1
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [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: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Engelhardt E, Resende EDPF, Gomes KB. Physiopathological mechanisms underlying Alzheimer's disease: a narrative review. Dement Neuropsychol 2024; 18:e2024VR01. [PMID: 39697643 PMCID: PMC11654088 DOI: 10.1590/1980-5764-dn-2024-vr01] [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: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 12/20/2024] Open
Abstract
The neuropathological signature of Alzheimer's disease (AD) comprises mainly amyloid plaques, and neurofibrillary tangles, resulting in synaptic and neuronal loss. These pathological structures stem from amyloid dysfunctional metabolism according to the amyloid cascade hypothesis, leading to the formation of plaques, and apparently inducing the initiation of the abnormal tau pathway, with phosphorylation and aggregation of these proteins, ultimately causing the formation of tangles. In this narrative review, the existing hypothesis related to the pathophysiology of AD were compiled, and biological pathways were highlighted in order to identify the molecules that could represent biological markers of the disease, necessary to establish early diagnosis, as well as the selection of patients for therapeutical interventional strategies.
Collapse
Affiliation(s)
- Eliasz Engelhardt
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo Horizonte MG, Brazil
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte MG, Brazil
| | - Karina Braga Gomes
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Belo Horizonte MG, Brazil
| |
Collapse
|
3
|
Shinomoto M, Takeuchi C, Tatebe H, Kitani-Morii F, Ohmichi T, Fujino Y, Menjo K, Terada N, Osako M, Mochizuki Y, Teramukai S, Tokuda T, Mizuno T, Kasai T. Comparison between DSQIID total / sub-item scores and plasma p-tau elevation in adults with Down's syndrome. PLoS One 2024; 19:e0311878. [PMID: 39652530 PMCID: PMC11627409 DOI: 10.1371/journal.pone.0311878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/25/2024] [Indexed: 12/12/2024] Open
Abstract
The Dementia Screening Questionnaire for Individuals with Intellectual Disabilities (DSQIID) is an appropriate screening tool for detecting dementia in Down's syndrome patients. However, whether this questionnaire reflects the neuropsychiatric signs of biomarker-confirmed Alzheimer's disease in DS (DS-AD) remains unknown. To address this issue, we compared the plasma phosphorylated tau (P181tau: p-tau) level of a representative AD biomarker with the total score and each sub-score of the DSQIID. The DSQIID was completed by 43 of the 56 individuals enrolled in the study. The DSQIID total scores tended to be positively associated with age, and some sub-scores increased in an age-dependent manner. DSQIID total scores and some sub-scores were also positively correlated with plasma p-tau levels, while all significant correlations disappeared after adjusting for age. Moreover, one sub-score appeared to have a significant negative correlation with plasma p-tau levels after adjusting for age. The DSQIID likely reflects age-associated behavioral changes in patients with DS. Meanwhile, their scores did not correlate with plasma p-tau after adjusting for age, suggesting that there might be room for improvement in the DSQIID for detecting DS-AD.
Collapse
Affiliation(s)
- Makiko Shinomoto
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chisen Takeuchi
- Department of Neurology, Tokyo Metropolitan Kita Medical and Rehabilitation Center for the Disabled, Tokyo, Japan
- Department of Genetic Medicine, Jikei University, Tokyo, Japan
| | - Harutsugu Tatebe
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Fukiko Kitani-Morii
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Molecular Pathobiology of Brain Diseases, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuma Ohmichi
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuzo Fujino
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanako Menjo
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Miho Osako
- Department of Neurology, Tokyo Metropolitan Kita Medical and Rehabilitation Center for the Disabled, Tokyo, Japan
| | - Yoko Mochizuki
- Department of Neurology, Tokyo Metropolitan Kita Medical and Rehabilitation Center for the Disabled, Tokyo, Japan
| | - Satoshi Teramukai
- Department of Biostatistics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiko Tokuda
- Department of Genetic Medicine, Jikei University, Tokyo, Japan
| | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takashi Kasai
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Huang T, Lam XJ, Lim CT, Jusoh N, Fakurazi S, Cheah PS, Ling KH. Understanding perspectives and research trends in Down syndrome neuropathogenesis: A bibliometric analysis. JOURNAL OF INTELLECTUAL DISABILITIES : JOID 2024:17446295241299160. [PMID: 39533897 DOI: 10.1177/17446295241299160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Down syndrome (DS), characterised by compromised brain development and intellectual challenges, often manifests Alzheimer's disease (AD) -like symptoms. Utilising the Web of Science Core Collection (WOSCC) database from January 1, 2000, to July 31, 2023, we conducted a comprehensive bibliometric analysis using VOSviewer, CiteSpace, and the R package "bibliometrix." Analyses included co-authorship, co-citation, co-occurrence, cooperative network, reference, and keyword burst citation. Analysing 5,082 papers, the U.S. demonstrated prominence with the highest number of research organisations and citations. Keyword analysis revealed promising research areas, including "Alzheimer's disease," "development," "inflammation," and "neurogenesis". This 22-year survey of the brain with trisomy 21 research unveils key trends, contributors, and focal areas in DS neuropathogenesis. Notably, Alzheimer 's-related genes and proteins play a pervasive role in DS neuropathological processes across patients' lifespans. The study contributes foundational knowledge for advancing research and care in the DS neuropathogenesis domain.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Xin-Jieh Lam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Chong-Teik Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Norhazlin Jusoh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia
- Malaysian Research Institute on Ageing (MyAgeing®), Universiti Putra Malaysia, Malaysia
| |
Collapse
|
5
|
Maure-Blesa L, Rodríguez-Baz I, Carmona-Iragui M, Fortea J. What Can We Learn About Alzheimer's Disease from People with Down Syndrome? Curr Top Behav Neurosci 2024. [PMID: 39509049 DOI: 10.1007/7854_2024_546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Down syndrome (DS) is the most frequent cause of intellectual disability of genetic origin, estimated to affect about 1 in 700 babies born worldwide (CDC 2023). In Europe and the United States, current estimates indicate a population prevalence of 5.6 and 6.7 per 10,000 individuals, respectively, which translates to more than 200,000 people in the United States, more than 400,000 people in Europe, and approximately six million worldwide. Advances in healthcare and the treatment of accompanying conditions have significantly prolonged the lifespan of those with DS over the past 50 years. Consequently, there is a pressing need to address the challenges associated with ageing among this population, with Alzheimer's disease (AD) being the primary concern. In this chapter, we will review the significance of studying this population to understand AD biology, the insights gained on AD in DS (DSAD), and how this knowledge can help us understand the AD not only in DS but also in the general population. We will conclude by exploring the objectives that remain to be accomplished.
Collapse
Affiliation(s)
- Lucia Maure-Blesa
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Iñigo Rodríguez-Baz
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Facultad de Medicina, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain.
| |
Collapse
|
6
|
Kaltschmidt B, Czaniera NJ, Schulten W, Kaltschmidt C. NF-κB in Alzheimer's Disease: Friend or Foe? Opposite Functions in Neurons and Glial Cells. Int J Mol Sci 2024; 25:11353. [PMID: 39518906 PMCID: PMC11545113 DOI: 10.3390/ijms252111353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a devasting neurodegenerative disease afflicting mainly glutamatergic neurons together with a massive neuroinflammation mediated by the transcription factor NF-κB. A 65%-plus increase in Alzheimer's patients by 2050 might be a major threat to society. Hallmarks of AD are neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau and amyloid beta (Aβ) plaques. Here, we review the potential involvement of transcription factor NF-κB by hereditary mutations of the tumor necrosis factor pathway in AD patients. One of the greatest genetic risk factors is APOE4. Recently, it was shown that the APOE4 allele functions as a null allele in human astrocytes not repressing NF-κB anymore. Moreover, NF-κB seems to be involved in the repair of DNA double-strand breaks during healthy learning and memory, a function blunted in AD. NF-κB could be a friend to healthy neurons by repressing apoptosis and necroptosis. But a loss of neuronal NF-κB and activation of glial NF-κB in AD makes it a foe of neuronal survival. Hopeful therapies include TNFR2 receptor bodies relieving the activation of glial NF-κB by TNFα.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| | - Nele Johanne Czaniera
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Wiebke Schulten
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| |
Collapse
|
7
|
Wohlfert AJ, Phares J, Granholm AC. The mTOR Pathway: A Common Link Between Alzheimer's Disease and Down Syndrome. J Clin Med 2024; 13:6183. [PMID: 39458132 PMCID: PMC11508835 DOI: 10.3390/jcm13206183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Down syndrome (DS) is a chromosomal condition that causes many systemic dysregulations, leading to several possible age-related diseases including Alzheimer's disease (AD). This may be due to the triplication of the Amyloid precursor protein (APP) gene or other alterations in mechanistic pathways, such as the mTOR pathway. Impairments to upstream regulators of mTOR, such as insulin, PI3K/AKT, AMPK, and amino acid signaling, have been linked to amyloid beta plaques (Aβ) and neurofibrillary tangles (NFT), the most common AD pathologies. However, the mechanisms involved in the progression of pathology in human DS-related AD (DS-AD) are not fully investigated to date. Recent advancements in omics platforms are uncovering new insights into neurodegeneration. Genomics, spatial transcriptomics, proteomics, and metabolomics are novel methodologies that provide more data in greater detail than ever before; however, these methods have not been used to analyze the mTOR pathways in connection to DS-AD. Using these new techniques can unveil unexpected insights into pathological cellular mechanisms through an unbiased approach.
Collapse
Affiliation(s)
- Abigail J. Wohlfert
- Department of Modern Human Anatomy and Cell & Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Jeremiah Phares
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| | - Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| |
Collapse
|
8
|
Lifshitz H, Shnitzer-Meirovich S, Koslovsky M, Yozevitch R. Exploring crystallized and fluid intelligence in down syndrome using graph theory. Sci Rep 2024; 14:23738. [PMID: 39390071 PMCID: PMC11467378 DOI: 10.1038/s41598-024-74815-5] [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: 02/06/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
This cross-sectional study examined the cognitive performance of crystallized intelligence (Gc) and fluid intelligence (Gf) in 340 individuals, comparing adults (aged 22-45) to adolescents (aged 16-21) in two groups of etiologies. Down syndrome (DS) and non-specific intellectual disability (NSID). The aim was to estimate whether their cognitive performance reflected accelerated, stable, or continuous trajectories. Participants were assessed using the Vocabulary, Similarities, Block Design, and Raven Matrix tests. ANOVA analysis indicated that adults exhibited higher scores than adolescents on three of the crystallized and fluid intelligence tests, with similar trends observed in the Raven Matrix test, thus supporting the Compensation Age Theory. Participants with NSID exhibited higher scores in Vocabulary than participants with DS. Participants with DS exhibited higher scores in Block Design and Raven than participants with NSID. There was no difference between the groups in Similarities, suggesting that the verbal ability of individuals with DS is not so impaired relative to participants with NSID. Graph analysis demonstrated divergent Gc-Gf networks between the two groups of etiologies. The DS etiology revealed more coherent connections between crystallized and fluid intelligence, especially in adulthood, compared to the diffuse and absent connections seen in adults with NSID. Thus, the relative strength in Similarities and the more coherent Gc-Gf interconnections in the DS etiology suggested a more coherent and not-so-impaired profile in a clear diagnostic etiology such as DS, especially in adulthood, compared to unclear genetic etiologies such as NSID. The findings hold educational implications for adults with ID with and without Down syndrome at least until their 40's as a time for growth and development, perhaps serving as a protective factor against possible cognitive decline in the future.
