1
|
Goncalves K, Przyborski S. Modulation of the Nogo signaling pathway to overcome amyloid-β-mediated neurite inhibition in human pluripotent stem cell-derived neurites. Neural Regen Res 2025; 20:2645-2654. [PMID: 39105379 PMCID: PMC11801276 DOI: 10.4103/nrr.nrr-d-23-01628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/29/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00026/figure1/v/2024-11-05T132919Z/r/image-tiff Neuronal cell death and the loss of connectivity are two of the primary pathological mechanisms underlying Alzheimer's disease. The accumulation of amyloid-β peptides, a key hallmark of Alzheimer's disease, is believed to induce neuritic abnormalities, including reduced growth, extension, and abnormal growth cone morphology, all of which contribute to decreased connectivity. However, the precise cellular and molecular mechanisms governing this response remain unknown. In this study, we used an innovative approach to demonstrate the effect of amyloid-β on neurite dynamics in both two-dimensional and three-dimensional culture systems, in order to provide more physiologically relevant culture geometry. We utilized various methodologies, including the addition of exogenous amyloid-β peptides to the culture medium, growth substrate coating, and the utilization of human-induced pluripotent stem cell technology, to investigate the effect of endogenous amyloid-β secretion on neurite outgrowth, thus paving the way for potential future applications in personalized medicine. Additionally, we also explore the involvement of the Nogo signaling cascade in amyloid-β-induced neurite inhibition. We demonstrate that inhibition of downstream ROCK and RhoA components of the Nogo signaling pathway, achieved through modulation with Y-27632 (a ROCK inhibitor) and Ibuprofen (a Rho A inhibitor), respectively, can restore and even enhance neuronal connectivity in the presence of amyloid-β. In summary, this study not only presents a novel culture approach that offers insights into the biological process of neurite growth and inhibition, but also proposes a specific mechanism for reduced neural connectivity in the presence of amyloid-β peptides, along with potential intervention points to restore neurite growth. Thereby, we aim to establish a culture system that has the potential to serve as an assay for measuring preclinical, predictive outcomes of drugs and their ability to promote neurite outgrowth, both generally and in a patient-specific manner.
Collapse
Affiliation(s)
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, UK
- Reprocell Europe Ltd., Glasgow, UK
| |
Collapse
|
2
|
Fernández SG, Oria CG, Petit D, Annaert W, Ringman JM, Fox NC, Ryan NS, Chávez-Gutiérrez L. Spectrum of γ-Secretase dysfunction as a unifying predictor of ADAD age at onset across PSEN1, PSEN2 and APP causal genes. Mol Neurodegener 2025; 20:48. [PMID: 40281586 PMCID: PMC12032737 DOI: 10.1186/s13024-025-00832-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/30/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Autosomal Dominant Alzheimer's Disease (ADAD), caused by mutations in Presenilins (PSEN1/2) and Amyloid Precursor Protein (APP) genes, typically manifests with early onset (< 65 years). Age at symptom onset (AAO) is relatively consistent among carriers of the same PSEN1 mutation, but more variable for PSEN2 and APP variants, with these mutations associated with later AAOs than PSEN1. Understanding this clinical variability is crucial for understanding disease mechanisms, developing predictive models and tailored interventions in ADAD, with potential implications for sporadic AD. METHODS We performed biochemical assessment of γ-secretase dysfunction on 28 PSEN2 and 19 APP mutations, including disease-associated, unclear and benign variants. This analysis has been valuable in the assessment of PSEN1 variant pathogenicity, disease onset and progression. RESULTS Our analysis reveals linear correlations between the molecular composition of Aβ profiles and AAO for both PSEN2 (R2 = 0.52) and APP (R2 = 0.69) mutations. The integration of PSEN1, PSEN2 and APP correlation data shows parallel but shifted lines, suggesting a common pathogenic mechanism with gene-specific shifts in onset. We found overall "delays" in AAOs of 27 years for PSEN2 and 8 years for APP variants, compared to PSEN1. Notably, extremely inactivating PSEN1 variants delayed onset, suggesting that reduced contribution to brain APP processing underlies the later onset of PSEN2 variants. CONCLUSION This study supports a unified model of ADAD pathogenesis wherein γ-secretase dysfunction and the resulting shifts in Aβ profiles are central to disease onset across all causal genes. While similar shifts in Aβ occur across causal genes, their impact on AAO varies in the function of their contribution to APP processing in the brain. This biochemical analysis establishes quantitative relationships that enable predictive AAO modelling with implications for clinical practice and genetic research. Our findings also support the development of therapeutic strategies modulating γ-secretase across different genetic ADAD forms and potentially more broadly in AD.
Collapse
Affiliation(s)
- Sara Gutiérrez Fernández
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 Box 602, Louvain, 3000, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 Box 602, Louvain, 3000, Belgium
| | - Cristina Gan Oria
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 Box 602, Louvain, 3000, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 Box 602, Louvain, 3000, Belgium
| | - Dieter Petit
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 Box 602, Louvain, 3000, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 Box 602, Louvain, 3000, Belgium
| | - Wim Annaert
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 Box 602, Louvain, 3000, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 Box 602, Louvain, 3000, Belgium
| | - John M Ringman
- Department of Neurology, Alzheimer's Disease Research Center, Center for Health Professions, University of Southern California, 1520 Alcazar Street, Suite 210, Los Angeles, CA, 90033, USA
| | - Nick C Fox
- Dementia Research Institute at UCL, Queen Square, London, WC1 N 3BG, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1 N 3BG, UK
| | - Natalie S Ryan
- Dementia Research Institute at UCL, Queen Square, London, WC1 N 3BG, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1 N 3BG, UK
| | - Lucía Chávez-Gutiérrez
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 Box 602, Louvain, 3000, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 Box 602, Louvain, 3000, Belgium.
| |
Collapse
|
3
|
Wolfe MS. Presenilin, γ-Secretase, and the Search for Pathogenic Triggers of Alzheimer's Disease. Biochemistry 2025; 64:1662-1672. [PMID: 39996369 DOI: 10.1021/acs.biochem.4c00830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Cerebral plaques of the amyloid β-peptide (Aβ) are a defining pathology in Alzheimer's disease (AD). The amyloid hypothesis of AD pathogenesis has dominated the field for over 30 years, ostensibly validated by rare AD-causing mutations in the substrate and enzyme that produce Aβ. The γ-secretase complex carries out intramembrane proteolysis of the substrate derived from the amyloid precursor protein (APP). Mutations in APP and presenilin, the catalytic component of γ-secretase, typically increase the ratio of aggregation-prone 42-residue Aβ (Aβ42) over the more soluble 40-residue form (Aβ40). Nevertheless, the inability to clarify how Aβ aggregation leads to neurodegeneration, along with poor progress in developing effective AD therapeutics that target Aβ, raises concern about whether Aβ is the primary disease driver. γ-Secretase carries out processive proteolysis on the APP substrate, producing long Aβ peptides that are generally trimmed in tripeptide intervals to shorter secreted peptides. Recent studies on effects of AD-causing mutations on the complicated proteolytic processing of the APP substrate by γ-secretase has led to the discovery that these mutations reduce─but do not abolish─processive proteolysis. Reduced proteolysis is apparently due to stabilization of enzyme-substrate complexes, and these stalled substrate-bound γ-secretase complexes can trigger synaptic degeneration even in the absence of Aβ production. Thus, the stalled process rather than the proteolytic products may be a principal initiator of AD pathogenesis. This new amyloid-independent hypothesis suggests that pharmacological agents that rescue stalled γ-secretase enzyme-substrate complexes might be effective therapeutics for AD prevention and/or treatment.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
4
|
Chou CC, Vest R, Prado MA, Wilson-Grady J, Paulo JA, Shibuya Y, Moran-Losada P, Lee TT, Luo J, Gygi SP, Kelly JW, Finley D, Wernig M, Wyss-Coray T, Frydman J. Proteostasis and lysosomal repair deficits in transdifferentiated neurons of Alzheimer's disease. Nat Cell Biol 2025; 27:619-632. [PMID: 40140603 PMCID: PMC11991917 DOI: 10.1038/s41556-025-01623-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 01/21/2025] [Indexed: 03/28/2025]
Abstract
Ageing is the most prominent risk factor for Alzheimer's disease (AD). However, the cellular mechanisms linking neuronal proteostasis decline to the characteristic aberrant protein deposits in the brains of patients with AD remain elusive. Here we develop transdifferentiated neurons (tNeurons) from human dermal fibroblasts as a neuronal model that retains ageing hallmarks and exhibits AD-linked vulnerabilities. Remarkably, AD tNeurons accumulate proteotoxic deposits, including phospho-tau and amyloid β, resembling those in APP mouse brains and the brains of patients with AD. Quantitative tNeuron proteomics identify ageing- and AD-linked deficits in proteostasis and organelle homeostasis, most notably in endosome-lysosomal components. Lysosomal deficits in aged tNeurons, including constitutive lysosomal damage and ESCRT-mediated lysosomal repair defects, are exacerbated in AD tNeurons and linked to inflammatory cytokine secretion and cell death. Providing support for the centrality of lysosomal deficits in AD, compounds ameliorating lysosomal function reduce amyloid β deposits and cytokine secretion. Thus, the tNeuron model system reveals impaired lysosomal homeostasis as an early event of ageing and AD.
Collapse
Affiliation(s)
- Ching-Chieh Chou
- Department of Biology, Stanford University, Stanford, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Ryan Vest
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Qinotto Inc., San Carlos, CA, USA
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yohei Shibuya
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia Moran-Losada
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ting-Ting Lee
- Department of Biology, Stanford University, Stanford, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research Inc. (PAVIR), Palo Alto, CA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Marius Wernig
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Karagas N, Young JE, Blue EE, Jayadev S. The Spectrum of Genetic Risk in Alzheimer Disease. Neurol Genet 2025; 11:e200224. [PMID: 39885961 PMCID: PMC11781270 DOI: 10.1212/nxg.0000000000200224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/13/2024] [Indexed: 02/01/2025]
Abstract
Alzheimer disease (AD), the most common dementing syndrome in the United States, is currently established by the presence of amyloid-β and tau protein biomarkers in the setting of clinical cognitive impairment. These straightforward diagnostic parameters belie an immense complexity of genetic architecture underlying risk and presentation in AD. In this review, we provide a focused overview of the current state of AD genetics. We discuss the discovery of familial autosomal dominant genes, the identification of candidate genes associated with AD, and genetic variants conferring higher risk of developing AD compared with the general population. In particular, we discuss important features of AD risk due to the APOE ε4 allele. In addition to risk, we describe how the field has made headway understanding genetic factors that may protect from AD. The biological implications and practical limitations of information gleaned from genome-wide association studies in AD over the years are also discussed. The readers will have an up-to-date understanding of where we are in our efforts to understand the layers of genetic complexity in AD.
Collapse
Affiliation(s)
- Nicholas Karagas
- Department of Neurology, Adjunct Medicine, Division Medical Genetics, University of Washington, Seattle
| | - Jessica E Young
- Department of Lab Medicine and Pathology, University of Washington, Seattle; and
| | - Elizabeth E Blue
- Division Medical Genetics, Department of Medicine, University of Washington, Seattle
| | - Suman Jayadev
- Department of Neurology, Adjunct Medicine, Division Medical Genetics, University of Washington, Seattle
| |
Collapse
|
6
|
Krishnamurthy HK, Jayaraman V, Krishna K, Wang T, Bei K, Changalath C, Rajasekaran JJ. An overview of the genes and biomarkers in Alzheimer's disease. Ageing Res Rev 2025; 104:102599. [PMID: 39612989 DOI: 10.1016/j.arr.2024.102599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and neurodegenerative disease characterized by neurofibrillary tangles (NFTs) and amyloid plaque. Familial AD is caused by mutations in the APP, PSEN1, and PSEN2 genes and these mutations result in the early onset of the disease. Sporadic AD usually affects older adults over the age of 65 years and is, therefore classified as late-onset AD (LOAD). Several risk factors associated with LOAD including the APOE gene have been identified. Moreover, GWAS studies have identified a wide array of genes and polymorphisms that are associated with LOAD risk. Currently, the diagnosis of AD involves the evaluation of memory and personality changes, cognitive impairment, and medical and family history to rule out other diseases. Laboratory tests to assess the biomarkers in the body fluids as well as MRI, CT, and PET scans to analyze the presence of plaques and NFTs are also included in the diagnosis of AD. It is important to diagnose AD before the onset of clinical symptoms, i.e. during the preclinical stage, to delay the progression and for better management of the disease. Research has been conducted to identify biomarkers of AD in the CSF, serum, saliva, and urine during the preclinical stage. Current research has identified several biomarkers and potential biomarkers in the body fluids that enhance diagnostic accuracy. Aside from genetics, other factors such as diet, physical activity, and lifestyle factors may influence the risk of developing AD. Clinical trials are underway to find potential biomarkers, diagnostic measures, and treatments for AD mainly in the preclinical stage. This review provides an overview of the genes and biomarkers of AD.
