J. Plasma amyloid beta levels and platelet mitochondrial respiration in patients with Alzheimer's disease. Clin Biochem 2019; 72:71-80. [PMID: 30954436 DOI: 10.1016/j.clinbiochem.2019.04.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Altered amyloid metabolism and mitochondrial dysfunction play key roles in the development of Alzheimer's disease (AD). We asked whether an association exists between disturbed platelet mitochondrial respiration and the plasma concentrations of Aβ40 and Aβ42 in patients with AD. DESIGN AND METHODS Plasma Aβ40 and Aβ42 concentrations and mitochondrial respiration in intact and permeabilized platelets were measured in 50 patients with AD, 15 patients with vascular dementia and 25 control subjects. A pilot longitudinal study was performed to monitor the progression of AD in a subgroup 11 patients with AD. RESULTS The mean Aβ40, Aβ42 and Aβ42/Aβ40 levels were not significantly altered in patients with AD compared with controls. The mitochondrial respiratory rate in intact platelets was significantly reduced in patients with AD compared to controls, particularly the basal respiratory rate, maximum respiratory capacity, and respiratory reserve; however, the flux control ratio for basal respiration was increased. A correlation between the plasma Aβ42 concentration and mitochondrial respiration in both intact and permeabilized platelets differs in controls and patients with AD. CONCLUSIONS Based on our data, (1) mitochondrial respiration in intact platelets, but not the Aβ level itself, may be included in a panel of biomarkers for AD; (2) dysfunctional mitochondrial respiration in platelets is not explained by changes in plasma Aβ concentrations; and (3) the association between mitochondrial respiration in platelets and plasma Aβ levels differs in patients with AD and controls. The results supported the hypothesis that mitochondrial dysfunction is the primary factor contributing to the development of AD.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Martina Zvěřová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Vladimír Setnička
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czech Republic.
| | - Lucie Habartová
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czech Republic.
| | - Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Zdislava Vaníčková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| |
Collapse
|
94
|
Overk C, Masliah E. Dale Schenk One Year Anniversary: Fighting to Preserve the Memories. J Alzheimers Dis 2019; 62:1-13. [PMID: 29439357 DOI: 10.3233/jad-171071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It has been a year since we lost Dale Schenk on September 30, 2016. Dale's visionary work resulted in the remarkable discovery in 1999 that an experimental amyloid-β (Aβ) vaccine reduced the neurodegeneration in a transgenic model of Alzheimer's disease (AD). Following Dale's seminal work, several active and passive immunotherapies have since been developed and tested in the clinic for AD, Parkinson's disease (PD), and other neurodegenerative disorders. Here we provide a brief overview of the current state of development of immunotherapy for AD, PD, and other neurodegenerative disorders in the context of this anniversary. The next steps in the development of immunotherapies will require combinatorial approaches mixing antibodies against various targets (e.g., Aβ, α-syn, Tau, and TDP43) with small molecules that block toxicity, aggregation, inflammation, and promote cell survival.
Collapse
Affiliation(s)
- Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,Division of Neurosciences and Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
95
|
Palomo V, Tosat-Bitrian C, Nozal V, Nagaraj S, Martin-Requero A, Martinez A. TDP-43: A Key Therapeutic Target beyond Amyotrophic Lateral Sclerosis. ACS Chem Neurosci 2019; 10:1183-1196. [PMID: 30785719 DOI: 10.1021/acschemneuro.9b00026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Accumulation of TDP-43 in the cytoplasm of diseased neurons is the pathological hallmark of frontotemporal dementia-TDP (FTLD-TDP) and amyotrophic lateral sclerosis (ALS), two diseases that lack efficacious medicine to prevent or to stop disease progression. The discovery of mutations in the TARDBP gene (encoding the nuclear protein known as TDP-43) in both FTLD and ALS patients provided evidence for a link between TDP-43 alterations and neurodegeneration. Our understanding of TDP-43 function has advanced profoundly in the past several years; however, its complete role and the molecular mechanisms that lead to disease are not fully understood. Here we summarize the recent studies of this protein, its relation to neurodegenerative diseases, and the therapeutic strategies for restoring its homeostasis with small molecules. Finally, we briefly discuss the available cellular and animal models that help to shed light on TDP-43 pathology and could serve as tools for the discovery of pharmacological agents for the treatment of TDP-43-related diseases.