Collapse
Affiliation(s)
| | | | - Meny Koslovsky
- Department of Psychology, Ariel University, Ariel, Israel
| | - Roi Yozevitch
- Department of Computer and Software Engineering, Ariel University, Ariel, Israel.
| |
Collapse
|
9
|
Hüer T, Weitzel M, Giebel GD, Raszke P, Wasem J, Levin J, Nübling G, Wagemann O, Wlasich E, Pantel J, Tesky V, Schall A, Walendzik A. [Alzheimer's dementia in people with Down syndrome : Results of guideline-assisted expert interviews on healthcare deficits in the diagnostics and treatment as well as solution approaches]. Z Gerontol Geriatr 2024:10.1007/s00391-024-02371-8. [PMID: 39384617 DOI: 10.1007/s00391-024-02371-8] [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: 05/24/2024] [Accepted: 09/12/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND People with Down syndrome have a genetically increased risk of developing early onset Alzheimer's dementia. An interview study with healthcare providers, patient representatives and employees in residential and work facilities was conducted to identify deficits in the healthcare process and approaches to overcoming them. METHOD In this study 14 semi-structured interviews were conducted and analyzed using qualitative content analysis. RESULTS A lack of knowledge and experience on the part of medical service providers in dealing with and providing medical care for people with Down syndrome was identified as a key challenge. In addition, the diagnosis of dementia in people with Down syndrome is difficult for various reasons (including lack of appropriate diagnostic tools in standard care and lack of time or financial resources). Doubts were expressed about the efficacy of antidementia medications and the reasons for the increased use of sedatives were discussed. Attentive observation of behavior and involvement of caregivers, regular review and reduction of polypharmacy and the use of alternative behavior modification techniques were mentioned as possible solutions. CONCLUSION The identified deficits in the medical care of the target population and the approaches to solving them will be incorporated into the development of health policy recommendations in order to optimize the care situation of those affected in the long term.
Collapse
Affiliation(s)
- Theresa Hüer
- Lehrstuhl für Medizinmanagement, Universität Duisburg-Essen, Thea-Leymann-Str. 9, 45127, Essen, Deutschland.
| | - Milena Weitzel
- Lehrstuhl für Medizinmanagement, Universität Duisburg-Essen, Thea-Leymann-Str. 9, 45127, Essen, Deutschland
| | - Godwin Denk Giebel
- Lehrstuhl für Medizinmanagement, Universität Duisburg-Essen, Thea-Leymann-Str. 9, 45127, Essen, Deutschland
| | - Pascal Raszke
- Lehrstuhl für Medizinmanagement, Universität Duisburg-Essen, Thea-Leymann-Str. 9, 45127, Essen, Deutschland
| | - Jürgen Wasem
- Lehrstuhl für Medizinmanagement, Universität Duisburg-Essen, Thea-Leymann-Str. 9, 45127, Essen, Deutschland
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Deutschland
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Standort München, München, Deutschland
- Munich Cluster for Systems Neurology (SyNergy), München, Deutschland
| | - Georg Nübling
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Deutschland
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Standort München, München, Deutschland
| | - Olivia Wagemann
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Deutschland
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Standort München, München, Deutschland
| | - Elisabeth Wlasich
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Deutschland
| | - Johannes Pantel
- Institut für Allgemeinmedizin, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Deutschland
| | - Valentina Tesky
- Institut für Allgemeinmedizin, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Deutschland
| | - Arthur Schall
- Institut für Allgemeinmedizin, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Deutschland
| | - Anke Walendzik
- Essener Forschungsinstitut für Medizinmanagement GmbH, EsFoMed, Essen, Deutschland
| |
Collapse
|
10
|
Murray A, Muñiz-García A, Alić I, Nižetić D. It's good to know what to BACE the specificity of your inhibitors on. J Clin Invest 2024; 134:e183677. [PMID: 39145447 PMCID: PMC11324289 DOI: 10.1172/jci183677] [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: 08/16/2024] Open
Abstract
Production, aggregation, and clearance of the amyloid β peptide (Aβ) are important processes governing the initial pathogenesis of Alzheimer's disease (AD). Inhibition of β-site amyloid precursor protein (APP) cleaving enzyme (BACE1) (one of two key proteases responsible for Aβ production) as an AD-therapeutic approach so far has failed to yield a successful drug. BACE1 and its homologue BACE2 are frequently inhibited by the same inhibitors. Several genetic and cerebral organoid modeling studies suggest that BACE2 has dose-dependent AD-suppressing activity, which makes its unwanted inhibition potentially counterproductive for AD treatment. The in vivo effects of an unwanted cross inhibition of BACE2 have so far been impossible to monitor because of the lack of an easily accessible pharmacodynamic marker specific for BACE2 cleavage. In this issue of the JCI, work led by Stefan F. Lichtenthaler identifies soluble VEGFR3 (sVEGFR3) as a pharmacodynamic plasma marker for BACE2 activity not shared with BACE1.
Collapse
Affiliation(s)
- Aoife Murray
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ana Muñiz-García
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ivan Alić
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Dean Nižetić
- The Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
11
|
Schworer EK, Zammit MD, Wang J, Handen BL, Betthauser T, Laymon CM, Tudorascu DL, Cohen AD, Zaman SH, Ances BM, Mapstone M, Head E, Klunk WE, Christian BT, Hartley SL. Amyloid age and tau PET timeline to symptomatic Alzheimer's disease in Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311702. [PMID: 39211859 PMCID: PMC11361254 DOI: 10.1101/2024.08.08.24311702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Adults with Down syndrome (DS) are at risk for Alzheimer's disease (AD). Recent natural history cohort studies have characterized AD biomarkers, with a focus on PET amyloid-beta (Aβ) and PET tau. Leveraging these well-characterized biomarkers, the present study examined the timeline to symptomatic AD based on estimated years since reaching Aβ+, referred to as "amyloid age", and in relation to tau in a large cohort of individuals with DS. Methods In this multicenter cohort study, 25 - 57-year-old adults with DS (n = 167) were assessed twice from 2017 to 2022, with approximately 32 months between visits as part of the Alzheimer Biomarker Consortium - Down Syndrome. Adults with DS completed amyloid and tau PET scans, and were administered the modified Cued Recall Test and the Down Syndrome Mental Status Examination. Study partners completed the National Task Group-Early Detection Screen for Dementia. Findings Mixed linear regressions showed significant quadratic associations between amyloid age and cognitive performance and cubic associations between amyloid age and tau, both at baseline and across 32 months. Using broken stick regression models, differences in mCRT scores were detected beginning 2.7 years following Aβ+ in cross-sectional models, with an estimated decline of 1.3 points per year. Increases in tau began, on average, 2.7 - 6.1 years following Aβ+. On average, participants with mild cognitive impairment were 7.4 years post Aβ+ and those with dementia were 12.7 years post Aβ+. Interpretation There is a short timeline to initial cognitive decline and dementia from Aβ+ (Centiloid = 18) and tau deposition in DS relative to late onset AD. The established timeline based on amyloid age (or equivalent Centiloid values) is important for clinical practice and informing AD clinical trials, and avoids limitations of timelines based on chronological age. Funding. National Institute on Aging and the National Institute for Child Health and Human Development. Research in Context Evidence before this study: We searched PubMed for articles published involving the progression of Aβ and tau deposition in adults with Down syndrome from database inception to March 1, 2024. Terms included "amyloid", "Down syndrome", "tau", "Alzheimer's disease", "cognitive decline", and "amyloid chronicity," with no language restrictions. One previous study outlined the progression of tau in adults with Down syndrome without consideration of cognitive decline or clinical status. Other studies reported cognitive decline associated with Aβ burden and estimated years to AD symptom onset in Down syndrome. Amyloid age estimates have also been created for older neurotypical adults and compared to cognitive performance, but this has not been investigated in Down syndrome.Added value of this study: The timeline to symptomatic Alzheimer's disease in relation to amyloid, expressed as duration of Aβ+, and tau has yet to be described in adults with Down syndrome. Our longitudinal study is the first to provide a timeline of cognitive decline and transition to mild cognitive impairment and dementia in relation to Aβ+.Implications of all the available evidence: In a cohort study of 167 adults with Down syndrome, cognitive decline began 2.7 - 5.4 years and tau deposition began 2.7 - 6.1 years following Aβ+ (Centiloid = 18). Adults with Down syndrome converted to MCI after ~7 years and dementia after ~12-13 years of Aβ+. This shortened timeline to AD symptomology from Aβ+ and tau deposition in DS based on amyloid age (or corresponding Centiloid values) can inform clinical AD intervention trials and is of use in clinical settings.
Collapse
|
12
|
Rastogi M, Bartolucci M, Nanni M, Aloisio M, Vozzi D, Petretto A, Contestabile A, Cancedda L. Integrative multi-omic analysis reveals conserved cell-projection deficits in human Down syndrome brains. Neuron 2024; 112:2503-2523.e10. [PMID: 38810652 DOI: 10.1016/j.neuron.2024.05.002] [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: 01/02/2023] [Revised: 03/17/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
Down syndrome (DS) is the most common genetic cause of cognitive disability. However, it is largely unclear how triplication of a small gene subset may impinge on diverse aspects of DS brain physiopathology. Here, we took a multi-omic approach and simultaneously analyzed by RNA-seq and proteomics the expression signatures of two diverse regions of human postmortem DS brains. We found that the overexpression of triplicated genes triggered global expression dysregulation, differentially affecting transcripts, miRNAs, and proteins involved in both known and novel biological candidate pathways. Among the latter, we observed an alteration in RNA splicing, specifically modulating the expression of genes involved in cytoskeleton and axonal dynamics in DS brains. Accordingly, we found an alteration in axonal polarization in neurons from DS human iPSCs and mice. Thus, our study provides an integrated multilayer expression database capable of identifying new potential targets to aid in designing future clinical interventions for DS.
Collapse
Affiliation(s)
- Mohit Rastogi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Marina Nanni
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy
| | | | - Diego Vozzi
- Central RNA Laboratory, Istituto Italiano di Tecnologia, Genova 16152, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy.
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, Genova 16163, Italy; Dulbecco Telethon Institute, Rome 00185, Italy.
| |
Collapse
|
13
|
Kasri A, Camporesi E, Gkanatsiou E, Boluda S, Brinkmalm G, Stimmer L, Ge J, Hanrieder J, Villain N, Duyckaerts C, Vermeiren Y, Pape SE, Nicolas G, Laquerrière A, De Deyn PP, Wallon D, Blennow K, Strydom A, Zetterberg H, Potier MC. Amyloid-β peptide signature associated with cerebral amyloid angiopathy in familial Alzheimer's disease with APPdup and Down syndrome. Acta Neuropathol 2024; 148:8. [PMID: 39026031 PMCID: PMC11258176 DOI: 10.1007/s00401-024-02756-4] [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: 02/02/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024]
Abstract
Alzheimer's disease (AD) is characterized by extracellular amyloid plaques containing amyloid-β (Aβ) peptides, intraneuronal neurofibrillary tangles, extracellular neuropil threads, and dystrophic neurites surrounding plaques composed of hyperphosphorylated tau protein (pTau). Aβ can also deposit in blood vessel walls leading to cerebral amyloid angiopathy (CAA). While amyloid plaques in AD brains are constant, CAA varies among cases. The study focuses on differences observed between rare and poorly studied patient groups with APP duplications (APPdup) and Down syndrome (DS) reported to have higher frequencies of elevated CAA levels in comparison to sporadic AD (sAD), most of APP mutations, and controls. We compared Aβ and tau pathologies in postmortem brain tissues across cases and Aβ peptides using mass spectrometry (MS). We further characterized the spatial distribution of Aβ peptides with MS-brain imaging. While intraparenchymal Aβ deposits were numerous in sAD, DS with AD (DS-AD) and AD with APP mutations, these were less abundant in APPdup. On the contrary, Aβ deposits in the blood vessels were abundant in APPdup and DS-AD while only APPdup cases displayed high Aβ deposits in capillaries. Investigation of Aβ peptide profiles showed a specific increase in Aβx-37, Aβx-38 and Aβx-40 but not Aβx-42 in APPdup cases and to a lower extent in DS-AD cases. Interestingly, N-truncated Aβ2-x peptides were particularly increased in APPdup compared to all other groups. This result was confirmed by MS-imaging of leptomeningeal and parenchymal vessels from an APPdup case, suggesting that CAA is associated with accumulation of shorter Aβ peptides truncated both at N- and C-termini in blood vessels. Altogether, this study identified striking differences in the localization and composition of Aβ deposits between AD cases, particularly APPdup and DS-AD, both carrying three genomic copies of the APP gene. Detection of specific Aβ peptides in CSF or plasma of these patients could improve the diagnosis of CAA and their inclusion in anti-amyloid immunotherapy treatments.