Collapse
Affiliation(s)
| | | | - Karthik Krishna
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | | | | |
Collapse
|
7
|
Li Y, Wang H, Wang Y, Chen Z, Liu Y, Tian W, Kang X, Pashang A, Kulasiri D, Yang X, Li HW, Zhang Y. Alterations in the axon initial segment plasticity is involved in early pathogenesis in Alzheimer's disease. MedComm (Beijing) 2024; 5:e768. [PMID: 39415847 PMCID: PMC11473794 DOI: 10.1002/mco2.768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, characterized by the early presence of amyloid-β (Aβ) and hyperphosphorylated tau. Identifying the neuropathological changes preceding cognitive decline is crucial for early intervention. Axon initial segment (AIS) maintains the orderly structure of the axon and is responsible for initiating action potentials (APs). To investigate the role of AIS in early stages of AD pathogenesis, we focused on alterations in the AIS of neurons from APP/PS1 mouse models harboring familial AD mutations. AIS length and electrophysiological properties were assessed in neurons using immunostaining and patch-clamp techniques. The expression and function of ankyrin G (AnkG) isoforms were evaluated by western blot and rescue experiments. We observed a significant shortening of AIS in APP/PS1 mice, which correlated with impaired action potential propagation. Furthermore, a decrease in the 480 kDa isoform of AnkG was observed. Rescue of this isoform restored AIS plasticity and improved long-term potentiation in APP/PS1 neurons. Our study implicates AIS plasticity alterations and AnkG dysregulation as early events in AD. The restoration of AIS integrity by the 480 kDa AnkG isoform presents a potential therapeutic strategy for AD, underscoring the importance of targeting AIS stability in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Li
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Han Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Yiming Wang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Zhiya Chen
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Yiqiong Liu
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Wu Tian
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Xinrui Kang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| | - Abolghasem Pashang
- Centre for Advanced Computational Solutions (C‐fACS)AGLS FacultyLincoln UniversityCanterburyNew Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C‐fACS)AGLS FacultyLincoln UniversityCanterburyNew Zealand
| | - Xiaoli Yang
- Division of Life Sciences and MedicineDepartment of NeurologyInstitute on Aging and Brain DisordersThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Neurodegenerative Disorder Research CenterAnhui Province Key Laboratory of Biomedical Aging ResearchDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Hung Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Yan Zhang
- State Key Laboratory of Membrane BiologySchool of Life SciencesPeking UniversityBeijingChina
| |
Collapse
|
8
|
Botella Lucena P, Heneka MT. Inflammatory aspects of Alzheimer's disease. Acta Neuropathol 2024; 148:31. [PMID: 39196440 DOI: 10.1007/s00401-024-02790-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Alzheimer´s disease (AD) stands out as the most common chronic neurodegenerative disorder. AD is characterized by progressive cognitive decline and memory loss, with neurodegeneration as its primary pathological feature. The role of neuroinflammation in the disease course has become a focus of intense research. While microglia, the brain's resident macrophages, have been pivotal to study central immune inflammation, recent evidence underscores the contributions of other cellular entities to the neuroinflammatory process. In this article, we review the inflammatory role of microglia and astrocytes, focusing on their interactions with AD's core pathologies, amyloid beta deposition, and tau tangle formation. Additionally, we also discuss how different modes of regulated cell death in AD may impact the chronic neuroinflammatory environment. This review aims to highlight the evolving landscape of neuroinflammatory research in AD and underscores the importance of considering multiple cellular contributors when developing new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo Botella Lucena
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
9
|
Sun Y, Shi Y, Li C, Shi H. Histidine Protonation Behaviors on Structural Properties and Aggregation Properties of Aβ(1-42) Mature Fibril: Approaching by Edge Effects. J Phys Chem B 2024; 128:7341-7349. [PMID: 39018428 DOI: 10.1021/acs.jpcb.4c02343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
The histidine behavior plays a crucial role in the structural and aggregation properties of protein folding and misfolding. Understanding the histidine behavior at the edge of the protein structure is critical for finding ways to disrupt fibril elongation and growth, but this impact remains poorly understood. In the current study, we used molecular dynamics simulations to investigate the edge substitution effect of histidine protonation on the structural and aggregation properties. Our data showed that ΔG1 contributed the most to binding affinity compared to ΔG2 and ΔG3. The different protonation states at the edge chain significantly impacted the secondary structure properties of the edge chain. Specifically, we found that such protonation behavior significantly affected specific regions, particularly the N-terminus (G9-Q15) and C-terminus (K28-A30). Further analysis confirmed that H6, H13, and H14 were directly involved in H-bonding networks with the C1_H14//C2_H13 interchain interactions critical for maintaining the interchain stability. Furthermore, we confirmed that H6, H13, and H14 were directly involved in the loss of the carbon skeleton contact in the N-terminus. Our findings indicate that the edge condition is more susceptible to changes in structural properties than the middle condition. The current study is helpful for understanding the histidine behavior hypothesis in related misfolding diseases.
Collapse
Affiliation(s)
- Yue Sun
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Yaru Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Changgui Li
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Hu Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| |
Collapse
|
10
|
Zhang J, Jiang Y, Dong X, Meng Z, Ji L, Kang Y, Liu M, Zhou W, Song W. Alpha-lipoic acid alleviates cognitive deficits in transgenic APP23/PS45 mice through a mitophagy-mediated increase in ADAM10 α-secretase cleavage of APP. Alzheimers Res Ther 2024; 16:160. [PMID: 39030577 PMCID: PMC11264788 DOI: 10.1186/s13195-024-01527-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 07/13/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Alpha-lipoic acid (ALA) has a neuroprotective effect on neurodegenerative diseases. In the clinic, ALA can improve cognitive impairments in patients with Alzheimer's disease (AD) and other dementias. Animal studies have confirmed the anti-amyloidosis effect of ALA, but its underlying mechanism remains unclear. In particular, the role of ALA in amyloid-β precursor protein (APP) metabolism has not been fully elucidated. OBJECTIVE To investigate whether ALA can reduce the amyloidogenic effect of APP in a transgenic mouse model of AD, and to study the mechanism underlying this effect. METHODS ALA was infused into 2-month-old APP23/PS45 transgenic mice for 4 consecutive months and their cognitive function and AD-like pathology were then evaluated. An ALA drug concentration gradient was applied to 20E2 cells in vitro to evaluate its effect on the expression of APP proteolytic enzymes and metabolites. The mechanism by which ALA affects APP processing was studied using GI254023X, an inhibitor of A Disintegrin and Metalloproteinase 10 (ADAM10), as well as the mitochondrial toxic drug carbonyl cyanide m-chlorophenylhydrazone (CCCP). RESULTS Administration of ALA ameliorated amyloid plaque neuropathology in the brain tissue of APP23/PS45 mice and reduced learning and memory impairment. ALA also increased the expression of ADAM10 in 20E2 cells and the non-amyloidogenic processing of APP to produce the 83 amino acid C-terminal fragment (C83). In addition to activating autophagy, ALA also significantly promoted mitophagy. BNIP3L-knockdown reduced the mat/pro ratio of ADAM10. By using CCCP, ALA was found to regulate BNIP3L-mediated mitophagy, thereby promoting the α-cleavage of APP. CONCLUSIONS The enhanced α-secretase cleavage of APP by ADAM10 is the primary mechanism through which ALA ameliorates the cognitive deficits in APP23/PS45 transgenic mice. BNIP3L-mediated mitophagy contributes to the anti-amyloid properties of ALA by facilitating the maturation of ADAM10. This study provides novel experimental evidence for the treatment of AD with ALA.
Collapse
Affiliation(s)
- Jie Zhang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanshuang Jiang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangjun Dong
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zijun Meng
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liangye Ji
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Kang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Mingjing Liu
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihui Zhou
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Weihong Song
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and the Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325001, China.
| |
Collapse
|
11
|
Shi Y, Sun Y, Li C, Wang S, Wang J, Shi H. Edge Substitution Effects of Histidine Tautomerization Behaviors on the Structural Properties and Aggregation Properties of Aβ(1-42) Mature Fibril. ACS Chem Neurosci 2024; 15:1055-1062. [PMID: 38379141 DOI: 10.1021/acschemneuro.4c00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024] Open
Abstract
Histidine behaviors play critical roles in folding and misfolding processes due to the changes in net charge and the various N/N-H orientations on imidazole rings. However, the effect of histidine tautomerization (HIE (Nε-H, ε) and HID (Nδ-H, δ) states) behaviors on the edge chain of Aβ mature fibrils remains inadequately understood, which is critical for finding a strategy to disturb fibril elongation and growth. In the current study, eight independent molecular dynamics simulations were conducted to investigate such impacts on the structural and aggregation properties. Our results from three different binding models revealed that the binding contributions of edge substitution effects are primarily located between chains 1 and 2. Histidine states significantly influence the secondary structure of each domain. Further analysis confirmed that the C1_H6//C1_E11 intrachain interaction is essential in maintaining the internal stability of chain 1, while the C1_H13//C2_H13 and C1_H14//C2_H13 interchain interactions are critical in maintaining the interchain stability of the fibril structure. Our subsequent analysis revealed that the current edge substitution leads to the loss of the C1_H13//C1_E11 intrachain and C1_H13//C2_H14 interchain interactions. The N-terminal regularity was significantly directly influenced by histidine states, particularly by the residue of C1_H13. Our study provides valuable insights into the effect of histidine behaviors on the edge chain of Aβ mature fibril, advancing our understanding of the histidine behavior hypothesis in misfolding diseases.
Collapse
Affiliation(s)
- Yaru Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Yue Sun
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Changgui Li
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Shuo Wang
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Jinping Wang
- Institute of Surface Analysis and Chemical Biology, University of Jinan, Jinan 250022, Shandong, China
| | - Hu Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| |
Collapse
|
12
|
Tian Y, Shang Q, Liang R, Viles JH. Copper(II) Can Kinetically Trap Arctic and Italian Amyloid-β 40 as Toxic Oligomers, Mimicking Cu(II) Binding to Wild-Type Amyloid-β 42: Implications for Familial Alzheimer's Disease. JACS AU 2024; 4:578-591. [PMID: 38425915 PMCID: PMC10900208 DOI: 10.1021/jacsau.3c00687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
The self-association of amyloid-β (Aβ) peptide into neurotoxic oligomers is believed to be central to Alzheimer's disease (AD). Copper is known to impact Aβ assembly, while disrupted copper homeostasis impacts phenotype in Alzheimer's models. Here we show the presence of substoichiometric Cu(II) has very different impacts on the assembly of Aβ40 and Aβ42 isoforms. Globally fitting microscopic rate constants for fibril assembly indicates copper will accelerate fibril formation of Aβ40 by increasing primary nucleation, while seeding experiments confirm that elongation and secondary nucleation rates are unaffected by Cu(II). In marked contrast, Cu(II) traps Aβ42 as prefibrillar oligomers and curvilinear protofibrils. Remarkably, the Cu(II) addition to preformed Aβ42 fibrils causes the disassembly of fibrils back to protofibrils and oligomers. The very different behaviors of the two Aβ isoforms are centered around differences in their fibril structures, as highlighted by studies of C-terminally amidated Aβ42. Arctic and Italian familiar mutations also support a key role for fibril structure in the interplay of Cu(II) with Aβ40/42 isoforms. The Cu(II) dependent switch in behavior between nonpathogenic Aβ40 wild-type and Aβ40 Arctic or Italian mutants suggests heightened neurotoxicity may be linked to the impact of physiological Cu(II), which traps these familial mutants as oligomers and curvilinear protofibrils, which cause membrane permeability and Ca(II) cellular influx.