Collapse
Affiliation(s)
- Valle Palomo
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | | | - Vanesa Nozal
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Siranjeevi Nagaraj
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Angeles Martin-Requero
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| |
Collapse
|
96
|
Martinelli AHS, Lopes FC, John EBO, Carlini CR, Ligabue-Braun R. Modulation of Disordered Proteins with a Focus on Neurodegenerative Diseases and Other Pathologies. Int J Mol Sci 2019; 20:ijms20061322. [PMID: 30875980 PMCID: PMC6471803 DOI: 10.3390/ijms20061322] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/03/2019] [Accepted: 02/12/2019] [Indexed: 12/15/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) do not have rigid 3D structures, showing changes in their folding depending on the environment or ligands. Intrinsically disordered proteins are widely spread in eukaryotic genomes, and these proteins participate in many cell regulatory metabolism processes. Some IDPs, when aberrantly folded, can be the cause of some diseases such as Alzheimer′s, Parkinson′s, and prionic, among others. In these diseases, there are modifications in parts of the protein or in its entirety. A common conformational variation of these IDPs is misfolding and aggregation, forming, for instance, neurotoxic amyloid plaques. In this review, we discuss some IDPs that are involved in neurodegenerative diseases (such as beta amyloid, alpha synuclein, tau, and the “IDP-like” PrP), cancer (p53, c-Myc), and diabetes (amylin), focusing on the structural changes of these IDPs that are linked to such pathologies. We also present the IDP modulation mechanisms that can be explored in new strategies for drug design. Lastly, we show some candidate drugs that can be used in the future for the treatment of diseases caused by misfolded IDPs, considering that cancer therapy has more advanced research in comparison to other diseases, while also discussing recent and future developments in this area of research. Therefore, we aim to provide support to the study of IDPs and their modulation mechanisms as promising approaches to combat such severe diseases.
Collapse
Affiliation(s)
- Anne H S Martinelli
- Department of Molecular Biology and Biotechnology & Department of Biophysics, Biosciences Institute-IB, (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
| | - Fernanda C Lopes
- Center for Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
- Graduate Program in Cell and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
| | - Elisa B O John
- Center for Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
- Graduate Program in Cell and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
| | - Célia R Carlini
- Graduate Program in Cell and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre CEP 91501-970, RS, Brazil.
- Graduate Program in Medicine and Health Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre CEP 91410-000, RS, Brazil.
- Brain Institute-InsCer, Laboratory of Neurotoxins, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre CEP 90610-000, RS, Brazil.
| | - Rodrigo Ligabue-Braun
- Department of Pharmaceutical Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre CEP 90050-170, RS, Brazil.
| |
Collapse
|
97
|
Cox TO, Gunther EC, Brody AH, Chiasseu MT, Stoner A, Smith LM, Haas LT, Hammersley J, Rees G, Dosanjh B, Groves M, Gardener M, Dobson C, Vaughan T, Chessell I, Billinton A, Strittmatter SM. Anti-PrP C antibody rescues cognition and synapses in transgenic alzheimer mice. Ann Clin Transl Neurol 2019; 6:554-574. [PMID: 30911579 PMCID: PMC6414488 DOI: 10.1002/acn3.730] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/28/2022] Open
Abstract
Objective Amyloid-beta oligomers (Aßo) trigger the development of Alzheimer's disease (AD) pathophysiology. Cellular prion protein (PrPC) initiates synaptic damage as a high affinity receptor for Aßo. Here, we evaluated the preclinical therapeutic efficacy of a fully human monoclonal antibody against PrPC. This AZ59 antibody selectively targets the Aβo binding site in the amino-terminal unstructured domain of PrPC to avoid any potential risk of direct toxicity. Methods Potency of AZ59 was evaluated by binding to PrPC, blockade of Aβo interaction and interruption of Aβo signaling. AZ59 was administered to mice by weekly intraperitoneal dosing and brain antibody measured. APP/PS1 transgenic mice were treated with AZ59 and assessed by memory tests, by brain biochemistry and by histochemistry for Aß, gliosis and synaptic density. Results AZ59 binds PrPC with 100 pmol/L affinity and blocks human brain Aßo binding to PrPC, as well as prevents synaptotoxic signaling. Weekly i.p. dosing of 20 mg/kg AZ59 in a murine form achieves trough brain antibody levels greater than 10 nmol/L. Aged symptomatic APP/PS1 transgenic mice treated with AZ59 for 5-7 weeks show a full rescue of behavioral and synaptic loss phenotypes. This recovery occurs without clearance of plaque pathology or elimination of gliosis. AZ59 treatment also normalizes synaptic signaling abnormalities in transgenic brain. These benefits are dose-dependent and persist for at least 1 month after the last dose. Interpretation Preclinical data demonstrate that systemic AZ59 therapy rescues central synapses and memory function from transgenic Alzheimer's disease pathology, supporting a disease-modifying therapeutic potential.