Collapse
Affiliation(s)
- Amal Kasri
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Elena Camporesi
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Eleni Gkanatsiou
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Susana Boluda
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lev Stimmer
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Junyue Ge
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jörg Hanrieder
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Nicolas Villain
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
| | - Charles Duyckaerts
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Yannick Vermeiren
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group Nutritional Biology, Wageningen University and Research (WUR), Wageningen, The Netherlands
| | - Sarah E Pape
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Gaël Nicolas
- Department of Genetics, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Annie Laquerrière
- Department of Pathology, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, F-76000, Rouen, France
| | - Peter Paul De Deyn
- Department of Biomedical Sciences, Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - David Wallon
- Department of Neurology, CNRMAJ, Univ Rouen Normandie, Normandie Univ, Inserm U1245 and CHU Rouen, 76000, Rouen, France
| | - Kaj Blennow
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute On Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Andre Strydom
- Institute of Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, UK
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
- Department of Neurology and Alzheimer Center, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
- UK Dementia Research Institute at UCL, London, UK.
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China.
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA.
| | - Marie-Claude Potier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, APHP, Hôpital de La Pitié Salpêtrière, InsermParis, France.
| |
Collapse
|
14
|
Streng BMM, Van Coillie J, Wildenbeest JG, Binnendijk RS, Smits G, den Hartog G, Wang W, Nouta J, Linty F, Visser R, Wuhrer M, Vidarsson G, Bont LJ. IgG1 glycosylation highlights premature aging in Down syndrome. Aging Cell 2024; 23:e14167. [PMID: 38616780 PMCID: PMC11258452 DOI: 10.1111/acel.14167] [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: 12/14/2023] [Revised: 03/12/2024] [Accepted: 03/24/2024] [Indexed: 04/16/2024] Open
Abstract
Down syndrome (DS) is characterized by lowered immune competence and premature aging. We previously showed decreased antibody response following SARS-CoV-2 vaccination in adults with DS. IgG1 Fc glycosylation patterns are known to affect the effector function of IgG and are associated with aging. Here, we compare total and anti-spike (S) IgG1 glycosylation patterns following SARS-CoV-2 vaccination in DS and healthy controls (HC). Total and anti-Spike IgG1 Fc N-glycan glycoprofiles were measured in non-exposed adults with DS and controls before and after SARS-CoV-2 vaccination by liquid chromatography-mass spectrometry (LC-MS) of Fc glycopeptides. We recruited N = 44 patients and N = 40 controls. We confirmed IgG glycosylation patterns associated with aging in HC and showed premature aging in DS. In DS, we found decreased galactosylation (50.2% vs. 59.0%) and sialylation (6.7% vs. 8.5%) as well as increased fucosylation (97.0% vs. 94.6%) of total IgG. Both cohorts showed similar bisecting GlcNAc of total and anti-S IgG1 with age. In contrast, anti-S IgG1 of DS and HC showed highly comparable glycosylation profiles 28 days post vaccination. The IgG1 glycoprofile in DS exhibits strong premature aging. The combination of an early decrease in IgG1 Fc galactosylation and sialylation and increase in fucosylation is predicted to reduce complement activity and decrease FcγRIII binding and subsequent activation, respectively. The altered glycosylation patterns, combined with decreased antibody concentrations, help us understand the susceptibility to severe infections in DS. The effect of premature aging highlights the need for individuals with DS to receive tailored vaccines and/or vaccination schedules.
Collapse
Affiliation(s)
- Bianca M. M. Streng
- Department of Paediatric Infectious Diseases and Immunology, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Julie Van Coillie
- Sanquin Research and Landsteiner LaboratoryAmsterdam University Medical Center, University of AmsterdamAmsterdamThe Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
| | - Joanne G. Wildenbeest
- Department of Paediatric Infectious Diseases and Immunology, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Rob S. Binnendijk
- Centre for Immunology of Infectious Diseases and VaccinesNational Institute of Public Health and the EnvironmentBilthovenThe Netherlands
| | - Gaby Smits
- Centre for Immunology of Infectious Diseases and VaccinesNational Institute of Public Health and the EnvironmentBilthovenThe Netherlands
| | - Gerco den Hartog
- Centre for Immunology of Infectious Diseases and VaccinesNational Institute of Public Health and the EnvironmentBilthovenThe Netherlands
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical CenterLeidenThe Netherlands
| | - Jan Nouta
- Center for Proteomics and Metabolomics, Leiden University Medical CenterLeidenThe Netherlands
| | - Federica Linty
- Sanquin Research and Landsteiner LaboratoryAmsterdam University Medical Center, University of AmsterdamAmsterdamThe Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
| | - Remco Visser
- Sanquin Research and Landsteiner LaboratoryAmsterdam University Medical Center, University of AmsterdamAmsterdamThe Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical CenterLeidenThe Netherlands
| | - Gestur Vidarsson
- Sanquin Research and Landsteiner LaboratoryAmsterdam University Medical Center, University of AmsterdamAmsterdamThe Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
| | - Louis J. Bont
- Department of Paediatric Infectious Diseases and Immunology, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | |
Collapse
|
15
|
Bansal P, Banda EC, Glatt-Deeley HR, Stoddard CE, Linsley JW, Arora N, Deleschaux C, Ahern DT, Kondaveeti Y, Massey RE, Nicouleau M, Wang S, Sabariego-Navarro M, Dierssen M, Finkbeiner S, Pinter SF. A dynamic in vitro model of Down syndrome neurogenesis with trisomy 21 gene dosage correction. SCIENCE ADVANCES 2024; 10:eadj0385. [PMID: 38848354 PMCID: PMC11160455 DOI: 10.1126/sciadv.adj0385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/03/2024] [Indexed: 06/09/2024]
Abstract
Excess gene dosage from chromosome 21 (chr21) causes Down syndrome (DS), spanning developmental and acute phenotypes in terminal cell types. Which phenotypes remain amenable to intervention after development is unknown. To address this question in a model of DS neurogenesis, we derived trisomy 21 (T21) human induced pluripotent stem cells (iPSCs) alongside, otherwise, isogenic euploid controls from mosaic DS fibroblasts and equipped one chr21 copy with an inducible XIST transgene. Monoallelic chr21 silencing by XIST is near-complete and irreversible in iPSCs. Differential expression reveals that T21 neural lineages and iPSCs share suppressed translation and mitochondrial pathways and activate cellular stress responses. When XIST is induced before the neural progenitor stage, T21 dosage correction suppresses a pronounced skew toward astrogenesis in neural differentiation. Because our transgene remains inducible in postmitotic T21 neurons and astrocytes, we demonstrate that XIST efficiently represses genes even after terminal differentiation, which will empower exploration of cell type-specific T21 phenotypes that remain responsive to chr21 dosage.
Collapse
Affiliation(s)
- Prakhar Bansal
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Erin C. Banda
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Heather R. Glatt-Deeley
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Christopher E. Stoddard
- Cell and Genome Engineering Core, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Jeremy W. Linsley
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Neha Arora
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
| | - Cécile Deleschaux
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Darcy T. Ahern
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Yuvabharath Kondaveeti
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Rachael E. Massey
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| | - Michael Nicouleau
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
| | - Shijie Wang
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
| | - Miguel Sabariego-Navarro
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Human Pharmacology and Clinical Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, CA, USA
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA, USA
- Neuroscience and Biomedical Sciences Graduate Programs, University of California San Francisco, San Francisco, CA, USA
| | - Stefan F. Pinter
- Graduate Program in Genetics and Developmental Biology, UCONN Health, University of Connecticut, Farmington, CT, USA
- Department of Genetics and Genome Sciences, UCONN Health, University of Connecticut, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| |
Collapse
|
16
|
Sauvé F, Kacimi L, Prévot V. The hypothalamic-pituitary-gonadal axis and the enigma of Alzheimer disease sex differences. Nat Rev Endocrinol 2024; 20:317-318. [PMID: 38514817 PMCID: PMC7616426 DOI: 10.1038/s41574-024-00981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Affiliation(s)
- Florent Sauvé
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 days for health, DistAlz, EGID, Lille, France
| | - Loïc Kacimi
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 days for health, DistAlz, EGID, Lille, France
| | - Vincent Prévot
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 days for health, DistAlz, EGID, Lille, France.
| |
Collapse
|
17
|
Piro-Gambetti B, Schworer EK, Handen B, Glukhovskaya M, Hartley SL. Does Employment Complexity Promote Healthy Cognitive Aging in Down Syndrome? JOURNAL OF INTELLECTUAL DISABILITIES : JOID 2024; 28:499-513. [PMID: 37040598 PMCID: PMC10564966 DOI: 10.1177/17446295231169379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Adults with Down syndrome (DS) experience high risk for Alzheimer's disease (AD), but there is variability in the timing of transition from a cognitively stable state to prodromal AD and dementia. The present study examined the association between a modifiable lifestyle factor, employment complexity, and cognitive decline across two time points in adults with DS. Employment complexity, defined as the degree of problem-solving or critical thinking required for employment activities, was operationalized using the Dictionary of Occupational Titles, a system which classifies occupations based on three categories: Data, People, and Things. Eighty-seven adults with DS (M = 36.28 years, SD = 6.90 years) were included in analyses. Partial correlations revealed that lower employment complexity involving People and Things were associated with increased dementia symptoms. Lower employment complexity involving Things was also associated with memory decline. These findings have implications for vocational programs focused on job training and placement for adults with DS.
Collapse
Affiliation(s)
- Brianna Piro-Gambetti
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Human Development & Family Studies, University of Wisconsin-Madison, Madison, WI, USA
| | - Emily K Schworer
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Human Development & Family Studies, University of Wisconsin-Madison, Madison, WI, USA
| | - Benjamin Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Sigan L Hartley
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Human Development & Family Studies, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
18
|
Lee D, Antonsdottir IM, Clark ED, Porsteinsson AP. Review of valiltramiprosate (ALZ-801) for the treatment of Alzheimer's disease: a novel small molecule with disease modifying potential. Expert Opin Pharmacother 2024; 25:791-799. [PMID: 38814590 DOI: 10.1080/14656566.2024.2360069] [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: 03/31/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative condition characterized by progressive cognitive deterioration, functional impairments, and neuropsychiatric symptoms. Valiltramiprosate is a tramiprosate prodrug being investigated as a novel treatment for AD. AREAS COVERED The online databases PubMed, Embase, Web of Science, Cochrane Library, and ClinicalTrials.gov were searched using the terms 'ALZ-801' or 'valiltramiprosate.' Alzheon press releases were reviewed for emerging clinical information. Valiltramiprosate is an oral, well-tolerated synthetic valine-conjugate prodrug of tramiprosate. Valiltramiprosate's active metabolite include tramiprosate and 3-sulfopropanoic acid. Proposed mechanism of action is multiligand binding to Aβ42 which stabilizes amyloid monomers to prevent peptide aggregation and oligomerization. Pharmacokinetic studies show 52% oral bioavailability, rapid absorption, approximately 40% brain-drug exposure, and near complete renal clearance. Compared to tramiprosate, valiltramiprosate extends plasma tramiprosate half-life and improves interindividual pharmacokinetic variability. Interim analyses from valiltramiprosate's phase II biomarker trial show: (1) significant reductions in plasma p-tau181 and related AD fluid biomarkers; (2) brain structure preservation and reduced hippocampal atrophy by MRI; and (3) improvements on cognitive assessments at multiple timepoints. Its phase III clinical trial in ApoE ε4 homozygotes is near completion. EXPERT OPINION Valiltramiprosate's clinical trial data show early indications of efficacy with potential disease modifying effect in AD.