Collapse
Affiliation(s)
- Yao Tian
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - Qi Shang
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - Ruina Liang
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - John H. Viles
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| |
Collapse
|
13
|
Yokoyama K, Barbour E, Hirschkind R, Martinez Hernandez B, Hausrath K, Lam T. Protein Corona Formation and Aggregation of Amyloid β 1-40-Coated Gold Nanocolloids. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:1728-1746. [PMID: 38194428 DOI: 10.1021/acs.langmuir.3c02923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Amyloid fibrillogenesis is a pathogenic protein aggregation process that occurs through a highly ordered process of protein-protein interactions. To better understand the protein-protein interactions involved in amyloid fibril formation, we formed nanogold colloid aggregates by stepwise additions of ∼2 nmol of amyloid β 1-40 peptide (Aβ1-40) at pH ∼3.7 and ∼25 °C. The processes of protein corona formation and building of gold colloid [diameters (d) of 20 and 80 nm] aggregates were confirmed by a red-shift of the surface plasmon resonance (SPR) band, λpeak, as the number of Aβ1-40 peptides [N(Aβ1-40)] increased. The normalized red-shift of λpeak, Δλ, was correlated with the degree of protein aggregation, and this process was approximated as the adsorption isotherm explained by the Langmuir-Freundlich model. As the coverage fraction (θ) was analyzed as a function of ϕ, which is the N(Aβ1-40) per total surface area of nanogold colloids available for adsorption, the parameters for explaining the Langmuir-Freundlich model were in good agreement for both 20 and 80 nm gold, indicating that ϕ could define the stage of the aggregation process. Surface-enhanced Raman scattering (SERS) imaging was conducted at designated values of ϕ and suggested that a protein-gold surface interaction during the initial adsorption stage may be dependent on the nanosize. The 20 nm gold case seems to prefer a relatively smaller contacting section, such as a -C-N or C═C bond, but a plane of the benzene ring may play a significant role for 80 nm gold. Regardless of the size of the particles, the β-sheet and random coil conformations were considered to be used to form gold colloid aggregates. The methodology developed in this study allows for new insights into protein-protein interactions at distinct stages of aggregation.
Collapse
Affiliation(s)
- Kazushige Yokoyama
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Eli Barbour
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Rachel Hirschkind
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Bryan Martinez Hernandez
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Kaylee Hausrath
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| | - Theresa Lam
- Department of Chemistry, The State University of New York Geneseo College, 1 College Circle, Geneseo, New York 14454, United States
| |
Collapse
|
14
|
Acharya NK, Grossman HC, Clifford PM, Levin EC, Light KR, Choi H, Swanson II RL, Kosciuk MC, Venkataraman V, Libon DJ, Matzel LD, Nagele RG. A Chronic Increase in Blood-Brain Barrier Permeability Facilitates Intraneuronal Deposition of Exogenous Bloodborne Amyloid-Beta1-42 Peptide in the Brain and Leads to Alzheimer's Disease-Relevant Cognitive Changes in a Mouse Model. J Alzheimers Dis 2024; 98:163-186. [PMID: 38393907 PMCID: PMC10977376 DOI: 10.3233/jad-231028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2023] [Indexed: 02/25/2024]
Abstract
Background Increased blood-brain barrier (BBB) permeability and amyloid-β (Aβ) peptides (especially Aβ1-42) (Aβ42) have been linked to Alzheimer's disease (AD) pathogenesis, but the nature of their involvement in AD-related neuropathological changes leading to cognitive changes remains poorly understood. Objective To test the hypothesis that chronic extravasation of bloodborne Aβ42 peptide and brain-reactive autoantibodies and their entry into the brain parenchyma via a permeable BBB contribute to AD-related pathological changes and cognitive changes in a mouse model. Methods The BBB was rendered chronically permeable through repeated injections of Pertussis toxin (PT), and soluble monomeric, fluorescein isothiocyanate (FITC)-labeled or unlabeled Aβ42 was injected into the tail-vein of 10-month-old male CD1 mice at designated intervals spanning ∼3 months. Acquisition of learned behaviors and long-term retention were assessed via a battery of cognitive and behavioral tests and linked to neuropathological changes. Results Mice injected with both PT and Aβ42 demonstrated a preferential deficit in the capacity for long-term retention and an increased susceptibility to interference in selective attention compared to mice exposed to PT or saline only. Immunohistochemical analyses revealed increased BBB permeability and entry of bloodborne Aβ42 and immunoglobulin G (IgG) into the brain parenchyma, selective neuronal binding of IgG and neuronal accumulation of Aβ42 in animals injected with both PT and Aβ42 compared to controls. Conclusion Results highlight the potential synergistic role of BBB compromise and the influx of bloodborne Aβ42 into the brain in both the initiation and progression of neuropathologic and cognitive changes associated with AD.
Collapse
Affiliation(s)
- Nimish K. Acharya
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Biomarker Discovery Center, New Jersey Institute for Successful Aging (NJISA), Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
- Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Rowan University, Glassboro, NJ, USA
| | | | - Peter M. Clifford
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- HNL Lab Medicine, Allentown, PA, USA
| | - Eli C. Levin
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Graduate Medical Education, Bayhealth Medical Center, Dover, DE, USA
| | - Kenneth R. Light
- Department of Psychology, Barnard College of Columbia University, New York, NY, USA
| | - Hana Choi
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
| | - Randel L. Swanson II
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Rehab Medicine Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Physical Medicine and Rehabilitation, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mary C. Kosciuk
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Venkat Venkataraman
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Academic and Student Affairs, Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
| | - David J. Libon
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Psychology, Rowan University, Glassboro, NJ, USA
| | - Louis D. Matzel
- Department of Psychology, Rutgers University, Piscataway, NJ, USA
| | - Robert G. Nagele
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Biomarker Discovery Center, New Jersey Institute for Successful Aging (NJISA), Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
- Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Rowan University, Glassboro, NJ, USA
| |
Collapse
|
15
|
Singh A, Ansari VA, Mahmood T, Ahsan F, Maheshwari S. Repercussion of Primary Nucleation Pathway: Dementia and Cognitive Impairment. Curr Aging Sci 2024; 17:196-204. [PMID: 38083895 DOI: 10.2174/0118746098243327231117113748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 07/05/2023] [Accepted: 09/08/2023] [Indexed: 09/10/2024]
Abstract
Neurodegenerative diseases, such as Alzheimer's, Parkinson's, and prion disease, are characterized by the conversion of normally soluble proteins or peptides into aggregated amyloidal fibrils. These diseases result in the permanent loss of specific types of neurons, making them incurable and devastating. Research on animal models of memory problems mentioned in this article contributes to our knowledge of brain health and functionality. Neurodegenerative disorders, which often lead to cognitive impairment and dementia, are becoming more prevalent as global life expectancy increases. These diseases cause severe neurological impairment and neuronal death, making them highly debilitating. Exploring and understanding these complex diseases offer significant insights into the fundamental processes essential for maintaining brain health. Exploring the intricate mechanisms underlying neurodegenerative diseases not only holds promise for potential treatments but also enhances our understanding of fundamental brain health and functionality. By unraveling the complexities of these disorders, researchers can pave the way for advancements in diagnosis, treatment, and ultimately, improving the lives of individuals affected by neurodegenerative diseases.
Collapse
Affiliation(s)
- Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Vaseem A Ansari
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Farogh Ahsan
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | | |
Collapse
|
16
|
Aurelian S, Ciobanu A, Cărare R, Stoica SI, Anghelescu A, Ciobanu V, Onose G, Munteanu C, Popescu C, Andone I, Spînu A, Firan C, Cazacu IS, Trandafir AI, Băilă M, Postoiu RL, Zamfirescu A. Topical Cellular/Tissue and Molecular Aspects Regarding Nonpharmacological Interventions in Alzheimer's Disease-A Systematic Review. Int J Mol Sci 2023; 24:16533. [PMID: 38003723 PMCID: PMC10671501 DOI: 10.3390/ijms242216533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
One of the most complex and challenging developments at the beginning of the third millennium is the alarming increase in demographic aging, mainly-but not exclusively-affecting developed countries. This reality results in one of the harsh medical, social, and economic consequences: the continuously increasing number of people with dementia, including Alzheimer's disease (AD), which accounts for up to 80% of all such types of pathology. Its large and progressive disabling potential, which eventually leads to death, therefore represents an important public health matter, especially because there is no known cure for this disease. Consequently, periodic reappraisals of different therapeutic possibilities are necessary. For this purpose, we conducted this systematic literature review investigating nonpharmacological interventions for AD, including their currently known cellular and molecular action bases. This endeavor was based on the PRISMA method, by which we selected 116 eligible articles published during the last year. Because of the unfortunate lack of effective treatments for AD, it is necessary to enhance efforts toward identifying and improving various therapeutic and rehabilitative approaches, as well as related prophylactic measures.
Collapse
Affiliation(s)
- Sorina Aurelian
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Gerontology and Geriatrics Clinic Division, St. Luca Hospital for Chronic Illnesses, 041915 Bucharest, Romania
| | - Adela Ciobanu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Roxana Cărare
- Faculty of Medicine, University of Southampton, Southampton SO16 7NS, UK;
| | - Simona-Isabelle Stoica
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Aurelian Anghelescu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Midwifery and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Vlad Ciobanu
- Computer Science Department, Politehnica University of Bucharest, 060042 Bucharest, Romania;
| | - Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Constantin Munteanu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Cristina Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Ioana Andone
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Aura Spînu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Carmen Firan
- NeuroRehabilitation Compartment, The Physical and Rehabilitation Medicine & Balneology Clinic Division, Teaching Emergency Hospital of the Ilfov County, 022104 Bucharest, Romania;
| | - Ioana Simona Cazacu
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Andreea-Iulia Trandafir
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Mihai Băilă
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Ruxandra-Luciana Postoiu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- NeuroRehabilitation Clinic Division, Teaching Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania; (S.-I.S.); (A.A.); (I.S.C.)
| | - Andreea Zamfirescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (S.A.); (A.C.); (C.P.); (I.A.); (A.S.); (A.-I.T.); (M.B.); (R.-L.P.); (A.Z.)
- Gerontology and Geriatrics Clinic Division, St. Luca Hospital for Chronic Illnesses, 041915 Bucharest, Romania
| |
Collapse
|
17
|
Li M, Jiang H, Wang Y, Xu Z, Xu H, Chen Y, Zhu J, Lin Z, Zhang M. Effect of arctigenin on neurological diseases: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 315:116642. [PMID: 37236381 DOI: 10.1016/j.jep.2023.116642] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/25/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Arctium lappa L. is a common specie of Asteraceae. Its main active ingredient, Arctigenin (AG), in mature seeds exerts pharmacological effects on the Central Nervous System (CNS). AIM OF THE STUDY To review studies on the specific effects of the AG mechanism on various CNS diseases and elucidate signal transduction mechanisms and their pharmacological actions. MATERIALS AND METHODS This investigation reviewed the essential role of AG in treating neurological disorders. Basic information on Arctium lappa L. was retrieved from the Pharmacopoeia of the People's Republic of China. The related articles from 1981 to 2022 on the network database (including CNKI, PubMed, and Wan Fang and so on) were reviewed using AG and CNS diseases-related terms such as Arctigenin and Epilepsy. RESULTS It was confirmed that AG has a therapeutic effect on Alzheimer's disease, Glioma, infectious CNS diseases (such as Toxoplasma and Japanese Encephalitis Virus), Parkinson's disease, Epilepsy, etc. In these diseases, related experiments such as a Western blot analysis revealed that AG could alter the content of some key factors (such as the reduction of Aβ in Alzheimer's disease). However, in-vivo AG's metabolic process and possible metabolites are still undetermined. CONCLUSION Based on this review, the existing pharmacological research has indeed made objective progress to elucidate how AG prevents and treats CNS diseases, especially senile degenerative disease such as Alzheimer's diseases. It was revealed that AG could be used as a potential nervous system drug as it has a wide range of effects in theory with markedly high application value, especially in the elder group. However, the existing studies are limited to in-vitro experiments; therefore, little is known about how AG metabolizes and functions in-vivo, limiting its clinical application and requiring further research.