Collapse
Affiliation(s)
- Timothy O. Cox
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Erik C. Gunther
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - A. Harrison Brody
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Marius T. Chiasseu
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Austin Stoner
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Levi M. Smith
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Laura T. Haas
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| | - Jayne Hammersley
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Gareth Rees
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Bhupinder Dosanjh
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Maria Groves
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Matthew Gardener
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Claire Dobson
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Tristan Vaughan
- Antibody Discovery and Protein EngineeringMedImmuneGranta ParkCambridgeCB21 6GHUK
| | - Iain Chessell
- NeuroscienceIMED Biotech UnitAstraZenecaGranta ParkCambridgeCB21 6GHUK
| | - Andrew Billinton
- NeuroscienceIMED Biotech UnitAstraZenecaGranta ParkCambridgeCB21 6GHUK
| | - Stephen M. Strittmatter
- Cellular Neuroscience Neurodegeneration & RepairDepartments of Neurology and of NeuroscienceYale University School of MedicineNew Haven06536Connecticut
| |
Collapse
|
98
|
Liu L, Ding L, Rovere M, Wolfe MS, Selkoe DJ. A cellular complex of BACE1 and γ-secretase sequentially generates Aβ from its full-length precursor. J Cell Biol 2019; 218:644-663. [PMID: 30626721 PMCID: PMC6363461 DOI: 10.1083/jcb.201806205] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/12/2018] [Accepted: 11/26/2018] [Indexed: 01/13/2023] Open
Abstract
Intramembrane proteolysis of transmembrane substrates by the presenilin-γ-secretase complex is preceded and regulated by shedding of the substrate's ectodomain by α- or β-secretase. We asked whether β- and γ-secretases interact to mediate efficient sequential processing of APP, generating the amyloid β (Aβ) peptides that initiate Alzheimer's disease. We describe a hitherto unrecognized multiprotease complex containing active β- and γ-secretases. BACE1 coimmunoprecipitated and cofractionated with γ-secretase in cultured cells and in mouse and human brain. An endogenous high molecular weight (HMW) complex (∼5 MD) containing β- and γ-secretases and holo-APP was catalytically active in vitro and generated a full array of Aβ peptides, with physiological Aβ42/40 ratios. The isolated complex responded properly to γ-secretase modulators. Alzheimer's-causing mutations in presenilin altered the Aβ42/40 peptide ratio generated by the HMW β/γ-secretase complex indistinguishably from that observed in whole cells. Thus, Aβ is generated from holo-APP by a BACE1-γ-secretase complex that provides sequential, efficient RIP processing of full-length substrates to final products.