Collapse
Affiliation(s)
- Daniel Lee
- Alzheimer's Disease Care, Research and Education (AD-CARE), Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Inga M Antonsdottir
- Johns Hopkins School of Nursing, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Department of Psychiatry and Behavioral Sciences, Johns Hopkins Bayview, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Emily D Clark
- Alzheimer's Disease Care, Research and Education (AD-CARE), Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Anton P Porsteinsson
- Alzheimer's Disease Care, Research and Education (AD-CARE), Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
19
|
Staurenghi E, Testa G, Leoni V, Cecci R, Floro L, Giannelli S, Barone E, Perluigi M, Leonarduzzi G, Sottero B, Gamba P. Altered Brain Cholesterol Machinery in a Down Syndrome Mouse Model: A Possible Common Feature with Alzheimer's Disease. Antioxidants (Basel) 2024; 13:435. [PMID: 38671883 PMCID: PMC11047305 DOI: 10.3390/antiox13040435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Down syndrome (DS) is a complex chromosomal disorder considered as a genetically determined form of Alzheimer's disease (AD). Maintenance of brain cholesterol homeostasis is essential for brain functioning and development, and its dysregulation is associated with AD neuroinflammation and oxidative damage. Brain cholesterol imbalances also likely occur in DS, concurring with the precocious AD-like neurodegeneration. In this pilot study, we analyzed, in the brain of the Ts2Cje (Ts2) mouse model of DS, the expression of genes encoding key enzymes involved in cholesterol metabolism and of the levels of cholesterol and its main precursors and products of its metabolism (i.e., oxysterols). The results showed, in Ts2 mice compared to euploid mice, the downregulation of the transcription of the genes encoding the enzymes 3-hydroxy-3-methylglutaryl-CoA reductase and 24-dehydrocholesterol reductase, the latter originally recognized as an indicator of AD, and the consequent reduction in total cholesterol levels. Moreover, the expression of genes encoding enzymes responsible for brain cholesterol oxidation and the amounts of the resulting oxysterols were modified in Ts2 mouse brains, and the levels of cholesterol autoxidation products were increased, suggesting an exacerbation of cerebral oxidative stress. We also observed an enhanced inflammatory response in Ts2 mice, underlined by the upregulation of the transcription of the genes encoding for α-interferon and interleukin-6, two cytokines whose synthesis is increased in the brains of AD patients. Overall, these results suggest that DS and AD brains share cholesterol cycle derangements and altered oxysterol levels, which may contribute to the oxidative and inflammatory events involved in both diseases.
Collapse
Affiliation(s)
- Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20832 Desio, Italy;
| | - Rebecca Cecci
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Lucrezia Floro
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Marzia Perluigi
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University, 00185 Roma, Italy; (E.B.); (M.P.)
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (E.S.); (R.C.); (L.F.); (S.G.); (G.L.); (B.S.); (P.G.)
| |
Collapse
|
20
|
Sarangi S, Minaeva O, Ledoux DM, Parsons DS, Moncaster JA, Black CA, Hollander J, Tripodis Y, Clark JI, Hunter DG, Goldstein LE. In vivo quasi-elastic light scattering detects molecular changes in the lenses of adolescents with Down syndrome. Exp Eye Res 2024; 241:109818. [PMID: 38422787 DOI: 10.1016/j.exer.2024.109818] [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: 05/24/2023] [Revised: 12/08/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
Down syndrome (DS) is the most common chromosomal disorder in humans. DS is associated with increased prevalence of several ocular sequelae, including characteristic blue-dot cerulean cataract. DS is accompanied by age-dependent accumulation of Alzheimer's disease (AD) amyloid-β (Aβ) peptides and amyloid pathology in the brain and comorbid early-onset Aβ amyloidopathy and colocalizing cataracts in the lens. Quasi-elastic light scattering (QLS) is an established optical technique that noninvasively measures changes in protein size distributions in the human lens in vivo. In this cross-sectional study, lenticular QLS correlation time was decreased in adolescent subjects with DS compared to age-matched control subjects. Clinical QLS was consistent with alterations in relative particle hydrodynamic radius in lenses of adolescents with DS. These correlative results suggest that noninvasive QLS can be used to evaluate molecular changes in the lenses of individuals with DS.
Collapse
Affiliation(s)
- Srikant Sarangi
- Molecular Aging & Development Laboratory, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA; Boston University Photonics Center, Boston University, Boston, MA, USA; Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Olga Minaeva
- Molecular Aging & Development Laboratory, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA; Boston University Photonics Center, Boston University, Boston, MA, USA; Department of Biomedical Engineering, Boston University, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
| | - Danielle M Ledoux
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Douglas S Parsons
- Molecular Aging & Development Laboratory, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA; Boston University Photonics Center, Boston University, Boston, MA, USA
| | - Juliet A Moncaster
- Molecular Aging & Development Laboratory, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA; Boston University Photonics Center, Boston University, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Caitlin A Black
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
| | - Jeffrey Hollander
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - John I Clark
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - David G Hunter
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Lee E Goldstein
- Molecular Aging & Development Laboratory, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA; Boston University Photonics Center, Boston University, Boston, MA, USA; Department of Biomedical Engineering, Boston University, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
21
|
Schworer EK, Handen BL, Petersen M, O'Bryant S, Peven JC, Tudorascu DL, Lee L, Krinsky‐McHale SJ, Hom CL, Clare ICH, Christian BT, Schupf N, Lee JH, Head E, Mapstone M, Lott I, Ances BM, Zaman S, Brickman AM, Lai F, Rosas HD, Hartley SL. Cognitive and functional performance and plasma biomarkers of early Alzheimer's disease in Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12582. [PMID: 38623384 PMCID: PMC11016818 DOI: 10.1002/dad2.12582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION People with Down syndrome (DS) have a 75% to 90% lifetime risk of Alzheimer's disease (AD). AD pathology begins a decade or more prior to onset of clinical AD dementia in people with DS. It is not clear if plasma biomarkers of AD pathology are correlated with early cognitive and functional impairments in DS, and if these biomarkers could be used to track the early stages of AD in DS or to inform inclusion criteria for clinical AD treatment trials. METHODS This large cross-sectional cohort study investigated the associations between plasma biomarkers of amyloid beta (Aβ)42/40, total tau, and neurofilament light chain (NfL) and cognitive (episodic memory, visual-motor integration, and visuospatial abilities) and functional (adaptive behavior) impairments in 260 adults with DS without dementia (aged 25-81 years). RESULTS In general linear models lower plasma Aβ42/40 was related to lower visuospatial ability, higher total tau was related to lower episodic memory, and higher NfL was related to lower visuospatial ability and lower episodic memory. DISCUSSION Plasma biomarkers may have utility in tracking AD pathology associated with early stages of cognitive decline in adults with DS, although associations were modest. Highlights Plasma Alzheimer's disease (AD) biomarkers correlate with cognition prior to dementia in Down syndrome.Lower plasma amyloid beta 42/40 was related to lower visuospatial abilities.Higher plasma total tau and neurofilament light chain were associated with lower cognitive performance.Plasma biomarkers show potential for tracking early stages of AD symptomology.
Collapse
Affiliation(s)
| | - Benjamin L. Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Melissa Petersen
- Department of Family Medicine and Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Sid O'Bryant
- Department of Family Medicine and Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Jamie C. Peven
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dana L. Tudorascu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Laisze Lee
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Sharon J. Krinsky‐McHale
- New York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Christy L. Hom
- Department of Psychiatry and Human BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | | | | | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainSergievsky Centerand Department of NeurologyVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Joseph H. Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainSergievsky Centerand Department of NeurologyVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Elizabeth Head
- Department of Pathology & Laboratory MedicineUniversity of CaliforniaIrvine School of MedicineIrvineCaliforniaUSA
| | - Mark Mapstone
- Department of NeurologyUniversity of CaliforniaIrvine School of MedicineIrvineCaliforniaUSA
| | - Ira Lott
- Department of NeurologyUniversity of CaliforniaIrvine School of MedicineIrvineCaliforniaUSA
| | - Beau M. Ances
- Department of NeurologyWashington University at St. LouisSt. LouisMissouriUSA
| | - Shahid Zaman
- Department of PsychiatryUniversity of CambridgeCambridgeUK
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainSergievsky Centerand Department of NeurologyVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Florence Lai
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - H. Diana Rosas
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Center for Neuro‐imaging of Aging and Neurodegenerative DiseasesMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Sigan L. Hartley
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- School of Human EcologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | |
Collapse
|
22
|
Yazdi MK, Alavi MS, Roohbakhsh A. The role of ATP-binding cassette transporter G1 (ABCG1) in Alzheimer's disease: A review of the mechanisms. Basic Clin Pharmacol Toxicol 2024; 134:423-438. [PMID: 38275217 DOI: 10.1111/bcpt.13981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
The maintenance of cholesterol homeostasis is essential for central nervous system function. Consequently, factors that affect cholesterol homeostasis are linked to neurological disorders and pathologies. Among them, ATP-binding cassette transporter G1 (ABCG1) plays a significant role in atherosclerosis. However, its role in Alzheimer's disease (AD) is unclear. There is inconsistent information regarding ABCG1's role in AD. It can increase or decrease amyloid β (Aβ) levels in animals' brains. Clinical studies show that ABCG1 is involved in AD patients' impairment of cholesterol efflux capacity (CEC) in the cerebrospinal fluid (CSF). Lower Aβ levels in the CSF are correlated with ABCG1-mediated CEC dysfunction. ABCG1 modulates α-, β-, and γ-secretase activities in the plasma membrane and may affect Aβ production in the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM) cell compartment. Despite contradictory findings regarding ABCG1's role in AD, this review shows that ABCG1 has a role in Aβ generation via modulation of membrane secretases. It is, however, necessary to investigate the underlying mechanism(s). ABCG1 may also contribute to AD pathology through its role in apoptosis and oxidative stress. As a result, ABCG1 plays a role in AD and is a candidate for drug development.
Collapse
Affiliation(s)
- Mohsen Karbasi Yazdi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Sukreet S, Rafii MS, Rissman RA. From understanding to action: Exploring molecular connections of Down syndrome to Alzheimer's disease for targeted therapeutic approach. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12580. [PMID: 38623383 PMCID: PMC11016820 DOI: 10.1002/dad2.12580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/17/2024]
Abstract
Down syndrome (DS) is caused by a third copy of chromosome 21. Alzheimer's disease (AD) is a neurodegenerative condition characterized by the deposition of amyloid-beta (Aβ) plaques and neurofibrillary tangles in the brain. Both disorders have elevated Aβ, tau, dysregulated immune response, and inflammation. In people with DS, Hsa21 genes like APP and DYRK1A are overexpressed, causing an accumulation of amyloid and neurofibrillary tangles, and potentially contributing to an increased risk of AD. As a result, people with DS are a key demographic for research into AD therapeutics and prevention. The molecular links between DS and AD shed insights into the underlying causes of both diseases and highlight potential therapeutic targets. Also, using biomarkers for early diagnosis and treatment monitoring is an active area of research, and genetic screening for high-risk individuals may enable earlier intervention. Finally, the fundamental mechanistic parallels between DS and AD emphasize the necessity for continued research into effective treatments and prevention measures for DS patients at risk for AD. Genetic screening with customized therapy approaches may help the DS population in current clinical studies and future biomarkers.