Collapse
Affiliation(s)
- Mopu Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haibin Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanan Wang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zidi Xu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hang Xu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuetong Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianghu Zhu
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China.
| | - Zhenlang Lin
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China.
| | - Min Zhang
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China.
| |
Collapse
|
18
|
Kurth V, Ogorek I, Münch C, Lopez-Rios J, Ousson S, Lehmann S, Nieweg K, Roebroek AJM, Pietrzik CU, Beher D, Weggen S. Pathogenic Aβ production by heterozygous PSEN1 mutations is intrinsic to the mutant protein and not mediated by conformational hindrance of wild-type PSEN1. J Biol Chem 2023; 299:104997. [PMID: 37394008 PMCID: PMC10413157 DOI: 10.1016/j.jbc.2023.104997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 07/04/2023] Open
Abstract
Presenilin-1 (PSEN1) is the catalytic subunit of the intramembrane protease γ-secretase and undergoes endoproteolysis during its maturation. Heterozygous mutations in the PSEN1 gene cause early-onset familial Alzheimer's disease (eFAD) and increase the proportion of longer aggregation-prone amyloid-β peptides (Aβ42 and/or Aβ43). Previous studies had suggested that PSEN1 mutants might act in a dominant-negative fashion by functional impediment of wild-type PSEN1, but the exact mechanism by which PSEN1 mutants promote pathogenic Aβ production remains controversial. Using dual recombinase-mediated cassette exchange (dRMCE), here we generated a panel of isogenic embryonic and neural stem cell lines with heterozygous, endogenous expression of PSEN1 mutations. When catalytically inactive PSEN1 was expressed alongside the wild-type protein, we found the mutant accumulated as a full-length protein, indicating that endoproteolytic cleavage occurred strictly as an intramolecular event. Heterozygous expression of eFAD-causing PSEN1 mutants increased the Aβ42/Aβ40 ratio. In contrast, catalytically inactive PSEN1 mutants were still incorporated into the γ-secretase complex but failed to change the Aβ42/Aβ40 ratio. Finally, interaction and enzyme activity assays demonstrated the binding of mutant PSEN1 to other γ-secretase subunits, but no interaction between mutant and wild-type PSEN1 was observed. These results establish that pathogenic Aβ production is an intrinsic property of PSEN1 mutants and strongly argue against a dominant-negative effect in which PSEN1 mutants would compromise the catalytic activity of wild-type PSEN1 through conformational effects.
Collapse
Affiliation(s)
- Vanessa Kurth
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Isabella Ogorek
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany; Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Carolina Münch
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Javier Lopez-Rios
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucia, Sevilla, Spain
| | | | - Sandra Lehmann
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Katja Nieweg
- Institute of Pharmacology and Clinical Pharmacy, Philipps-University, Marburg, Germany
| | | | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | | | - Sascha Weggen
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
19
|
Tambini MD, Yin T, Yesiltepe M, Breuillaud L, Zehntner SP, d'Abramo C, Giliberto L, D'Adamio L. Aβ43 levels determine the onset of pathological amyloid deposition. J Biol Chem 2023; 299:104868. [PMID: 37257821 PMCID: PMC10404620 DOI: 10.1016/j.jbc.2023.104868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/18/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023] Open
Abstract
About 2% of Alzheimer's disease (AD) cases have early onset (FAD) and are caused by mutations in either Presenilins (PSEN1/2) or amyloid-β precursor protein (APP). PSEN1/2 catalyze production of Aβ peptides of different length from APP. Aβ peptides are the major components of amyloid plaques, a pathological lesion that characterizes AD. Analysis of mechanisms by which PSEN1/2 and APP mutations affect Aβ peptide compositions lead to the implication of the absolute or relative increase in Aβ42 in amyloid-β plaques formation. Here, to elucidate the formation of pathogenic Aβ cocktails leading to amyloid pathology, we utilized FAD rat knock-in models carrying the Swedish APP (Apps allele) and the PSEN1 L435F (Psen1LF allele) mutations. To accommodate the differences in the pathogenicity of rodent and human Aβ, these rat models are genetically engineered to express human Aβ species as both the Swedish mutant allele and the WT rat allele (called Apph) have been humanized in the Aβ-coding region. Analysis of the eight possible FAD mutant permutations indicates that the CNS levels of Aβ43, rather than absolute or relative increases in Aβ42, determine the onset of pathological amyloid deposition in FAD knock-in rats. Notably, Aβ43 was found in amyloid plaques in late onset AD and mild cognitive impairment cases, suggesting that the mechanisms initiating amyloid pathology in FAD knock-in rat reflect disease mechanisms driving amyloid pathology in late onset AD. This study helps clarifying the molecular determinants initiating amyloid pathology and supports therapeutic interventions targeting Aβ43 in AD.
Collapse
Affiliation(s)
- Marc D Tambini
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Tao Yin
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Metin Yesiltepe
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | | | | | - Cristina d'Abramo
- Litwin-Zucker Center for the Study of Alzheimer's Disease and Memory Disorders, Feinstein Institutes for Medical Research, Institute of Molecular Medicine, Northwell Health System, Manhasset, New York, USA
| | - Luca Giliberto
- Litwin-Zucker Center for the Study of Alzheimer's Disease and Memory Disorders, Feinstein Institutes for Medical Research, Institute of Molecular Medicine, Northwell Health System, Manhasset, New York, USA; Institute of Neurology and Neurosurgery, Northwell Health System, Manhasset, New York, USA
| | - Luciano D'Adamio
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA.
| |
Collapse
|
20
|
Susmitha G, Kumar R. Role of microbial dysbiosis in the pathogenesis of Alzheimer's disease. Neuropharmacology 2023; 229:109478. [PMID: 36871788 DOI: 10.1016/j.neuropharm.2023.109478] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly and detected during the advanced stages where the chances of reversal are minimum. The gut-brain axis mediates a bidirectional communication between the gut and brain, which is dependent on bacterial products such as short chain fatty acids (SCFA) and neurotransmitters. Accumulating lines of evidence suggests that AD is associated with significant alteration in the composition of gut microbiota. Furthermore, transfer of gut microbiota from healthy individuals to patients can reshape the gut microbiota structure and thus holds the potential to be exploited for the treatment of various neurodegenerative disease. Moreover, AD-associated gut dysbiosis can be partially reversed by using probiotics, prebiotics, natural compounds and dietary modifications, but need further validations. Reversal of AD associated gut dysbiosis alleviate AD-associated pathological feature and therefore can be explored as a therapeutic approach in the future. The current review article will describe various studies suggesting that AD dysbiosis occurs with AD and highlights the causal role by focussing on the interventions that hold the potential to reverse the gut dysbiosis partially.
Collapse
Affiliation(s)
- Gudimetla Susmitha
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India
| | - Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Vishakhapatnam, India.
| |
Collapse
|
21
|
Singh A, Ansari VA, Ansari TM, Hasan SM, Ahsan F, Singh K, Wasim R, Maheshwari S, Ahmad A. Consequence of Dementia and Cognitive Impairment by Primary Nucleation Pathway. Horm Metab Res 2023; 55:304-314. [PMID: 37130536 DOI: 10.1055/a-2052-8462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
An acquired loss of cognition in several cognitive domains that is severe enough to interfere with social or professional functioning is called dementia. As well as a moderately in-depth mental status examination by a clinician to identify impairments in memory, language, attention, visuospatial cognition, such as spatial orientation, executive function, and mood, the diagnosis of dementia requires a history evaluating for cognitive decline and impairment in daily activities, with confirmation from a close friend or family member. The start and organization of the cognitive assessment can be helped by short screening tests for cognitive impairment. Clinical presentations show that neurodegenerative diseases are often incurable because patients permanently lose some types of neurons. It has been determined through an assessment that, at best, our understanding of the underlying processes is still rudimentary, which presents exciting new targets for further study as well as the development of diagnostics and drugs. A growing body of research suggests that they also advance our knowledge of the processes that are probably crucial for maintaining the health and functionality of the brain. We concentrate on a number of the animal models of memory problems that have been mentioned in this review article because dementia has numerous etiologies. Serious neurological impairment and neuronal death are the main features of neurodegenerative illnesses, which are also extremely crippling ailments. The most prevalent neurodegenerative disorders are followed by those primary nucleation pathways responsible for cognitive impairment and dementia.
Collapse
Affiliation(s)
- Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | | | | | - Farogh Ahsan
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Kuldeep Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Rufaida Wasim
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | - Asad Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, India
| |
Collapse
|
22
|
Malter JS. Pin1 and Alzheimer's disease. Transl Res 2023; 254:24-33. [PMID: 36162703 PMCID: PMC10111655 DOI: 10.1016/j.trsl.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/29/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is an immense and growing public health crisis. Despite over 100 years of investigation, the etiology remains elusive and therapy ineffective. Despite current gaps in knowledge, recent studies have identified dysfunction or loss-of-function of Pin1, a unique cis-trans peptidyl prolyl isomerase, as an important step in AD pathogenesis. Here I review the functionality of Pin1 and its role in neurodegeneration.
Collapse
Affiliation(s)
- James S Malter
- Department of Pathology, UT Southwestern Medical Center, 5333 Harry Hines Blvd, Dallas, TX 75390.
| |
Collapse
|
23
|
PSEN2 and ABCA7 variants causing early-onset preclinical pathological changes in Alzheimer's disease: a case report and literature review. Neurol Sci 2023; 44:1987-2001. [PMID: 36701017 DOI: 10.1007/s10072-023-06602-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a debilitating and highly heritable neurodegenerative disease. Early-onset AD (EOAD) was defined as AD occurring before age 65. Although it has a high genetic risk, EOAD due to PSEN2 variation is very rare. ABCA7 is an important risk gene for AD. Previously reported cases mainly carried variations in a single pathogenic or risk gene. METHODS AND RESULTS: In this study, we report a 35-year-old female carrying variants in both the PSEN2 gene (c.640G > T p.V214L) and ABCA7 gene (c.2848G > A p.V950M). Four previously reported cases carried PSEN2 V214L, and no reported cases carried ABCA7 V950M. She had a history of migraine, patent foramen ovale, spontaneous subarachnoid hemorrhage without aneurysm, and multiple cerebral microhemorrhages. Her MMSE score was 24/30, and her MoCA score was 22/30. The concentration of Aβ42 and the ratio of Aβ42 to Aβ40 in cerebral spinal fluid were obviously decreased. Published variants of PSEN2 and ABCA7 in PubMed were reviewed, and the patients' characteristics were summarized and compared to provide information for the clinical diagnosis of AD. CONCLUSIONS It is necessary to conduct genetic screening in cases with atypical manifestations.
Collapse
|
24
|
Shi H, Sun Y, Yao Z, Bai M. New Insights into the Structural and Binding Properties on Aβ Mature Fibrils Due to Histidine Protonation Behaviors. ACS Chem Neurosci 2023; 14:218-225. [PMID: 36604946 DOI: 10.1021/acschemneuro.2c00487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Histidine tautomeric behaviors have been considered origin factors for controlling the structure and aggregation properties of misfolding peptides. Except for tautomeric behaviors, histidine protonation behaviors definitely have the same capacities due to the net charge changes and the various N/N-H orientations on imidazole rings. However, such phenomena are still unknown. In the current study, Aβ mature fibrils substituted with various protonation states were performed by molecular dynamics simulations to investigate the structure and binding properties. Our results show that all kinds of protonation states can increase the ΔG1 stability and decrease ΔG2 and ΔG3 stabilities. A significantly higher averaged β-sheet content was detected in (εεp), (εpp), and (ppp) fibrils in one, two, and three protonation stages, respectively. Impressively, we found that the substituted fibril with specific protonated states can control the N-terminus structural properties. Further analysis confirmed that H6 and H13 are more important than H14 since the H-bond donor and receptor cooperate among C1/C3/C8_H6, C1/C3/C8_H13, and C1/C3/C8_E11. Furthermore, the mechanism of protonation behaviors was discussed. The current study is helpful for understanding the histidine protonation behaviors on one, two, and three protonation stages, which provides new horizons for exploring the origin of protein folding and misfolding.