Collapse
Affiliation(s)
- Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Li Ding
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Matteo Rovere
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michael S Wolfe
- University of Kansas School of Pharmacy, Department of Medical Chemistry, Lawrence, KS
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
99
|
Fan Y, Luo R, Su LY, Xiang Q, Yu D, Xu L, Chen JQ, Bi R, Wu DD, Zheng P, Yao YG. Does the Genetic Feature of the Chinese Tree Shrew (Tupaia belangeri chinensis) Support Its Potential as a Viable Model for Alzheimer's Disease Research? J Alzheimers Dis 2019; 61:1015-1028. [PMID: 29332044 DOI: 10.3233/jad-170594] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with increasing incidence across the world and no cure at the present time. An ideal animal model would facilitate the understanding of the pathogenesis of AD and discovery of potential therapeutic targets. The Chinese tree shrew (Tupaia belangeri chinensis) has a closer genetic affinity to primates relative to rodents, and can attain ages of 8 years or older, which represents another advantage for the study of neurodegenerative diseases such as AD compared to primates. Here, we analyzed 131 AD-related genes in the Chinese tree shrew brain tissues based on protein sequence identity, positive selection, mRNA, and protein expression by comparing with those of human, rhesus monkey, and mouse. In particular, we focused on the Aβ and neurofibrillary tangles formation pathways, which are crucial to AD pathogenesis. The Chinese tree shrew had a generally higher sequence identity with human than that of mouse versus human for the AD pathway genes. There was no apparent selection on the tree shrew lineage for the AD-related genes. Moreover, expression pattern of the Aβ and neurofibrillary tangle formation pathway genes in tree shrew brain tissues resembled that of human brain tissues, with a similar aging-dependent effect. Our results provided an essential genetic basis for future AD research using the tree shrew as a viable model.
Collapse
Affiliation(s)
- Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Rongcan Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ling-Yan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qun Xiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Jia-Qi Chen
- Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ping Zheng
- Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
100
|
Gunther EC, Smith LM, Kostylev MA, Cox TO, Kaufman AC, Lee S, Folta-Stogniew E, Maynard GD, Um JW, Stagi M, Heiss JK, Stoner A, Noble GP, Takahashi H, Haas LT, Schneekloth JS, Merkel J, Teran C, Naderi ZK, Supattapone S, Strittmatter SM. Rescue of Transgenic Alzheimer's Pathophysiology by Polymeric Cellular Prion Protein Antagonists. Cell Rep 2019; 26:145-158.e8. [PMID: 30605671 PMCID: PMC6358723 DOI: 10.1016/j.celrep.2018.12.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/17/2018] [Accepted: 12/05/2018] [Indexed: 10/27/2022] Open
Abstract
Cellular prion protein (PrPC) binds the scrapie conformation of PrP (PrPSc) and oligomeric β-amyloid peptide (Aβo) to mediate transmissible spongiform encephalopathy (TSE) and Alzheimer's disease (AD), respectively. We conducted cellular and biochemical screens for compounds blocking PrPC interaction with Aβo. A polymeric degradant of an antibiotic targets Aβo binding sites on PrPC with low nanomolar affinity and prevents Aβo-induced pathophysiology. We then identified a range of negatively charged polymers with specific PrPC affinity in the low to sub-nanomolar range, from both biological (melanin) and synthetic (poly [4-styrenesulfonic acid-co-maleic acid], PSCMA) origin. Association of PSCMA with PrPC prevents Aβo/PrPC-hydrogel formation, blocks Aβo binding to neurons, and abrogates PrPSc production by ScN2a cells. We show that oral PSCMA yields effective brain concentrations and rescues APPswe/PS1ΔE9 transgenic mice from AD-related synapse loss and memory deficits. Thus, an orally active PrPC-directed polymeric agent provides a potential therapeutic approach to address neurodegeneration in AD and TSE.
Collapse
Affiliation(s)
- Erik C Gunther
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Levi M Smith
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mikhail A Kostylev
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Timothy O Cox
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Adam C Kaufman
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Suho Lee
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ewa Folta-Stogniew
- W.M. Keck Biotechnology Resource Laboratory, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | - Ji Won Um
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Massimiliano Stagi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Jacqueline K Heiss
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Austin Stoner
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Geoff P Noble
- Departments of Biochemistry, Cell Biology, and Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Hideyuki Takahashi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Laura T Haas
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - John S Schneekloth
- Yale Center for Molecular Discovery, Yale University, 600 West Campus Drive, West Haven, CT 06516, USA
| | - Janie Merkel
- Yale Center for Molecular Discovery, Yale University, 600 West Campus Drive, West Haven, CT 06516, USA
| | - Christopher Teran
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Zahra K Naderi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Surachai Supattapone
- Departments of Biochemistry, Cell Biology, and Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|