Collapse
Affiliation(s)
- Sonal Sukreet
- Department of NeurosciencesUniversity of California‐San DiegoLa JollaCaliforniaUSA
| | - Michael S. Rafii
- Department of Neurology, Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Robert A. Rissman
- Department of NeurosciencesUniversity of California‐San DiegoLa JollaCaliforniaUSA
- Department Physiology and Neuroscience, Alzheimer’s Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| |
Collapse
|
24
|
Jin M, Ma Z, Dang R, Zhang H, Kim R, Xue H, Pascual J, Finkbeiner S, Head E, Liu Y, Jiang P. A Trisomy 21-linked Hematopoietic Gene Variant in Microglia Confers Resilience in Human iPSC Models of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584646. [PMID: 38559257 PMCID: PMC10979994 DOI: 10.1101/2024.03.12.584646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
While challenging, identifying individuals displaying resilience to Alzheimer's disease (AD) and understanding the underlying mechanism holds great promise for the development of new therapeutic interventions to effectively treat AD. Down syndrome (DS), or trisomy 21, is the most common genetic cause of AD. Interestingly, some people with DS, despite developing AD neuropathology, show resilience to cognitive decline. Furthermore, DS individuals are at an increased risk of myeloid leukemia due to somatic mutations in hematopoietic cells. Recent studies indicate that somatic mutations in hematopoietic cells may lead to resilience to neurodegeneration. Microglia, derived from hematopoietic lineages, play a central role in AD etiology. We therefore hypothesize that microglia carrying the somatic mutations associated with DS myeloid leukemia may impart resilience to AD. Using CRISPR-Cas9 gene editing, we introduce a trisomy 21-linked hotspot CSF2RB A455D mutation into human pluripotent stem cell (hPSC) lines derived from both DS and healthy individuals. Employing hPSC-based in vitro microglia culture and in vivo human microglia chimeric mouse brain models, we show that in response to pathological tau, the CSF2RB A455D mutation suppresses microglial type-1 interferon signaling, independent of trisomy 21 genetic background. This mutation reduces neuroinflammation and enhances phagocytic and autophagic functions, thereby ameliorating senescent and dystrophic phenotypes in human microglia. Moreover, the CSF2RB A455D mutation promotes the development of a unique microglia subcluster with tissue repair properties. Importantly, human microglia carrying CSF2RB A455D provide protection to neuronal function, such as neurogenesis and synaptic plasticity in chimeric mouse brains where human microglia largely repopulate the hippocampus. When co-transplanted into the same mouse brains, human microglia with CSF2RB A455D mutation phagocytize and replace human microglia carrying the wildtype CSF2RB gene following pathological tau treatment. Our findings suggest that hPSC-derived CSF2RB A455D microglia could be employed to develop effective microglial replacement therapy for AD and other age-related neurodegenerative diseases, even without the need to deplete endogenous diseased microglia prior to cell transplantation.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Rui Dang
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Haiwei Zhang
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Rachael Kim
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| | - Haipeng Xue
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Jesse Pascual
- Department of Pathology and Laboratory Medicine, Department of Neurology, University of California, Irvine, CA 92697, USA
| | - Steven Finkbeiner
- Ceter for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes; University of California, San Francisco, CA 94158, USA
- Departments of Neurology and Physiology, University of California, San Francisco, CA 94158, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, Department of Neurology, University of California, Irvine, CA 92697, USA
| | - Ying Liu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University New Brunswick, Piscataway, NJ 08854, USA
| |
Collapse
|
25
|
Boer PH. Functional fitness of adults with Down syndrome: a longitudinal study. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2024; 68:237-247. [PMID: 37964712 DOI: 10.1111/jir.13107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Individuals with Down syndrome (DS) are born with and develop many health-related complications. The purpose of this study was to determine the longitudinal functional fitness profile of adults with DS. METHODS The functional fitness of adults with DS was tested twice, 12 years apart. Sixty-six adults with DS were tested for body mass, stature and 10 functional fitness tests. Data were categorised according to gender and age-specific categories. RESULTS Static balance, shoulder flexibility, trunk strength and aerobic capacity deteriorated significantly with medium to large effect sizes for both DS men and women (most age categories). For women, dynamic balance deteriorated significantly, and for men, leg- and upper body-strength deteriorated significantly. CONCLUSIONS Practitioners working in the field of adapted physical activity should take cognisance of the functional fitness ageing profile of adults with DS and timeously develop habitual physical activity interventions to reduce the effect of accelerated ageing experienced by this population.
Collapse
Affiliation(s)
- P H Boer
- Department of Human Movement Science, Cape Peninsula University of Technology, Wellington, South Africa
| |
Collapse
|
26
|
Young AL, Oxtoby NP, Garbarino S, Fox NC, Barkhof F, Schott JM, Alexander DC. Data-driven modelling of neurodegenerative disease progression: thinking outside the black box. Nat Rev Neurosci 2024; 25:111-130. [PMID: 38191721 DOI: 10.1038/s41583-023-00779-6] [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] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
Data-driven disease progression models are an emerging set of computational tools that reconstruct disease timelines for long-term chronic diseases, providing unique insights into disease processes and their underlying mechanisms. Such methods combine a priori human knowledge and assumptions with large-scale data processing and parameter estimation to infer long-term disease trajectories from short-term data. In contrast to 'black box' machine learning tools, data-driven disease progression models typically require fewer data and are inherently interpretable, thereby aiding disease understanding in addition to enabling classification, prediction and stratification. In this Review, we place the current landscape of data-driven disease progression models in a general framework and discuss their enhanced utility for constructing a disease timeline compared with wider machine learning tools that construct static disease profiles. We review the insights they have enabled across multiple neurodegenerative diseases, notably Alzheimer disease, for applications such as determining temporal trajectories of disease biomarkers, testing hypotheses about disease mechanisms and uncovering disease subtypes. We outline key areas for technological development and translation to a broader range of neuroscience and non-neuroscience applications. Finally, we discuss potential pathways and barriers to integrating disease progression models into clinical practice and trial settings.
Collapse
Affiliation(s)
- Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Sara Garbarino
- Life Science Computational Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|
27
|
Sánchez Moreno B, Adán-Lirola L, Rubio-Serrano J, Real de Asúa D. Causes of mortality among adults with Down syndrome before and after the COVID-19 pandemic in Spain. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2024; 68:128-139. [PMID: 37779228 DOI: 10.1111/jir.13096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND The life expectancy of people with Down syndrome (DS) is limited by Alzheimer's disease (AD)-related deaths, mainly due to respiratory infections. The emergence of the COVID-19 pandemic could have changed known, past trends in mortality in this population. We analysed the differences in causes of mortality between individuals with DS deceased before and after the onset of the pandemic. METHOD This is a cross-sectional study of adults with DS recruited at a tertiary, university outpatient clinic in Madrid, Spain. Demographic and clinical data were retrospectively collected from their medical records, including information on their deaths, if any. RESULTS Five hundred seventy-two adults were included in the study, and 67 (11.7%) died. The main cause of death was respiratory infections, which occurred in 36 participants [9 (45.0%) before, and 27 (58.7%) after the appearance of COVID-19]. No significant differences were found in the determinants of pre-pandemic and post-pandemic death after adjusting for age and AD, except for an association between the use of psychotropic medication and death in the post-pandemic period (odds ratio: 2.24; 95% confidence interval: 1.04-4.82). Vaccination against COVID-19 showed a marked protective effect against mortality (odds ratio: 0.0002; 95% confidence interval: 6.7e10-6 to 0.004). CONCLUSIONS The appearance of COVID-19 has not impacted the overall trend of increase in mean age of death of adults with DS in our milieu, probably thanks to the very important protective effect of vaccination, which supports prioritising people with DS in future immunisation campaigns. The association between psychotropic medication use and mortality requires further exploration.
Collapse
Affiliation(s)
- Beatriz Sánchez Moreno
- Department of Internal Medicine, Adult Down Syndrome Unit, Hospital Universitario de La Princesa, Madrid, Spain
- Fundación de Investigación Biomédica del Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Adán-Lirola
- Department of Internal Medicine, Adult Down Syndrome Unit, Hospital Universitario de La Princesa, Madrid, Spain
- Fundación de Investigación Biomédica del Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Javier Rubio-Serrano
- Department of Internal Medicine, Adult Down Syndrome Unit, Hospital Universitario de La Princesa, Madrid, Spain
- Fundación de Investigación Biomédica del Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Diego Real de Asúa
- Department of Internal Medicine, Adult Down Syndrome Unit, Hospital Universitario de La Princesa, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
28
|
Gorijala P, Aslam MM, Dang LT, Xicota L, Fernandez MV, Sung YJ, Fan K, Feingold E, Surace EI, Chhatwal JP, Hom CL, Hartley SL, Hassenstab J, Perrin RJ, Mapstone M, Zaman SH, Ances BM, Kamboh MI, Lee JH, Cruchaga C. Alzheimer's polygenic risk scores are associated with cognitive phenotypes in Down syndrome. Alzheimers Dement 2024; 20:1038-1049. [PMID: 37855447 PMCID: PMC10916941 DOI: 10.1002/alz.13506] [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: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION This study aimed to investigate the influence of the overall Alzheimer's disease (AD) genetic architecture on Down syndrome (DS) status, cognitive measures, and cerebrospinal fluid (CSF) biomarkers. METHODS AD polygenic risk scores (PRS) were tested for association with DS-related traits. RESULTS The AD risk PRS was associated with disease status in several cohorts of sporadic late- and early-onset and familial late-onset AD, but not in familial early-onset AD or DS. On the other hand, lower DS Mental Status Examination memory scores were associated with higher PRS, independent of intellectual disability and APOE (PRS including APOE, PRSAPOE , p = 2.84 × 10-4 ; PRS excluding APOE, PRSnonAPOE , p = 1.60 × 10-2 ). PRSAPOE exhibited significant associations with Aβ42, tTau, pTau, and Aβ42/40 ratio in DS. DISCUSSION These data indicate that the AD genetic architecture influences cognitive and CSF phenotypes in DS adults, supporting common pathways that influence memory decline in both traits. HIGHLIGHTS Examination of the polygenic risk of AD in DS presented here is the first of its kind. AD PRS influences memory aspects in DS individuals, independently of APOE genotype. These results point to an overlap between the genes and pathways that leads to AD and those that influence dementia and memory decline in the DS population. APOE ε4 is linked to DS cognitive decline, expanding cognitive insights in adults.
Collapse
Affiliation(s)
- Priyanka Gorijala
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - M. Muaaz Aslam
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Lam‐Ha T. Dang
- Department of EpidemiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Sergievsky CenterTaub Institute for Research on Alzheimer's Disease and the Aging Brainand Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - L. Xicota
- Sergievsky CenterTaub Institute for Research on Alzheimer's Disease and the Aging Brainand Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Maria V. Fernandez
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Yun Ju Sung
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMissouriUSA
| | - Kang‐Hsien Fan
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Eleanor Feingold
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Ezequiel I. Surace
- Laboratory of Neurodegenerative Diseases ‐ Institute of Neurosciences (INEU‐Fleni‐ CONICET)Buenos AiresArgentina
| | - Jasmeer P Chhatwal
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Christy L. Hom
- Dept. of Psychiatry and Human BehaviorUniversity of CaliforniaIrvine School of MedicineCaliforniaUSA
| | | | | | - Sigan L. Hartley
- Waisman Center and School of Human EcologyUniversity of Wisconsin‐ MadisonMadisonWisconsinUSA
| | - Jason Hassenstab
- Department of Neurology and Psychological & Brain SciencesWashington UniversitySt. LouisMissouriUSA
| | - Richard J. Perrin
- Hope Center for Neurologic DiseasesWashington UniversitySt. LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Mark Mapstone
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Shahid H Zaman
- Cambridge Intellectual and Developmental Disabilities Research GroupDepartment of PsychiatryUniversity of CambridgeDouglas HouseCambridgeUK
- Cambridgeshire and Peterborough NHS Foundation TrustElizabeth HouseFulbourn HospitalFulbournCambridgeUK
| | - Beau M Ances
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - M. Ilyas Kamboh
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Joseph H Lee
- Department of EpidemiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Sergievsky CenterTaub Institute for Research on Alzheimer's Disease and the Aging Brainand Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Hope Center for Neurologic DiseasesWashington UniversitySt. LouisMissouriUSA
| |
Collapse
|
29
|
Hartley SL, Handen B, Tudorascu D, Lee L, Cohen A, Schworer EK, Peven JC, Zammit M, Klunk W, Laymon C, Minhas D, Luo W, Zaman S, Ances B, Preboske G, Christian BT. AT(N) biomarker profiles and Alzheimer's disease symptomology in Down syndrome. Alzheimers Dement 2024; 20:366-375. [PMID: 37641428 PMCID: PMC10840615 DOI: 10.1002/alz.13446] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Down syndrome (DS) is a genetic cause of early-onset Alzheimer's disease (AD). The National Institute on Aging-Alzheimer's Association AT(N) Research Framework is a staging model for AD biomarkers but has not been assessed in DS. METHOD Data are from the Alzheimer's Biomarker Consortium-Down Syndrome. Positron emission tomography (PET) amyloid beta (Aβ; 15 mCi of [11 C]Pittsburgh compound B) and tau (10 mCi of [18 F]AV-1451) were used to classify amyloid (A) -/+ and tau (T) +/-. Hippocampal volume classified neurodegeneration (N) -/+. The modified Cued Recall Test assessed episodic memory. RESULTS Analyses included 162 adults with DS (aged M = 38.84 years, standard deviation = 8.41). Overall, 69.8% of participants were classified as A-/T-/(N)-, 11.1% were A+/T-/(N)-, 5.6% were A+/T+/(N)-, and 9.3% were A+/T+/(N)+. Participants deemed cognitively stable were most likely to be A-T-(N)- and A+T-(N)-. Tau PET (T+) most closely aligning with memory impairment and AD clinical status. DISCUSSION Findings add to understanding of AT(N) biomarker profiles in DS. HIGHLIGHTS Overall, 69.8% of adults with Down syndrome (DS) aged 25 to 61 years were classified as amyloid (A)-/tau (T)-/neurodegeneration (N)-, 11.1% were A+/T-/(N)-, 5.6% were A+/T+/(N)-, and 9.3% were A+/T+/(N)+. The AT(N) profiles were associated with clinical Alzheimer's disease (AD) status and with memory performance, with the presence of T+ aligned with AD clinical symptomology. Findings inform models for predicting the transition to the prodromal stage of AD in DS.