Collapse
Affiliation(s)
- Hu Shi
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.,Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Yue Sun
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Zeshuai Yao
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Min Bai
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030006, China
| |
Collapse
|
25
|
Jiang Y, Wan M, Xiao X, Lin Z, Liu X, Zhou Y, Liao X, Lin J, Zhou H, Zhou L, Weng L, Wang J, Guo J, Jiang H, Zhang Z, Xia K, Li J, Tang B, Jiao B, Shen L. GSN gene frameshift mutations in Alzheimer's disease. J Neurol Neurosurg Psychiatry 2023; 94:436-447. [PMID: 36650038 DOI: 10.1136/jnnp-2022-330465] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND The pathogenic missense mutations of the gelsolin (GSN) gene lead to familial amyloidosis of the Finnish type (FAF); however, our previous study identified GSN frameshift mutations existed in patients with Alzheimer's disease (AD). The GSN genotype-phenotype heterogeneity and the role of GSN frameshift mutations in patients with AD are unclear. METHOD In total, 1192 patients with AD and 1403 controls were screened through whole genome sequencing, and 884 patients with AD were enrolled for validation. Effects of GSN mutations were evaluated in vitro. GSN, Aβ42, Aβ40 and Aβ42/40 were detected in both plasma and cerebrospinal fluid (CSF). RESULTS Six patients with AD with GSN P3fs and K346fs mutations (0.50%, 6/1192) were identified, who were diagnosed with AD but not FAF. In addition, 13 patients with AD with GSN frameshift mutations were found in the validation cohort (1.47%, 13/884). Further in vitro experiments showed that both K346fs and P3fs mutations led to the GSN loss of function in inhibiting Aβ-induced toxicity. Moreover, a higher level of plasma (p=0.001) and CSF (p=0.005) GSN was observed in AD cases than controls, and a positive correlation was found between the CSF GSN and CSF Aβ42 (r=0.289, p=0.009). Besides, the GSN level was initially increasing and then decreasing with the disease course and cognitive decline. CONCLUSIONS GSN frameshift mutations may be associated with AD. An increase in plasma GSN is probably a compensatory reaction in AD, which is a potential biomarker for early AD.
Collapse
Affiliation(s)
- Yaling Jiang
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Meidan Wan
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - XueWen Xiao
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Zhuojie Lin
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Xixi Liu
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Yafang Zhou
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China.,Department of Geriatrics Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Xinxin Liao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China.,Department of Geriatrics Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Jingyi Lin
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Hui Zhou
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Lu Zhou
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Ling Weng
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Zhuohua Zhang
- Institute of Molecular Precision Medicine, Key Laboratory of Molecular Precision Medicine of Hunan Province, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Kun Xia
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jiada Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital Central South University, Changsha, Hunan, China .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Central South University, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
26
|
Xie D, Deng T, Zhai Z, Sun T, Xu Y. The cellular model for Alzheimer's disease research: PC12 cells. Front Mol Neurosci 2023; 15:1016559. [PMID: 36683856 PMCID: PMC9846650 DOI: 10.3389/fnmol.2022.1016559] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD) is a common age-related neurodegenerative disease characterized by progressive cognitive decline and irreversible memory impairment. Currently, several studies have failed to fully elucidate AD's cellular and molecular mechanisms. For this purpose, research on related cellular models may propose potential predictive models for the drug development of AD. Therefore, many cells characterized by neuronal properties are widely used to mimic the pathological process of AD, such as PC12, SH-SY5Y, and N2a, especially the PC12 pheochromocytoma cell line. Thus, this review covers the most systematic essay that used PC12 cells to study AD. We depict the cellular source, culture condition, differentiation methods, transfection methods, drugs inducing AD, general approaches (evaluation methods and metrics), and in vitro cellular models used in parallel with PC12 cells.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
27
|
Abstract
Probabilistic and parsimony-based arguments regarding available genetics data are used to propose that Hardy and Higgin's amyloid cascade hypothesis is valid but is commonly interpreted too narrowly to support, incorrectly, the primacy of the amyloid-β peptide (Aβ) in driving Alzheimer's disease pathogenesis. Instead, increased activity of the βCTF (C99) fragment of AβPP is the critical pathogenic determinant altered by mutations in the APP gene. This model is consistent with the regulation of APP mRNA translation via its 5' iron responsive element. Similar arguments support that the pathological effects of familial Alzheimer's disease mutations in the genes PSEN1 and PSEN2 are not exerted directly via changes in AβPP cleavage to produce different ratios of Aβ length. Rather, these mutations likely act through effects on presenilin holoprotein conformation and function, and possibly the formation and stability of multimers of presenilin holoprotein and/or of the γ-secretase complex. All fAD mutations in APP, PSEN1, and PSEN2 likely find unity of pathological mechanism in their actions on endolysosomal acidification and mitochondrial function, with detrimental effects on iron homeostasis and promotion of "pseudo-hypoxia" being of central importance. Aβ production is enhanced and distorted by oxidative stress and accumulates due to decreased lysosomal function. It may act as a disease-associated molecular pattern enhancing oxidative stress-driven neuroinflammation during the cognitive phase of the disease.
Collapse
Affiliation(s)
- Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
28
|
Liu L, Lauro BM, He A, Lee H, Bhattarai S, Wolfe MS, Bennett DA, Karch CM, Young-Pearse T, Dominantly Inherited Alzheimer Network (DIAN), Selkoe DJ. Identification of the Aβ37/42 peptide ratio in CSF as an improved Aβ biomarker for Alzheimer's disease. Alzheimers Dement 2023; 19:79-96. [PMID: 35278341 PMCID: PMC9464800 DOI: 10.1002/alz.12646] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Identifying CSF-based biomarkers for the β-amyloidosis that initiates Alzheimer's disease (AD) could provide inexpensive and dynamic tests to distinguish AD from normal aging and predict future cognitive decline. METHODS We developed immunoassays specifically detecting all C-terminal variants of secreted amyloid β-protein and identified a novel biomarker, the Aβ 37/42 ratio, that outperforms the canonical Aβ42/40 ratio as a means to evaluate the γ-secretase activity and brain Aβ accumulation. RESULTS We show that Aβ 37/42 can distinguish physiological and pathological status in (1) presenilin-1 mutant vs wild-type cultured cells, (2) AD vs control brain tissue, and (3) AD versus cognitively normal (CN) subjects in CSF, where 37/42 (AUC 0.9622) outperformed 42/40 (AUC 0.8651) in distinguishing CN from AD. DISCUSSION We conclude that the Aβ 37/42 ratio sensitively detects presenilin/γ-secretase dysfunction and better distinguishes CN from AD than Aβ42/40 in CSF. Measuring this novel ratio alongside promising phospho-tau analytes may provide highly discriminatory fluid biomarkers for AD.
Collapse
Affiliation(s)
- Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Bianca M. Lauro
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Amy He
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Hyo Lee
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Sanjay Bhattarai
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS USA
| | - Michael S. Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center Rush University Medical Center, Chicago, IL USA
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St. Louis, MO USA
- Hope Center for Neurologic Disorders, St. Louis, MO USA
| | - Tracy Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | | | - Dennis J. Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
29
|
Sun Y, Yao Z, Shi H. Structural properties of Aβ (1-40) peptide in protonation stage of one, two, and three: New insights from the histidine protonation behaviors. Int J Biol Macromol 2022; 223:1556-1561. [PMID: 36370861 DOI: 10.1016/j.ijbiomac.2022.11.061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Structural properties and aggregation tendency can be significantly influenced by histidine behaviors (histidine on Nδ-H state is defined as δ, likewise, Nε-H: ε and both Nδ-H and Nε-H: p). In current study, we investigated structural properties of Aβ(1-40) peptide during protonation evolution stage of one, two, and three by total 19 independent replica exchange molecular dynamics simulations using implicit solvent. Our results show that any kind of protonated state will promote β-sheet structure formation in comparison with deprotonated (εεε). With increase in number of protonation, the lowest β-sheet content increased. The highest averaged β-sheet structure content was detected in (δpδ) (46.0 %), (εpp) (36.8 %), and (ppp) (16.0 %) in each protonation stage. With three β-strand structures, (δpδ) shows more stable features and high hydrophobic properties. Further analysis confirmed that H13 and H14 are more important than H6. Specifically, H13 and H14 have a synergistic effect for structural formations by controlling H-bond networks in H13(p) with V39/V40 and H14(p/δ) with G37/G38. Finally, the Pearson correlation coefficient results confirmed that experimental result (ref. 44) is corresponding to our (εpp) system. Our current study will be conducive to understanding the effects of the histidine behaviors, it provides new insights for exploration protein folding and misfolding processes.
Collapse
Affiliation(s)
- Yue Sun
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Zeshuai Yao
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Hu Shi
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China; Institute of Molecular Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
30
|
Islam F, Islam MM, Khan Meem AF, Nafady MH, Islam MR, Akter A, Mitra S, Alhumaydhi FA, Emran TB, Khusro A, Simal-Gandara J, Eftekhari A, Karimi F, Baghayeri M. Multifaceted role of polyphenols in the treatment and management of neurodegenerative diseases. CHEMOSPHERE 2022; 307:136020. [PMID: 35985383 DOI: 10.1016/j.chemosphere.2022.136020] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 07/21/2022] [Accepted: 08/07/2022] [Indexed: 06/15/2023]
Abstract
Neurodegenerative diseases (NDDs) are conditions that cause neuron structure and/or function to deteriorate over time. Genetic alterations may be responsible for several NDDs. However, a multitude of physiological systems can trigger neurodegeneration. Several NDDs, such as Huntington's, Parkinson's, and Alzheimer's, are assigned to oxidative stress (OS). Low concentrations of reactive oxygen and nitrogen species are crucial for maintaining normal brain activities, as their increasing concentrations can promote neural apoptosis. OS-mediated neurodegeneration has been linked to several factors, including notable dysfunction of mitochondria, excitotoxicity, and Ca2+ stress. However, synthetic drugs are commonly utilized to treat most NDDs, and these treatments have been known to have side effects during treatment. According to providing empirical evidence, studies have discovered many occurring natural components in plants used to treat NDDs. Polyphenols are often safer and have lesser side effects. As, epigallocatechin-3-gallate, resveratrol, curcumin, quercetin, celastrol, berberine, genistein, and luteolin have p-values less than 0.05, so they are typically considered to be statistically significant. These polyphenols could be a choice of interest as therapeutics for NDDs. This review highlighted to discusses the putative effectiveness of polyphenols against the most prevalent NDDs.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Mohaimenul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Atkia Farzana Khan Meem
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Mohamed H Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza, 12568, Egypt
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Aklima Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 52571, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh; Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh.
| | - Ameer Khusro
- Department of Biotechnology, Hindustan College of Arts & Science, Padur, OMR, Chennai, 603103, India; Centre for Research and Development, Department of Biotechnology, Hindustan College of Arts & Science, Padur, OMR, Chennai, 603103, India
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, E32004, Ourense, Spain.
| | - Aziz Eftekhari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmacology & Toxicology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Karimi
- Department of Chemical Engineering, Quchan University of Technology, Quchan, Iran.
| | - Mehdi Baghayeri
- Department of Chemistry, Faculty of Science, Hakim Sabzevari University, PO. Box 397, Sabzevar, Iran.
| |
Collapse
|
31
|
Bera A, Lavanya G, Reshmi R, Dev K, Kumar R. Mechanistic and therapeutic role of Drp1 in the pathogenesis of Alzheimer's disease. Eur J Neurosci 2022; 56:5516-5531. [PMID: 35078269 DOI: 10.1111/ejn.15611] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, has emerged as the most common form of dementia in the elderly. Two major pathological hallmarks have been identified for AD: extracellular amyloid plaques and intracellular neurofibrillary tangles (NFT). Recently, dynamin-related protein 1 (Drp1) was recognized to contribute significantly towards the pathogenesis of AD. Drp1 is primarily located in the cytosol, from where it translocates to the mitochondrial outer membrane and drives the mitochondrial fission via GTP hydrolysis. Drp1 interacts with Aβ and phosphorylated tau, leading to excessive mitochondrial fragmentation, which in turn results in synaptic dysfunction, neuronal damage and cognitive decline. Several studies suggest an increase in the level of Drp1 in the post-mortem brain specimen collected from the AD patients and murine models of AD. Interestingly, heterozygous deletion of Drp1 in the transgenic murine model of AD ameliorates the mitochondrial dysfunction, improving learning and memory. The current review article discusses the possible mechanistic pathways by which Drp1 can influence the pathogenesis of AD. Besides, it will describe various inhibitors for Drp1 and their potential role as therapeutics for AD in the future.
Collapse
Affiliation(s)
- Arpita Bera
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Gantyada Lavanya
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Ravada Reshmi
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| |
Collapse
|
32
|
Sonar K, Mancera RL. Characterization of the Conformations of Amyloid Beta 42 in Solution That May Mediate Its Initial Hydrophobic Aggregation. J Phys Chem B 2022; 126:7916-7933. [PMID: 36179370 DOI: 10.1021/acs.jpcb.2c04743] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intrinsically disordered peptides, such as amyloid β42 (Aβ42), lack a well-defined structure in solution. Aβ42 can undergo abnormal aggregation and amyloidogenesis in the brain, forming fibrillar plaques, a hallmark of Alzheimer's disease. The insoluble fibrillar forms of Aβ42 exhibit well-defined, cross β-sheet structures at the molecular level and are less toxic than the soluble, intermediate disordered oligomeric forms. However, the mechanism of initial interaction of monomers and subsequent oligomerization is not well understood. The structural disorder of Aβ42 adds to the challenges of determining the structural properties of its monomers, making it difficult to understand the underlying molecular mechanism of pathogenic aggregation. Certain regions of Aβ42 are known to exhibit helical propensity in different physiological conditions. NMR spectroscopy has shown that the Aβ42 monomer at lower pH can adopt an α-helical conformation and as the pH is increased, the peptide switches to β-sheet conformation and aggregation occurs. CD spectroscopy studies of aggregation have shown the presence of an initial spike in the amount of α-helical content at the start of aggregation. Such an increase in α-helical content suggests a mechanism wherein the peptide can expose critical non-polar residues for interaction, leading to hydrophobic aggregation with other interacting peptides. We have used molecular dynamics simulations to characterize in detail the conformational landscape of monomeric Aβ42 in solution to identify molecular properties that may mediate the early stages of oligomerization. We hypothesized that conformations with α-helical structure have a higher probability of initiating aggregation because they increase the hydrophobicity of the peptide. Although random coil conformations were found to be the most dominant, as expected, α-helical conformations are thermodynamically accessible, more so than β-sheet conformations. Importantly, for the first time α-helical conformations are observed to increase the exposure of aromatic and hydrophobic residues to the aqueous solvent, favoring their hydrophobically driven interaction with other monomers to initiate aggregation. These findings constitute a first step toward characterizing the mechanism of formation of disordered, low-order oligomers of Aβ42.
Collapse
Affiliation(s)
- Krushna Sonar
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin Institute for Computation, Curtin University, P. O. Box U1987, Perth, Western Australia6845, Australia
| | - Ricardo L Mancera
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin Institute for Computation, Curtin University, P. O. Box U1987, Perth, Western Australia6845, Australia
| |
Collapse
|
33
|
Wuli W, Lin SZ, Chen SP, Tannous BA, Huang WS, Woon PY, Wu YC, Yang HH, Chen YC, Fleming RL, Rogers JT, Cahill CM, Ho TJ, Chiou TW, Harn HJ. Targeting PSEN1 by lnc-CYP3A43-2/miR-29b-2-5p to Reduce β Amyloid Plaque Formation and Improve Cognition Function. Int J Mol Sci 2022; 23:10554. [PMID: 36142465 PMCID: PMC9506169 DOI: 10.3390/ijms231810554] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Presenilin-1 (PSEN1) is a crucial subunit within the γ-secretase complex and regulates β-amyloid (Aβ) production. Accumulated evidence indicates that n-butylidenephthalide (BP) acts effectively to reduce Aβ levels in neuronal cells that are derived from trisomy 21 (Ts21) induced pluripotent stem cells (iPSCs). However, the mechanism underlying this effect remains unclear. This article aims to investigate the possible mechanisms through which BP ameliorates the development of Alzheimer's disease (AD) and verify the effectiveness of BP through animal experiments. Results from RNA microarray analysis showed that BP treatment in Ts21 iPSC-derived neuronal cells reduced long noncoding RNA (lncRNA) CYP3A43-2 levels and increased microRNA (miR)-29b-2-5p levels. Bioinformatics tool prediction analysis, biotin-labeled miR-29b-2-5p pull-down assay, and dual-luciferase reporter assay confirmed a direct negative regulatory effect for miRNA29b-2-5p on lnc-RNA-CYP3A43-2 and PSEN1. Moreover, BP administration improved short-term memory and significantly reduced Aβ accumulation in the hippocampus and cortex of 3xTg-AD mice but failed in miR-29b-2-5p mutant mice generated by CRISP/Cas9 technology. In addition, analysis of brain samples from patients with AD showed a decrease in microRNA-29b-2-5p expression in the frontal cortex region. Our results provide evidence that the LncCYP3A43-2/miR29-2-5p/PSEN1 network might be involved in the molecular mechanisms underlying BP-induced Aβ reduction.
Collapse
Affiliation(s)
- Wei Wuli
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Department of Life Science, National Dong Hwa University, Hualien 974301, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Department of Neurology, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan
| | - Shee-Ping Chen
- Buddhist Tzu Chi Stem Cells Centre, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan
| | - Bakhos A. Tannous
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA 02129, USA
| | - Wen-Sheng Huang
- Department of Nuclear Medicine, Taipei Medical University Hospital, Taipei 110301, Taiwan
| | - Peng Yeong Woon
- Taiwan Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung City 404, Taiwan
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung City 404, Taiwan
- The Biotechnology Department, College of Medical and Health Science, Asia University, Taichung City 404, Taiwan
| | - Hsueh-Hui Yang
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan
| | - Yi-Cheng Chen
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Renata Lopes Fleming
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA 02129, USA
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA 02129, USA
| | - Catherine M. Cahill
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, Boston, MA 02129, USA
| | - Tsung-Jung Ho
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Tzyy-Wen Chiou
- Department of Life Science, National Dong Hwa University, Hualien 974301, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Department of Pathology, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan
| |
Collapse
|
34
|
Haass C, Selkoe D. If amyloid drives Alzheimer disease, why have anti-amyloid therapies not yet slowed cognitive decline? PLoS Biol 2022; 20:e3001694. [PMID: 35862308 PMCID: PMC9302755 DOI: 10.1371/journal.pbio.3001694] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Strong genetic evidence supports an imbalance between production and clearance of amyloid β-protein (Aβ) in people with Alzheimer disease (AD). Microglia that are potentially involved in alternative mechanisms are actually integral to the amyloid cascade. Fluid biomarkers and brain imaging place accumulation of Aβ at the beginning of molecular and clinical changes in the disease. So why have clinical trials of anti-amyloid therapies not provided clear-cut benefits to patients with AD? Can anti-amyloid therapies robustly decrease Aβ in the human brain, and if so, could this lowering be too little, too late? These central questions in research on AD are being urgently addressed. Evidence suggests that an imbalance between production and clearance of amyloid-beta is an early, invariant feature of Alzheimer disease that drives its neuronal and glial pathology and precedes cognitive symptoms. So why are we still unable to slow cognitive decline with anti-amyloid therapies?
Collapse
Affiliation(s)
- Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- * E-mail: (CH); (DS)
| | - Dennis Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (CH); (DS)
| |
Collapse
|
35
|
Yokoyama M, Kobayashi H, Tatsumi L, Tomita T. Mouse Models of Alzheimer's Disease. Front Mol Neurosci 2022; 15:912995. [PMID: 35799899 PMCID: PMC9254908 DOI: 10.3389/fnmol.2022.912995] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by memory loss and personality changes, eventually leading to dementia. The pathological hallmarks of AD are senile plaques and neurofibrillary tangles, which comprise abnormally aggregated β-amyloid peptide (Aβ) and hyperphosphorylated tau protein. To develop preventive, diagnostic, and therapeutic strategies for AD, it is essential to establish animal models that recapitulate the pathophysiological process of AD. In this review, we will summarize the advantages and limitations of various mouse models of AD, including transgenic, knock-in, and injection models based on Aβ and tau. We will also discuss other mouse models based on neuroinflammation because recent genetic studies have suggested that microglia are crucial in the pathogenesis of AD. Although each mouse model has its advantages and disadvantages, further research on AD pathobiology will lead to the establishment of more accurate mouse models, and accelerate the development of innovative therapeutics.
Collapse
Affiliation(s)
| | | | | | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
36
|
Shen WB, Yang JJ, Yang P. RNA Hypomethylation and Unchanged DNA Methylation Levels in the Cortex of ApoE4 Carriers and Alzheimer's Disease Subjects. Curr Alzheimer Res 2022; 19:530-540. [PMID: 36045519 DOI: 10.2174/1567205019666220831125142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/07/2022] [Accepted: 07/22/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder, and ApoE4 variants are significant risk factors for AD. Epigenetic modifications are involved in AD pathology. However, it is unclear whether DNA/RNA methylation plays a role in AD pathology, and dysregulation of DNA/RNA methylation occurs in ApoE4 carriers. OBJECTIVE The present study aimed to determine whether dysregulation of DNA/RNA methylation is present in the brains of ApoE4 carriers and AD patients. METHODS In this study, postmortem brain tissues from carriers of ApoE4 and ApoE3, from AD and non- AD controls, were used in the analysis of DNA/RNA methylation, methyltransferases, and their demethylases. RESULTS Immunofluorescence staining indicates that RNA methylation is suppressed in ApoE4 carriers. Further analysis shows that the expression of RNA methyltransferases and an RNA methylation reader is suppressed in ApoE4 carriers, whereas RNA demethylase expression is increased. RNA hypomethylation occurs in NeuN+ neurons in ApoE4 carriers and AD patients. Furthermore, in ApoE4 carriers, both DNA methyltransferases and demethylases are downregulated, and overall DNA methylation levels are unchanged. CONCLUSION Our finding indicates that RNA methylation decreased in ApoE4 carriers before AD pathology and AD individuals. The expression of RNA methyltransferases and RNA methylation reader is inhibited, and RNA demethylase is upregulated in ApoE4 carriers, which leads to suppression of RNA methylation, and the suppression precedes the AD pathogenesis and persists through AD pathology.
Collapse
Affiliation(s)
- Wei-Bin Shen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201, USA
| | - James Jiao Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201, USA.,Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201, USA
| |
Collapse
|
37
|
Petit D, Fernández SG, Zoltowska KM, Enzlein T, Ryan NS, O'Connor A, Szaruga M, Hill E, Vandenberghe R, Fox NC, Chávez-Gutiérrez L. Aβ profiles generated by Alzheimer's disease causing PSEN1 variants determine the pathogenicity of the mutation and predict age at disease onset. Mol Psychiatry 2022; 27:2821-2832. [PMID: 35365805 PMCID: PMC9156411 DOI: 10.1038/s41380-022-01518-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023]
Abstract
Familial Alzheimer's disease (FAD), caused by mutations in Presenilin (PSEN1/2) and Amyloid Precursor Protein (APP) genes, is associated with an early age at onset (AAO) of symptoms. AAO is relatively consistent within families and between carriers of the same mutations, but differs markedly between individuals carrying different mutations. Gaining a mechanistic understanding of why certain mutations manifest several decades earlier than others is extremely important in elucidating the foundations of pathogenesis and AAO. Pathogenic mutations affect the protease (PSEN/γ-secretase) and the substrate (APP) that generate amyloid β (Aβ) peptides. Altered Aβ metabolism has long been associated with AD pathogenesis, with absolute or relative increases in Aβ42 levels most commonly implicated in the disease development. However, analyses addressing the relationships between these Aβ42 increments and AAO are inconsistent. Here, we investigated this central aspect of AD pathophysiology via comprehensive analysis of 25 FAD-linked Aβ profiles. Hypothesis- and data-driven approaches demonstrate linear correlations between mutation-driven alterations in Aβ profiles and AAO. In addition, our studies show that the Aβ (37 + 38 + 40) / (42 + 43) ratio offers predictive value in the assessment of 'unclear' PSEN1 variants. Of note, the analysis of PSEN1 variants presenting additionally with spastic paraparesis, indicates that a different mechanism underlies the aetiology of this distinct clinical phenotype. This study thus delivers valuable assays for fundamental, clinical and genetic research as well as supports therapeutic interventions aimed at shifting Aβ profiles towards shorter Aβ peptides.