Collapse
Affiliation(s)
- Sigan L. Hartley
- Waisman CenterUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- School of Human EcologyUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Benjamin Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dana Tudorascu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Laisze Lee
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Annie Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Jamie C. Peven
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Matthew Zammit
- Waisman CenterUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - William Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Charles Laymon
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of BioengineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Davneet Minhas
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Weiquan Luo
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Shahid Zaman
- Department of PsychiatryUniversity of CambridgeCambridgeUK
| | - Beau Ances
- Department of NeurologyWashington University at St. LouisSt. Louis, MissouriUSA
| | | | - Bradley T. Christian
- Waisman CenterUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | | |
Collapse
|
30
|
Sharma C, Mazumder A. A Comprehensive Review on Potential Molecular Drug Targets for the Management of Alzheimer's Disease. Cent Nerv Syst Agents Med Chem 2024; 24:45-56. [PMID: 38305393 DOI: 10.2174/0118715249263300231116062740] [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: 06/20/2023] [Revised: 08/25/2023] [Accepted: 10/04/2023] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is an onset and incurable neurodegenerative disorder that has been linked to various genetic, environmental, and lifestyle factors. Recent research has revealed several potential targets for drug development, such as the prevention of Aβ production and removal, prevention of tau hyperphosphorylation, and keeping neurons alive. Drugs that target numerous ADrelated variables have been developed, and early results are encouraging. This review provides a concise map of the different receptor signaling pathways associated with Alzheimer's Disease, as well as insight into drug design based on these pathways. It discusses the molecular mechanisms of AD pathogenesis, such as oxidative stress, aging, Aβ turnover, thiol groups, and mitochondrial activities, and their role in the disease. It also reviews the potential drug targets, in vivo active agents, and docking studies done in AD and provides prospects for future drug development. This review intends to provide more clarity on the molecular processes that occur in Alzheimer's patient's brains, which can be of use in diagnosing and preventing the condition.
Collapse
Affiliation(s)
- Chanchal Sharma
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida-201306, Uttar Pradesh, India
| |
Collapse
|
31
|
Cruciani F, Aparo A, Brusini L, Combi C, Storti SF, Giugno R, Menegaz G, Boscolo Galazzo I. Identifying the joint signature of brain atrophy and gene variant scores in Alzheimer's Disease. J Biomed Inform 2024; 149:104569. [PMID: 38104851 DOI: 10.1016/j.jbi.2023.104569] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The joint modeling of genetic data and brain imaging information allows for determining the pathophysiological pathways of neurodegenerative diseases such as Alzheimer's disease (AD). This task has typically been approached using mass-univariate methods that rely on a complete set of Single Nucleotide Polymorphisms (SNPs) to assess their association with selected image-derived phenotypes (IDPs). However, such methods are prone to multiple comparisons bias and, most importantly, fail to account for potential cross-feature interactions, resulting in insufficient detection of significant associations. Ways to overcome these limitations while reducing the number of traits aim at conveying genetic information at the gene level and capturing the integrated genetic effects of a set of genetic variants, rather than looking at each SNP individually. Their associations with brain IDPs are still largely unexplored in the current literature, though they can uncover new potential genetic determinants for brain modulations in the AD continuum. In this work, we explored an explainable multivariate model to analyze the genetic basis of the grey matter modulations, relying on the AD Neuroimaging Initiative (ADNI) phase 3 dataset. Cortical thicknesses and subcortical volumes derived from T1-weighted Magnetic Resonance were considered to describe the imaging phenotypes. At the same time the genetic counterpart was represented by gene variant scores extracted by the Sequence Kernel Association Test (SKAT) filtering model. Moreover, transcriptomic analysis was carried on to assess the expression of the resulting genes in the main brain structures as a form of validation. Results highlighted meaningful genotype-phenotype interactionsas defined by three latent components showing a significant difference in the projection scores between patients and controls. Among the significant associations, the model highlighted EPHX1 and BCAS1 gene variant scores involved in neurodegenerative and myelination processes, hence relevant for AD. In particular, the first was associated with decreased subcortical volumes and the second with decreasedtemporal lobe thickness. Noteworthy, BCAS1 is particularly expressed in the dentate gyrus. Overall, the proposed approach allowed capturing genotype-phenotype interactions in a restricted study cohort that was confirmed by transcriptomic analysis, offering insights into the underlying mechanisms of neurodegeneration in AD in line with previous findings and suggesting new potential disease biomarkers.
Collapse
Affiliation(s)
- Federica Cruciani
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
| | - Antonino Aparo
- Department of Computer Science, University of Verona, Verona, Italy
| | - Lorenza Brusini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Carlo Combi
- Department of Computer Science, University of Verona, Verona, Italy
| | - Silvia F Storti
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Verona, Italy
| | - Gloria Menegaz
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | | |
Collapse
|
32
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
33
|
Meijer L, Chrétien E, Ravel D. Leucettinib-21, a DYRK1A Kinase Inhibitor as Clinical Drug Candidate for Alzheimer's Disease and Down Syndrome. J Alzheimers Dis 2024; 101:S95-S113. [PMID: 39422950 DOI: 10.3233/jad-240078] [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: 10/19/2024]
Abstract
Alzheimer's disease (AD) and Down syndrome (DS) share a common therapeutic target, the dual-specificity, tyrosine phosphorylation activated kinase 1A (DYRK1A). Abnormally active DYRK1A is responsible for cognitive disorders (memory, learning, spatial localization) observed in both conditions. In DS, DYRK1A is overexpressed due to the presence of the DYRK1A gene on chromosome 21. In AD, calcium-activated calpains cleave full-length DYRK1A (FL-DYRK1A) into a more stable and more active, low molecular weight, kinase (LMW-DYRK1A). Genetic and pharmacological experiments carried out with animal models of AD and DS strongly support the idea that pharmacological inhibitors of DYRK1A might be able to correct memory/learning disorders in people with AD and DS. Starting from a marine sponge natural product, Leucettamine B, Perha Pharmaceuticals has optimized, through classical medicinal chemistry, and extensively characterized a small molecule drug candidate, Leucettinib-21. Regulatory preclinical safety studies in rats and minipigs have been completed and formulation of Leucettinib-21 has been optimized as immediate-release tablets. Leucettinib-21 is now undergoing a phase 1 clinical trial (120 participants, including 12 adults with DS and 12 patients with AD). The therapeutic potential of DYRK1A inhibitors in AD and DS is presented.
Collapse
Affiliation(s)
- Laurent Meijer
- Perha Pharmaceuticals, Hôtel de Recherche, Roscoff, Bretagne, France
| | - Emilie Chrétien
- Perha Pharmaceuticals, Hôtel de Recherche, Roscoff, Bretagne, France
| | | |
Collapse
|
34
|
Halperin SJ, Dhodapkar MM, Radford Z, Frumberg DB, Rubin LE, Grauer JN. Patients With Down Syndrome and Total Hip and Total Knee Arthroplasty: Outcome Measures Show Increased Risk of Perioperative Complications. J Am Acad Orthop Surg Glob Res Rev 2023; 7:01979360-202312000-00008. [PMID: 38054749 PMCID: PMC10697628 DOI: 10.5435/jaaosglobal-d-23-00108] [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] [Received: 06/05/2023] [Revised: 09/27/2023] [Accepted: 10/28/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Patients with Down syndrome (DS) are being considered for total joint arthroplasty. There is limited literature regarding outcomes of patients with DS after total hip arthroplasty (THA) and total knee arthroplasty (TKA). METHODS Data were abstracted from the 2010 to 2021 Q1 PearlDiver M151 database. THA and TKA osteoarthritis adult patients with and without DS were identified. Patients were matched 1:10. Ninety-day postoperative events and 2-year revision rates were compared. RESULTS For THA, 154 patients with DS were matched with 1,532 patients without DS. For TKA, 150 patients with DS were matched with 1,495 patients without DS. On multivariable logistic regression, THA and TKA patients with DS were at markedly greater odds of postoperative events including any adverse event, sepsis, minor adverse event, urinary tract infection (UTI), acute kidney injury (AKI), and pneumonia. For both THA and TKA, 2-year revision rates were not increased for those with DS. DISCUSSION This study represents the largest cohorts for matched patients with DS undergoing THA or TKA through 90 days postoperatively. For both procedures, DS patients were found to have greater risk of several adverse events, but not 2-year revisions. These findings may help guide perioperative risk assessment, patient/family counseling, and care pathways.
Collapse
Affiliation(s)
- Scott J Halperin
- From the Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, CT
| | | | | | | | | | | |
Collapse
|
35
|
Idris M, Saini F, Pape SE, Baksh RA, Cahart MS, Strydom A. Common mental health disorders and cognitive decline in a longitudinal Down syndrome cohort. BJPsych Open 2023; 9:e206. [PMID: 37920115 PMCID: PMC10753965 DOI: 10.1192/bjo.2023.590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/04/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Down syndrome is the most common genetic cause of intellectual disability and Alzheimer's disease. In the general population, common mental disorders (CMDs), including anxiety, depression and obsessive-compulsive disorder, are linked to cognitive decline and higher risk for dementia. It is not known how CMDs affect longer-term cognitive outcomes in Down syndrome, and there is often diagnostic uncertainty in older people with Down syndrome and psychiatric comorbidity. AIMS To study the influence of CMDs on cognitive ability and whether they are related longitudinally to development of clinical signs of Alzheimer's disease in Down syndrome. METHOD We followed 115 individuals with Down syndrome, 27 of whom were diagnosed with a CMD, over approximately 3 years. Changes in cognitive and behavioural outcomes between baseline and follow-up assessment were analysed, with comparisons made between those with and without a comorbid CMD. Age, gender, apolipoprotein E status and level of intellectual disability were included as covariates. RESULTS No significant association between presence of a CMD and poorer performance on cognitive tasks or informant-rated decline over time was observed (P > 0.05). CONCLUSIONS Our results suggest that a diagnosis of a CMD does not have a significant negative effect on long-term cognitive or behavioural outcomes in individuals with Down syndrome. In individuals with stable or treated CMD, subsequent cognitive decline is likely indicative of Alzheimer's disease rather than a consequence of mental disorder.