Collapse
Affiliation(s)
- Dieter Petit
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Sara Gutiérrez Fernández
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Katarzyna Marta Zoltowska
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Thomas Enzlein
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, 68163, Mannheim, Germany
| | - Natalie S Ryan
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Antoinette O'Connor
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Maria Szaruga
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Elizabeth Hill
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Herestraat 49 box 1027, 3000, Leuven, Belgium
- Neurology Department, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Nick C Fox
- UK Dementia Research Institute at UCL, Queen Square, WC1N 3BG, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, WC1N 3BG, London, UK
| | - Lucía Chávez-Gutiérrez
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49 box 602, 3000, Leuven, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49 box 602, 3000, Leuven, Belgium.
| |
Collapse
|
38
|
Syvänen S, Meier SR, Roshanbin S, Xiong M, Faresjö R, Gustavsson T, Bonvicini G, Schlein E, Aguilar X, Julku U, Eriksson J, Sehlin D. PET Imaging in Preclinical Anti-Aβ Drug Development. Pharm Res 2022; 39:1481-1496. [PMID: 35501533 PMCID: PMC9246809 DOI: 10.1007/s11095-022-03277-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/25/2022] [Indexed: 11/21/2022]
Abstract
Positron emission tomography (PET), a medical imaging technique allowing for studies of the living human brain, has gained an important role in clinical trials of novel drugs against Alzheimer’s disease (AD). For example, PET data contributed to the conditional approval in 2021 of aducanumab, an antibody directed towards amyloid-beta (Aβ) aggregates, by showing a dose-dependent reduction in brain amyloid after treatment. In parallel to clinical studies, preclinical studies in animal models of Aβ pathology may also benefit from PET as a tool to detect target engagement and treatment effects of anti-Aβ drug candidates. PET is associated with a high level of translatability between species as similar, non-invasive protocols allow for longitudinal rather than cross-sectional studies and can be used both in a preclinical and clinical setting. This review focuses on the use of preclinical PET imaging in genetically modified animals that express human Aβ, and its present and potential future role in the development of drugs aimed at reducing brain Aβ levels as a therapeutic strategy to halt disease progression in AD.
Collapse
Affiliation(s)
- Stina Syvänen
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden.
| | - Silvio R Meier
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Mengfei Xiong
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Rebecca Faresjö
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Tobias Gustavsson
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Gillian Bonvicini
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden.,BioArctic AB, Stockholm, Sweden
| | - Eva Schlein
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Ximena Aguilar
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Ulrika Julku
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.,PET Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Uppsala University, Dag Hammarskjöldsväg 20, 75185, Uppsala, Sweden
| |
Collapse
|
39
|
Advancements in the development of multi-target directed ligands for the treatment of Alzheimer's disease. Bioorg Med Chem 2022; 61:116742. [PMID: 35398739 DOI: 10.1016/j.bmc.2022.116742] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/01/2022] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial irreversible neurological disorder which results in cognitive impairment, loss of cholinergic neurons in synapses of the basal forebrain and neuronal death. Exact pathology of the disease is not yet known however, many hypotheses have been proposed for its treatment. The available treatments including monotherapies and combination therapies are not able to combat the disease effectively because of its complex pathological mechanism. A multipotent drug for AD has the potential to bind or inhibit multiple targets responsible for the progression of the disease like aggregated Aβ, hyperphosphorylated tau proteins, cholinergic and adrenergic receptors, MAO enzymes, overactivated N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor etc. The traditional approach of one disease-one target-one drug has been rationalized to one drug-multi targets for the chronic diseases like AD and cancer. Thus, over the last decade research focus has been shifted towards the development of multi target directed ligands (MTDLs) which can simultaneously inhibit multiple targets and stop or slow the progression of the disease. The MTDLs can be more effective against AD and eliminate any possibility of drug-drug interactions. Many important active pharmacophore units have been fused, merged or incorporated into different scaffolds to synthesize new potent drugs. In the current article, we have described various hypothesis for AD and effectiveness of the MTDLs treatment strategy is discussed in detail. Different chemical scaffolds and their synthetic strategies have been described and important functionalities are identified in the chemical scaffold that have the potential to bind to the multiple targets. The important leads identified in this study with MTDL characteristics have the potential to be developed as drug candidates for the effective treatment of AD.
Collapse
|
40
|
Yang JJ. Brain insulin resistance and the therapeutic value of insulin and insulin-sensitizing drugs in Alzheimer's disease neuropathology. Acta Neurol Belg 2022; 122:1135-1142. [PMID: 35482277 DOI: 10.1007/s13760-022-01907-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
The incidence of Alzheimer's disease (AD) is significantly higher in people with diabetes. Insulin and insulin receptor (IR) signaling intermediates are expressed in the brain. Insulin exerts multiple function in the brain. The role of compromised IR signaling in AD pathogenesis and the therapeutic value of insulin attract broad attention. This review summarizes the collective insulin action in the brain related to key factors of AD pathogenesis, updates the key features of insulin resistance in the AD brain and assesses the therapeutic potential of insulin and insulin-sensitizing drugs. Insulin stimulates neural growth and survival, suppresses amyloidogenic processing of the amyloid precursor protein (AβPP) and inhibits the Tau phosphorylation kinase, glycogen synthase kinase 3β. Central nervous IR signaling regulates systemic metabolism and increases glucose availability to neurons. The expression of IR and its downstream effectors is reduced in AD brain tissues. Insulin and insulin-sensitizing drugs can improve cognitive function in AD patients and AD animal models. Systemic insulin delivery is less effective than intranasal insulin treatment. The penetrance of insulin-sensitizing drugs to the blood brain barrier is problematic and new brain-prone drugs need be developed. Insulin resistance manifested by the degradation and the altered phosphorylation of IR intermediates precedes overt AD syndrome. Type 3 diabetes as a pure form of brain insulin resistance without systemic insulin resistance is proposed as a causal factor in AD. Further research is needed for the identification of critical factors leading to impaired IR signaling and the development of new molecules to stimulate brain IR signaling.
Collapse
Affiliation(s)
- James J Yang
- Marriotts Ridge High School, 12100 Woodford Dr, Marriottsville, MD, 21104, USA.
- , 3060 Seneca Chief Trail, Ellicott City, MD, 21042, USA.
| |
Collapse
|
41
|
Pathak N, Vimal SK, Tandon I, Agrawal L, Hongyi C, Bhattacharyya S. Neurodegenerative Disorders of Alzheimer, Parkinsonism, Amyotrophic Lateral Sclerosis and Multiple Sclerosis: An Early Diagnostic Approach for Precision Treatment. Metab Brain Dis 2022; 37:67-104. [PMID: 34719771 DOI: 10.1007/s11011-021-00800-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative diseases (NDs) are characterised by progressive dysfunction of synapses, neurons, glial cells and their networks. Neurodegenerative diseases can be classified according to primary clinical features (e.g., dementia, parkinsonism, or motor neuron disease), anatomic distribution of neurodegeneration (e.g., frontotemporal degenerations, extrapyramidal disorders, or spinocerebellar degenerations), or principal molecular abnormalities. The most common neurodegenerative disorders are amyloidosis, tauopathies, a-synucleinopathy, and TAR DNA-binding protein 43 (TDP-43) proteopathy. The protein abnormalities in these disorders have abnormal conformational properties along with altered cellular mechanisms, and they exhibit motor deficit, mitochondrial malfunction, dysfunctions in autophagic-lysosomal pathways, synaptic toxicity, and more emerging mechanisms such as the roles of stress granule pathways and liquid-phase transitions. Finally, for each ND, microglial cells have been reported to be implicated in neurodegeneration, in particular, because the microglial responses can shift from neuroprotective to a deleterious role. Growing experimental evidence suggests that abnormal protein conformers act as seed material for oligomerization, spreading from cell to cell through anatomically connected neuronal pathways, which may in part explain the specific anatomical patterns observed in brain autopsy sample. In this review, we mention the human pathology of select neurodegenerative disorders, focusing on how neurodegenerative disorders (i.e., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis) represent a great healthcare problem worldwide and are becoming prevalent because of the increasing aged population. Despite many studies have focused on their etiopathology, the exact cause of these diseases is still largely unknown and until now with the only available option of symptomatic treatments. In this review, we aim to report the systematic and clinically correlated potential biomarker candidates. Although future studies are necessary for their use in early detection and progression in humans affected by NDs, the promising results obtained by several groups leads us to this idea that biomarkers could be used to design a potential therapeutic approach and preclinical clinical trials for the treatments of NDs.
Collapse
Affiliation(s)
- Nishit Pathak
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sunil Kumar Vimal
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Ishi Tandon
- Amity University Jaipur, Rajasthan, Jaipur, Rajasthan, India
| | - Lokesh Agrawal
- Graduate School of Comprehensive Human Sciences, Kansei Behavioural and Brain Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Cao Hongyi
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
42
|
Mckean NE, Handley RR, Snell RG. A Review of the Current Mammalian Models of Alzheimer's Disease and Challenges That Need to Be Overcome. Int J Mol Sci 2021; 22:13168. [PMID: 34884970 PMCID: PMC8658123 DOI: 10.3390/ijms222313168] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is one of the looming health crises of the near future. Increasing lifespans and better medical treatment for other conditions mean that the prevalence of this disease is expected to triple by 2050. The impact of AD includes both the large toll on individuals and their families as well as a large financial cost to society. So far, we have no way to prevent, slow, or cure the disease. Current medications can only alleviate some of the symptoms temporarily. Many animal models of AD have been created, with the first transgenic mouse model in 1995. Mouse models have been beset by challenges, and no mouse model fully captures the symptomatology of AD without multiple genetic mutations and/or transgenes, some of which have never been implicated in human AD. Over 25 years later, many mouse models have been given an AD-like disease and then 'cured' in the lab, only for the treatments to fail in clinical trials. This review argues that small animal models are insufficient for modelling complex disorders such as AD. In order to find effective treatments for AD, we need to create large animal models with brains and lifespan that are closer to humans, and underlying genetics that already predispose them to AD-like phenotypes.
Collapse
Affiliation(s)
- Natasha Elizabeth Mckean
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Renee Robyn Handley
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Russell Grant Snell
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, 3a Symonds Street, Auckland 1010, New Zealand; (N.E.M.); (R.R.H.)
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
43
|
Role of Receptors in Relation to Plaques and Tangles in Alzheimer's Disease Pathology. Int J Mol Sci 2021; 22:ijms222312987. [PMID: 34884789 PMCID: PMC8657621 DOI: 10.3390/ijms222312987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 12/23/2022] Open
Abstract
Despite the identification of Aβ plaques and NFTs as biomarkers for Alzheimer’s disease (AD) pathology, therapeutic interventions remain elusive, with neither an absolute prophylactic nor a curative medication available to impede the progression of AD presently available. Current approaches focus on symptomatic treatments to maintain AD patients’ mental stability and behavioral symptoms by decreasing neuronal degeneration; however, the complexity of AD pathology requires a wide range of therapeutic approaches for both preventive and curative treatments. In this regard, this review summarizes the role of receptors as a potential target for treating AD and focuses on the path of major receptors which are responsible for AD progression. This review gives an overall idea centering on major receptors, their agonist and antagonist and future prospects of viral mimicry in AD pathology. This article aims to provide researchers and developers a comprehensive idea about the different receptors involved in AD pathogenesis that may lead to finding a new therapeutic strategy to treat AD.
Collapse
|
44
|
Li C, Wang R, Zhang Y, Hu C, Ma Q. PIAS3 suppresses damage in an Alzheimer's disease cell model by inducing the STAT3-associated STAT3/Nestin/Nrf2/HO-1 pathway. Mol Med 2021; 27:150. [PMID: 34837964 PMCID: PMC8626961 DOI: 10.1186/s10020-021-00410-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/09/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most common form of dementia, is caused by the degeneration of the central nervous system (CNS). A previous study reported that signal transducer and activator of transcription 3 (STAT3) is activated during AD development; nonetheless, the related mechanism remains unknown. Thus, this study used a cell model to explore whether and how the protein inhibitor of activated STAT3 (PIAS3) is involved in AD development. METHODS Cerebrospinal fluid (CSF) specimens of 30 patients with AD and 10 normal participants were included in this study. SH-SY5Y cells were used to constructed AD model. Relevant indices were then detected and analyzed. RESULTS The results showed that compared with the control group, PIAS3 expression was substantially decreased in patients with AD and amyloid beta (Aβ)-treated SH-SY5Y cells. PIAS3 overexpression was able to reverse the detrimental effects of Aβ treatment on cell survival and growth. Further, it could also ameliorate apoptosis and oxidative stress in Aβ-treated SH-SY5Y cells. Additionally, PIAS3 was shown to reduce the activated form of STAT3 and increase the activity of the downstream Nestin/nuclear factor erythroid 2-related factor/heme oxygenase-1 pathway. CONCLUSIONS STAT3 reactivation by colivelin treatment negated the influence of PIAS3 on the survival, growth, apoptosis, and oxidative stress of Aβ-treated SH-SY5Y cells.
Collapse
Affiliation(s)
- Chen Li
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Ruili Wang
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Youyou Zhang
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Chunting Hu
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Qiaoya Ma
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xi’an, 710004 Shaanxi People’s Republic of China
| |
Collapse
|
45
|
Bao Y, Yang X, Fu Y, Li Z, Gong R, Lu W. NMDAR-dependent somatic potentiation of synaptic inputs is correlated with β amyloid-mediated neuronal hyperactivity. Transl Neurodegener 2021; 10:34. [PMID: 34496956 PMCID: PMC8424869 DOI: 10.1186/s40035-021-00260-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND β Amyloid (Aβ)-mediated neuronal hyperactivity, a key feature of the early stage of Alzheimer's disease (AD), is recently proposed to be initiated by the suppression of glutamate reuptake. Nevertheless, the underlying mechanism by which the impaired glutamate reuptake causes neuronal hyperactivity remains unclear. Chronic suppression of the glutamate reuptake causes accumulation of ambient glutamate that could diffuse from synaptic sites at the dendrites to the soma to elevate the tonic activation of somatic N-methyl-D-aspartate receptors (NMDARs). However, less attention has been paid to the potential role of tonic activity change in extrasynaptic glutamate receptors (GluRs) located at the neuronal soma on generation of neuronal hyperactivity. METHODS Whole-cell patch-clamp recordings were performed on CA1 pyramidal neurons in acute hippocampal slices exposed to TFB-threo-β-benzyloxyaspartic acid (TBOA) or human Aβ1-42 peptide oligomer. A series of dendritic patch-clamp recordings were made at different distances from the soma to identify the location of the changes in synaptic inputs. Moreover, single-channel recording in the cell-attached mode was performed to investigate the activity changes of single NMDARs at the soma. RESULTS Blocking glutamate uptake with either TBOA or the human Aβ1-42 peptide oligomer elicited potentiation of synaptic inputs in CA1 hippocampal neurons. Strikingly, this potentiation specifically occurred at the soma, depending on the activation of somatic GluN2B-containing NMDARs (GluN2B-NMDARs) and accompanied by a substantial and persistent increment in the open probability of somatic NMDARs. Blocking the activity of GluN2B-NMDARs at the soma completely reversed both the TBOA-induced or the Aβ1-42-induced somatic potentiation and neuronal hyperactivity. CONCLUSIONS The somatic potentiation of synaptic inputs may represent a novel amplification mechanism that elevates cell excitability and thus contributes to neuronal hyperactivity initiated by impaired glutamate reuptake in AD.
Collapse
Affiliation(s)
- Yifei Bao
- MOE Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Xin Yang
- MOE Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Yi Fu
- MOE Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Zhengyan Li
- MOE Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Ru Gong
- MOE Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Wei Lu
- MOE Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China. .,Department of Neurology, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontier Center for Brain Science, Fudan University, Shanghai, 200032, China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
46
|
Pope CA, Wilkins HM, Swerdlow RH, Wolfe MS. Mutations in the Amyloid-β Protein Precursor Reduce Mitochondrial Function and Alter Gene Expression Independent of 42-Residue Amyloid-β Peptide. J Alzheimers Dis 2021; 83:1039-1049. [PMID: 34366346 DOI: 10.3233/jad-210366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Dominant missense mutations in the amyloid-β protein precursor (AβPP) cause early-onset familial Alzheimer's disease (FAD) and are associated with changes in the production or properties of the amyloid-β peptide (Aβ), particularly of the 42-residue variant (Aβ 42) that deposits in the Alzheimer's disease (AD) brain. Recent findings, however, show that FAD mutations in AβPP also lead to increased production of longer Aβ variants of 45-49 residues in length. OBJECTIVE We aimed to test neurotoxicity of Aβ 42 vis-á-vis longer variants, focusing specifically on mitochondrial function, as dysfunctional mitochondria are implicated in the pathogenesis of AD. METHODS We generated SH-SY5Y human neuroblastoma cells stably expressing AβPP mutations that lead to increased production of long Aβ peptides with or without Aβ 42. These AβPP-expressing cells were tested for oxygen consumption rates (OCR) under different conditions designed to interrogate mitochondrial function. These cell lines were also examined for expression of genes important for mitochondrial or neuronal structure and function. RESULTS The mutant AβPP-expressing cells showed decreased basal OCRs as well as decreased OCRs associated with mitochondrial ATP production, even more so in the absence of Aβ 42 production. Moreover, mutant AβPP-expressing cells producing longer forms of Aβ displayed altered expression of certain mitochondrial- and neuronal-associated genes, whether or not Aβ 42 was produced. CONCLUSION These findings suggest that mutant AβPP can cause mitochondrial dysfunction that is associated with long Aβ but not with Aβ 42.
Collapse
Affiliation(s)
- Chad A Pope
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
47
|
Chen J, He HJ, Ye Q, Feng F, Wang WW, Gu Y, Han R, Xie C. Defective Autophagy and Mitophagy in Alzheimer's Disease: Mechanisms and Translational Implications. Mol Neurobiol 2021; 58:5289-5302. [PMID: 34279771 DOI: 10.1007/s12035-021-02487-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/07/2021] [Indexed: 11/30/2022]
Abstract
The main histopathology of Alzheimer's disease (AD) is featured by the extracellular accumulation of amyloid-β (Aβ) plaques and intracellular tau neurofibrillary tangles (NFT) in the brain, which is likely to result from co-pathogenic interactions among multiple factors, e.g., aging or genes. The link between defective autophagy/mitophagy and AD pathologies is still under investigation and not fully established. In this review, we consider how AD is associated with impaired autophagy and mitophagy, and how these impact pathological hallmarks as well as the potential mechanisms. This complicated interplay between autophagy or mitophagy and histopathology in AD suggests that targeting autophagy or mitophagy probably is a promising anti-AD drug candidate. Finally, we review the implications of some new insights for induction of autophagy or mitophagy as the new therapeutic way that targets processes upstream of both NFT and Aβ plaques, and hence stops the neurodegenerative course in AD.
Collapse
Affiliation(s)
- Jie Chen
- Department of Rehabilitation Medicine, Ningbo Medical Center Li Huili Hospital, Ningbo, 315000, China
| | - Hai-Jun He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qianqian Ye
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Feifei Feng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wen-Wen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yingying Gu
- Department of Psychiatry, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ruiyu Han
- NHC Key Laboratory of Family Planning and Healthy, Hebei Key Laboratory of Reproductive Medicine, Hebei Research Institute for Family Planning Science and Technology, Shijiazhuang, 050071, Hebei, China.
| | - Chenglong Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China. .,Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China. .,Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou, China.
| |
Collapse
|
48
|
Chen J, Bassot A, Giuliani F, Simmen T. Amyotrophic Lateral Sclerosis (ALS): Stressed by Dysfunctional Mitochondria-Endoplasmic Reticulum Contacts (MERCs). Cells 2021; 10:cells10071789. [PMID: 34359958 PMCID: PMC8304209 DOI: 10.3390/cells10071789] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease for which there is currently no cure. Progress in the characterization of other neurodegenerative mechanisms has shifted the spotlight onto an intracellular structure called mitochondria-endoplasmic reticulum (ER) contacts (MERCs) whose ER portion can be biochemically isolated as mitochondria-associated membranes (MAMs). Within the central nervous system (CNS), these structures control the metabolic output of mitochondria and keep sources of oxidative stress in check via autophagy. The most relevant MERC controllers in the ALS pathogenesis are vesicle-associated membrane protein-associated protein B (VAPB), a mitochondria-ER tether, and the ubiquitin-specific chaperone valosin containing protein (VCP). These two systems cooperate to maintain mitochondrial energy output and prevent oxidative stress. In ALS, mutant VAPB and VCP take a central position in the pathology through MERC dysfunction that ultimately alters or compromises mitochondrial bioenergetics. Intriguingly, both proteins are targets themselves of other ALS mutant proteins, including C9orf72, FUS, or TDP-43. Thus, a new picture emerges, where different triggers cause MERC dysfunction in ALS, subsequently leading to well-known pathological changes including endoplasmic reticulum (ER) stress, inflammation, and motor neuron death.
Collapse
Affiliation(s)
- Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (J.C.); (A.B.)
| | - Arthur Bassot
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (J.C.); (A.B.)
| | - Fabrizio Giuliani
- Department of Medicine (Neurology), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada;
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (J.C.); (A.B.)
- Correspondence: ; Tel.: +1-780-492-1546
| |
Collapse
|
49
|
Costa-Rodrigues C, Couceiro J, Moreno E. Cell competition from development to neurodegeneration. Dis Model Mech 2021; 14:269331. [PMID: 34190316 PMCID: PMC8277968 DOI: 10.1242/dmm.048926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell competition is a process by which suboptimal cells are eliminated to the benefit of cells with higher fitness. It is a surveillance mechanism that senses differences in the fitness status by several modes, such as expression of fitness fingerprints, survival factor uptake rate and resistance to mechanical stress. Fitness fingerprints-mediated cell competition recognizes isoforms of the transmembrane protein Flower, and translates the relative fitness of cells into distinct fates through the Flower code. Impairments in cell competition potentiate the development of diseases like cancer and ageing-related pathologies. In cancer, malignant cells acquire a supercompetitor behaviour, killing the neighbouring cells and overtaking the tissue, thus avoiding elimination. Neurodegenerative disorders affect millions of people and are characterized by cognitive decline and locomotor deficits. Alzheimer's disease is the most common form of dementia, and one of the largely studied diseases. However, the cellular processes taking place remain unclear. Drosophila melanogaster is an emerging neurodegeneration model due to its versatility as a tool for genetic studies. Research in a Drosophila Alzheimer's disease model detected fitness markers in the suboptimal and hyperactive neurons, thus establishing a link between cell competition and Alzheimer's disease. In this Review, we overview cell competition and the new insights related to neurodegenerative disorders, and discuss how research in the field might contribute to the development of new therapeutic targets for these diseases.
Collapse
Affiliation(s)
| | - Joana Couceiro
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Eduardo Moreno
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
50
|
Poddar MK, Banerjee S, Chakraborty A, Dutta D. Metabolic disorder in Alzheimer's disease. Metab Brain Dis 2021; 36:781-813. [PMID: 33638805 DOI: 10.1007/s11011-021-00673-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD), a well known aging-induced neurodegenerative disease is related to amyloid proteinopathy. This proteinopathy occurs due to abnormalities in protein folding, structure and thereby its function in cells. The root cause of such kind of proteinopathy and its related neurodegeneration is a disorder in metabolism, rather metabolomics of the major as well as minor nutrients. Metabolomics is the most relevant "omics" platform that offers a great potential for the diagnosis and prognosis of neurodegenerative diseases as an individual's metabolome. In recent years, the research on such kinds of neurodegenerative diseases, especially aging-related disorders is broadened its scope towards metabolic function. Different neurotransmitter metabolisms are also involved with AD and its associated neurodegeneration. The genetic and epigenetic backgrounds are also noteworthy. In this review, the physiological changes of AD in relation to its corresponding biochemical, genetic and epigenetic involvements including its (AD) therapeutic aspects are discussed.
Collapse
Affiliation(s)
- Mrinal K Poddar
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India.
| | - Soumyabrata Banerjee
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Departrment of Psychology, Neuroscience Program, Field Neurosciences Institute Research Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Apala Chakraborty
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
| | - Debasmita Dutta
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, 58102, USA
| |
Collapse
|