Collapse
Affiliation(s)
- Mina Idris
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Fedal Saini
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Sarah E. Pape
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - R. Asaad Baksh
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Marie-Stephanie Cahart
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - André Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| |
Collapse
|
36
|
Mgaieth F, Baksh RA, Startin CM, Hamburg S, Hithersay R, Pape S, Zetterberg H, Ashton NJ, Tamayo‐Elizalde M, Saini F, Idris M, Consortium TL, Strydom A. Exploring semantic verbal fluency patterns and their relationship to age and Alzheimer's disease in adults with Down syndrome. Alzheimers Dement 2023; 19:5129-5137. [PMID: 37114906 PMCID: PMC11497345 DOI: 10.1002/alz.13097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023]
Abstract
INTRODUCTION Adults with Down syndrome (DS) are at ultra-high risk of developing Alzheimer's disease (AD), characterized by poor episodic memory and semantic fluency in the preclinical phase in the general population. We explored semantic fluency performance in DS and its relationship to age, AD, and blood biomarkers. METHODS A total of 302 adults with DS at baseline and 87 at follow-up from the London Down Syndrome Consortium cohort completed neuropsychological assessments. Blood biomarkers were measured with the single molecule array technique in a subset of 94 participants. RESULTS Poorer verbal fluency performance was observed as age increases. Number of correct words declined in those with AD compared to those without over 2 years and was negatively correlated with neurofilament light (r = -0.37, P = .001) and glial fibrillary acidic protein (r = -0.31, P = .012). DISCUSSION Semantic fluency may be useful as an early indicator of cognitive decline and provide additional information on AD-related change, showing associations with biomarkers in DS.
Collapse
Affiliation(s)
- Farah Mgaieth
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
| | - R. Asaad Baksh
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
- The LonDownS ConsortiumLondonUK
| | - Carla M. Startin
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
- The LonDownS ConsortiumLondonUK
- Division of PsychiatryUniversity College LondonLondonUK
- School of PsychologyUniversity of RoehamptonLondonUK
| | | | - Rosalyn Hithersay
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
- The LonDownS ConsortiumLondonUK
- Division of PsychiatryUniversity College LondonLondonUK
| | - Sarah Pape
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
- South London and Maudsley NHS Foundation TrustLondonUK
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Nicholas J. Ashton
- South London and Maudsley NHS Foundation TrustLondonUK
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Institute of PsychiatryPsychology and Neuroscience Maurice Wohl Institute Clinical Neuroscience InstituteKing's College LondonLondonUK
- Centre for Age‐Related MedicineStavanger University HospitalStavangerNorway
| | - Miren Tamayo‐Elizalde
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
| | - Fedal Saini
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
| | - Mina Idris
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
| | | | - Andre Strydom
- Department of Forensic and Neurodevelopmental SciencesInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
- The LonDownS ConsortiumLondonUK
- Division of PsychiatryUniversity College LondonLondonUK
- South London and Maudsley NHS Foundation TrustLondonUK
| |
Collapse
|
37
|
Prévot V, Tena-Sempere M, Pitteloud N. New Horizons: Gonadotropin-Releasing Hormone and Cognition. J Clin Endocrinol Metab 2023; 108:2747-2758. [PMID: 37261390 DOI: 10.1210/clinem/dgad319] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/02/2023]
Abstract
Pulsatile secretion of gonadotropin-releasing hormone (GnRH) is essential for activating and maintaining the function of the hypothalamic-pituitary-gonadal axis, which controls the onset of puberty and fertility. Two recent studies suggest that, in addition to controlling reproduction, the neurons in the brain that produce GnRH are also involved in the control of postnatal brain maturation, odor discrimination, and adult cognition. This review will summarize the development and establishment of the GnRH system, with particular attention to the importance of its first postnatal activation, a phenomenon known as minipuberty, for later reproductive and nonreproductive functions. In addition, we will discuss the beneficial effects of restoring physiological (ie, pulsatile) GnRH levels on olfactory and cognitive alterations in preclinical Down syndrome and Alzheimer disease models, as well as the potential risks associated with long-term continuous (ie, nonphysiological) GnRH administration in certain disorders. Finally, this review addresses the intriguing possibility that pulsatile GnRH therapy may hold therapeutic potential for the management of some neurodevelopmental cognitive disorders and pathological aging in elderly people.
Collapse
Affiliation(s)
- Vincent Prévot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR S1172, Lille F-59000, France
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14004 Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Córdoba, Spain
| | - Nelly Pitteloud
- Department of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne 1011, Switzerland
- Faculty of Biology and Medicine, Université of Lausanne, Lausanne 1005, Switzerland
| |
Collapse
|
38
|
Dodd D, Helsel B, Bodde AE, Danon JC, Sherman JR, Donnelly JE, Washburn RA, Ptomey LT. The association of increased body mass index on cardiorespiratory fitness, physical activity, and cognition in adults with down syndrome. Disabil Health J 2023; 16:101497. [PMID: 37407386 PMCID: PMC10680094 DOI: 10.1016/j.dhjo.2023.101497] [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: 02/10/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Obesity is a significant risk factor for Alzheimer's disease; however, this association has not been explored in adults with Down syndrome. OBJECTIVE To examine the association of obesity, assessed by body mass index (BMI), with factors related to Alzheimer's disease risk including cardiorespiratory fitness, physical activity, and cognition in adults with Down syndrome. METHODS Adults with Down syndrome attended a laboratory visit where BMI, cardiorespiratory fitness (VO2 peak), and cognitive function (CANTAB® DS Battery) were obtained. Physical activity (accelerometer) was collected over the week following the laboratory visit. Wilcoxon rank sum tests were used to evaluate differences in cardiorespiratory fitness, sedentary time, moderate-to-vigorous physical activity (MVPA), and cognition between adults with obesity (BMI≥ 30 kg/m2) and those with healthy weight or overweight (BMI <30 kg/m2). Spearman correlations and linear regressions were used to measure the impact of BMI on cardiorespiratory fitness, MVPA, sedentary time, and cognition. RESULTS Data was collected for 79 adults with Down syndrome (26.7 ± 9.0 years of age, 54% female, 54% with obesity). VO2 peak was significantly lower in participants with obesity (18.4 ± 2.5 ml/kg/min) compared to those with healthy weight or overweight (22.9 ± 4.0 ml/kg/min, p < 0.001). BMI was negatively associated with cardiorespiratory fitness (rho = -0.614, p < 0.001). No associations were observed between BMI and physical activity or cognition. CONCLUSIONS Lower BMI was associated with improved cardiorespiratory fitness. However, no associations were observed between BMI and cognition or physical activity. NCT REGISTRATION NCT04048759.
Collapse
Affiliation(s)
- Danica Dodd
- School of Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA; Department of Internal Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Brian Helsel
- Department of Neurology, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Amy E Bodde
- Department of Internal Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Jessica C Danon
- Department of Internal Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Joseph R Sherman
- Department of Internal Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Joseph E Donnelly
- Department of Internal Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Richard A Washburn
- Department of Internal Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Lauren T Ptomey
- Department of Internal Medicine, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
39
|
Réthelyi JM, Vincze K, Schall D, Glennon J, Berkel S. The role of insulin/IGF1 signalling in neurodevelopmental and neuropsychiatric disorders - Evidence from human neuronal cell models. Neurosci Biobehav Rev 2023; 153:105330. [PMID: 37516219 DOI: 10.1016/j.neubiorev.2023.105330] [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: 09/30/2022] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signalling play a central role in the development and maintenance of neurons in the brain, and human neurodevelopmental as well as neuropsychiatric disorders have been linked to impaired insulin and IGF1 signalling. This review focuses on the impairments of the insulin and IGF1 signalling cascade in the context of neurodevelopmental and neuropsychiatric disorders, based on evidence from human neuronal cell models. Clear evidence was obtained for impaired insulin and IGF1 receptor downstream signalling in neurodevelopmental disorders, while the evidence for its role in neuropsychiatric disorders was less substantial. Human neuronal model systems can greatly add to our knowledge about insulin/IGF1 signalling in the brain, its role in restoring dendritic maturity, and complement results from clinical studies and animal models. Moreover, they represent a useful model for the development of new therapeutic strategies. Further research is needed to systematically investigate the exact role of the insulin/IGF1 signalling cascades in neurodevelopmental and neuropsychiatric disorders, and to elucidate the respective therapeutic implications.
Collapse
Affiliation(s)
- János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Katalin Vincze
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
| | - Dorothea Schall
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; Interdisciplinary Centre of Neurosciences (IZN), Heidelberg University, Germany.
| |
Collapse
|
40
|
Videla L, Benejam B, Carmona-Iragui M, Barroeta I, Fernández S, Arranz J, Azzahchi SE, Altuna M, Padilla C, Valldeneu S, Pegueroles J, Montal V, Aranha MR, Vaqué-Alcázar L, Iulita MF, Alcolea D, Bejanin A, Videla S, Blesa R, Lleó A, Fortea J. Cross-sectional versus longitudinal cognitive assessments for the diagnosis of symptomatic Alzheimer's disease in adults with Down syndrome. Alzheimers Dement 2023; 19:3916-3925. [PMID: 37038748 DOI: 10.1002/alz.13073] [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: 01/11/2023] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Down syndrome (DS) is a genetic form of Alzheimer's disease (AD). However, clinical diagnosis is difficult, and experts emphasize the need for detecting intra-individual cognitive decline. OBJECTIVE To compare the performance of baseline and longitudinal neuropsychological assessments for the diagnosis of symptomatic AD in DS. METHODS Longitudinal cohort study of adults with DS. Individuals were classified as asymptomatic, prodromal AD, or AD dementia. We performed receiver operating characteristic curve analyses to compare baseline and longitudinal changes of CAMCOG-DS and mCRT. RESULTS We included 562 adults with DS. Baseline assessments showed good to excellent diagnostic performance for AD dementia (AUCs between 0.82 and 0.99) and prodromal AD, higher than the 1-year intra-individual cognitive decline (area under the ROC curve between 0.59 and 0.79 for AD dementia, lower for prodromal AD). Longer follow-ups increased the diagnostic performance of the intra-individual cognitive decline. DISCUSSION Baseline cognitive assessment outperforms the 1-year intra-individual cognitive decline in adults with DS.
Collapse
Affiliation(s)
- Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Bessy Benejam
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - María Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Susana Fernández
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Javier Arranz
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sumia Elbachiri Azzahchi
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- CITA-alzheimer foundation, Donostia-San Sebsatián, Spain
| | - Concepción Padilla
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Silvia Valldeneu
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Víctor Montal
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mateus Rozalem Aranha
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lídia Vaqué-Alcázar
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Maria Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Sebastià Videla
- Clinical Research Support Unit-[HUB-IDIBELL], Clinical Pharmacology Department, Bellvitge University Hospital, University of Barcelona, Barcelona, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
41
|
Prévot V, Duittoz A. A role for GnRH in olfaction and cognition: Implications for veterinary medicine. Reprod Domest Anim 2023; 58 Suppl 2:109-124. [PMID: 37329313 DOI: 10.1111/rda.14411] [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: 04/24/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
Pulsatile secretion of gonadotropin-releasing hormone (GnRH) is essential for the activation and maintenance of the function of the hypothalamic-pituitary-gonadal (HPG) axis, which controls the onset of puberty and fertility. Two provocative recent studies suggest that, in addition to control reproduction, the neurons in the brain that produce GnRH are also involved in the control postnatal brain maturation, odour discrimination and adult cognition. Long-acting GnRH antagonists and agonists are commonly used to control fertility and behaviour in veterinary medicine, primarily in males. This review puts into perspective the potential risks of these androgen deprivation therapies and immunization on olfactory and cognitive performances and well-aging in domestic animals, including pets. We will also discuss the results reporting beneficial effects of pharmacological interventions restoring physiological GnRH levels on olfactory and cognitive alterations in preclinical models of Alzheimer's disease, which shares many pathophysiological and behavioural hallmarks with canine cognitive dysfunction. These novel findings raise the intriguing possibility that pulsatile GnRH therapy holds therapeutic potential for the management of this behavioural syndrome affecting older dogs.
Collapse
Affiliation(s)
- Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
| | - Anne Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAE Val de Loire, IFCE, Université de Tours, Nouzilly, France
| |
Collapse
|
42
|
Brackhan M, Arribas-Blazquez M, Lastres-Becker I. Aging, NRF2, and TAU: A Perfect Match for Neurodegeneration? Antioxidants (Basel) 2023; 12:1564. [PMID: 37627559 PMCID: PMC10451380 DOI: 10.3390/antiox12081564] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Although the trigger for the neurodegenerative disease process is unknown, the relevance of aging stands out as a major risk for the development of neurodegeneration. In this review, we highlighted the relationship between the different cellular mechanisms that occur as a consequence of aging and transcription factor nuclear factor erythroid-2-related factor 2 (NRF2) and the connection with the TAU protein. We focused on the relevance of NRF2 in the main processes involved in neurodegeneration and associated with aging, such as genomic instability, protein degradation systems (proteasomes/autophagy), cellular senescence, and stem cell exhaustion, as well as inflammation. We also analyzed the effect of aging on TAU protein levels and its aggregation and spread process. Finally, we investigated the interconnection between NRF2 and TAU and the relevance of alterations in the NRF2 signaling pathway in both primary and secondary tauopathies. All these points highlight NRF2 as a possible therapeutic target for tauopathies.
Collapse
Affiliation(s)
- Mirjam Brackhan
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, c/Arturo Duperier 4, 28029 Madrid, Spain
| | - Marina Arribas-Blazquez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain;
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Isabel Lastres-Becker
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, c/Arturo Duperier 4, 28029 Madrid, Spain
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- Institute Teófilo Hernando for Drug Discovery, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| |
Collapse
|
43
|
Abu-Elfotuh K, Selim HMRM, Riad OKM, Hamdan AME, Hassanin SO, Sharif AF, Moustafa NM, Gowifel AM, Mohamed MYA, Atwa AM, Zaghlool SS, El-Din MN. The protective effects of sesamol and/or the probiotic, Lactobacillus rhamnosus, against aluminum chloride-induced neurotoxicity and hepatotoxicity in rats: Modulation of Wnt/β-catenin/GSK-3β, JAK-2/STAT-3, PPAR-γ, inflammatory, and apoptotic pathways. Front Pharmacol 2023; 14:1208252. [PMID: 37601053 PMCID: PMC10436218 DOI: 10.3389/fphar.2023.1208252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction: Aluminium (Al) is accumulated in the brain causing neurotoxicity and neurodegenerative disease like Alzheimer's disease (AD), multiple sclerosis, autism and epilepsy. Hence, attenuation of Al-induced neurotoxicity has become a "hot topic" in looking for an intervention that slow down the progression of neurodegenerative diseases. Objective: Our study aims to introduce a new strategy for hampering aluminum chloride (AlCl3)-induced neurotoxicity using a combination of sesamol with the probiotic bacteria; Lactobacillus rhamnosus (L. rhamnosus) and also to test their possible ameliorative effects on AlCl3-induced hepatotoxicity. Methods: Sprague-Dawley male rats were randomly divided into five groups (n = 10/group) which are control, AlCl3, AlCl3 + Sesamol, AlCl3 + L. rhamnosus and AlCl3 + Sesamol + L. rhamnosus. We surveilled the behavioral, biochemical, and histopathological alterations centrally in the brain and peripherally in liver. Results: This work revealed that the combined therapy of sesamol and L. rhamnosus produced marked reduction in brain amyloid-β, p-tau, GSK-3β, inflammatory and apoptotic biomarkers, along with marked elevation in brain free β-catenin and Wnt3a, compared to AlCl3-intoxicated rats. Also, the combined therapy exerted pronounced reduction in hepatic expressions of JAK-2/STAT-3, inflammatory (TNF-α, IL-6, NF-κB), fibrotic (MMP-2, TIMP-1, α-SMA) and apoptotic markers, (caspase-3), together with marked elevation in hepatic PPAR-γ expression, compared to AlCl3 -intoxicated rats. Behavioral and histopathological assessments substantiated the efficiency of this combined regimen in halting the effect of neurotoxicity. Discussion: Probiotics can be used as an add-on therapy with sesamol ameliorate AlCl3 -mediated neurotoxicity and hepatotoxicity.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Clinical Pharmacy Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Heba Mohammed Refat M. Selim
- Pharmaceutical Sciences Department, Faculty of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
- Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Omnia Karem M. Riad
- Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ahmed M. E. Hamdan
- Pharmacy Practice Department, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Soha Osama Hassanin
- Biochemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Asmaa F. Sharif
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
- Clinical Medical Sciences Department, College of Medicine, Dar Al Uloom University, Riyadh, Saudi Arabia
| | - Nouran Magdy Moustafa
- Basic Medical Science Department, College of Medicine, Dar Al Uloom University, Riyadh, Saudi Arabia
- Medical Microbiology and Immunology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ayah M.H. Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Marwa Y. A. Mohamed
- Biology Department, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Ahmed M. Atwa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Sameh S. Zaghlool
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Mahmoud Nour El-Din
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), Menoufia, Egypt
| |
Collapse
|
44
|
Murray A, Gough G, Cindrić A, Vučković F, Koschut D, Borelli V, Petrović DJ, Bekavac A, Plećaš A, Hribljan V, Brunmeir R, Jurić J, Pučić-Baković M, Slana A, Deriš H, Frkatović A, Groet J, O'Brien NL, Chen HY, Yeap YJ, Delom F, Havlicek S, Gammon L, Hamburg S, Startin C, D'Souza H, Mitrečić D, Kero M, Odak L, Krušlin B, Krsnik Ž, Kostović I, Foo JN, Loh YH, Dunn NR, de la Luna S, Spector T, Barišić I, Thomas MSC, Strydom A, Franceschi C, Lauc G, Krištić J, Alić I, Nižetić D. Dose imbalance of DYRK1A kinase causes systemic progeroid status in Down syndrome by increasing the un-repaired DNA damage and reducing LaminB1 levels. EBioMedicine 2023; 94:104692. [PMID: 37451904 PMCID: PMC10435767 DOI: 10.1016/j.ebiom.2023.104692] [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: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND People with Down syndrome (DS) show clinical signs of accelerated ageing. Causative mechanisms remain unknown and hypotheses range from the (essentially untreatable) amplified-chromosomal-instability explanation, to potential actions of individual supernumerary chromosome-21 genes. The latter explanation could open a route to therapeutic amelioration if the specific over-acting genes could be identified and their action toned-down. METHODS Biological age was estimated through patterns of sugar molecules attached to plasma immunoglobulin-G (IgG-glycans, an established "biological-ageing-clock") in n = 246 individuals with DS from three European populations, clinically characterised for the presence of co-morbidities, and compared to n = 256 age-, sex- and demography-matched healthy controls. Isogenic human induced pluripotent stem cell (hiPSCs) models of full and partial trisomy-21 with CRISPR-Cas9 gene editing and two kinase inhibitors were studied prior and after differentiation to cerebral organoids. FINDINGS Biological age in adults with DS is (on average) 18.4-19.1 years older than in chronological-age-matched controls independent of co-morbidities, and this shift remains constant throughout lifespan. Changes are detectable from early childhood, and do not require a supernumerary chromosome, but are seen in segmental duplication of only 31 genes, along with increased DNA damage and decreased levels of LaminB1 in nucleated blood cells. We demonstrate that these cell-autonomous phenotypes can be gene-dose-modelled and pharmacologically corrected in hiPSCs and derived cerebral organoids. Using isogenic hiPSC models we show that chromosome-21 gene DYRK1A overdose is sufficient and necessary to cause excess unrepaired DNA damage. INTERPRETATION Explanation of hitherto observed accelerated ageing in DS as a developmental progeroid syndrome driven by DYRK1A overdose provides a target for early pharmacological preventative intervention strategies. FUNDING Main funding came from the "Research Cooperability" Program of the Croatian Science Foundation funded by the European Union from the European Social Fund under the Operational Programme Efficient Human Resources 2014-2020, Project PZS-2019-02-4277, and the Wellcome Trust Grants 098330/Z/12/Z and 217199/Z/19/Z (UK). All other funding is described in details in the "Acknowledgements".
Collapse
Affiliation(s)
- Aoife Murray
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK.
| | - Gillian Gough
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Ana Cindrić
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Frano Vučković
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - David Koschut
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cellular Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Vincenzo Borelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy
| | - Dražen J Petrović
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Bekavac
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ante Plećaš
- Faculty of Veterinary Medicine, Department of Anatomy, Histology and Embryology, University of Zagreb, Zagreb, Croatia
| | - Valentina Hribljan
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Reinhard Brunmeir
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Julija Jurić
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | | | - Anita Slana
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Helena Deriš
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Azra Frkatović
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Jűrgen Groet
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK
| | - Niamh L O'Brien
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK
| | - Hong Yu Chen
- Institute of Molecular and Cell Biology (IMCB), A∗STAR, Singapore
| | - Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Frederic Delom
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Steven Havlicek
- Laboratory of Neurogenetics, Genome Institute of Singapore, A∗STAR, Singapore
| | - Luke Gammon
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Sarah Hamburg
- The London Down Syndrome Consortium (LonDownS), London, UK
| | - Carla Startin
- The London Down Syndrome Consortium (LonDownS), London, UK; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Division of Psychiatry, University College London, London, UK; School of Psychology, University of Roehampton, London, UK
| | - Hana D'Souza
- The London Down Syndrome Consortium (LonDownS), London, UK; Centre for Brain and Cognitive Development, Birkbeck, University of London, London, UK
| | - Dinko Mitrečić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mijana Kero
- Department of Medical Genetics, Children's Hospital Zagreb, Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ljubica Odak
- Department of Medical Genetics, Children's Hospital Zagreb, Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Božo Krušlin
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Laboratory of Neurogenetics, Genome Institute of Singapore, A∗STAR, Singapore
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), A∗STAR, Singapore
| | - Norris Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Institute of Molecular and Cell Biology (IMCB), A∗STAR, Singapore
| | - Susana de la Luna
- ICREA, Genome Biology Programme (CRG), Universitat Pompeu Fabra (UPF), CIBER of Rare Diseases, Barcelona, Spain
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Ingeborg Barišić
- Department of Medical Genetics, Children's Hospital Zagreb, Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Michael S C Thomas
- The London Down Syndrome Consortium (LonDownS), London, UK; Centre for Brain and Cognitive Development, Birkbeck, University of London, London, UK
| | - Andre Strydom
- The London Down Syndrome Consortium (LonDownS), London, UK; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Division of Psychiatry, University College London, London, UK
| | - Claudio Franceschi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy; Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| | - Gordan Lauc
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia; Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | | | - Ivan Alić
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; Faculty of Veterinary Medicine, Department of Anatomy, Histology and Embryology, University of Zagreb, Zagreb, Croatia.
| | - Dean Nižetić
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| |
Collapse
|
45
|
Holloway K, Neherin K, Dam KU, Zhang H. Cellular senescence and neurodegeneration. Hum Genet 2023; 142:1247-1262. [PMID: 37115318 DOI: 10.1007/s00439-023-02565-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Advancing age is a major risk factor of Alzheimer's disease (AD). The worldwide prevalence of AD is approximately 50 million people, and this number is projected to increase substantially. The molecular mechanisms underlying the aging-associated susceptibility to cognitive impairment in AD are largely unknown. As a hallmark of aging, cellular senescence is a significant contributor to aging and age-related diseases including AD. Senescent neurons and glial cells have been detected to accumulate in the brains of AD patients and mouse models. Importantly, selective elimination of senescent cells ameliorates amyloid beta and tau pathologies and improves cognition in AD mouse models, indicating a critical role of cellular senescence in AD pathogenesis. Nonetheless, the mechanisms underlying when and how cellular senescence contributes to AD pathogenesis remain unclear. This review provides an overview of cellular senescence and discusses recent advances in the understanding of the impact of cellular senescence on AD pathogenesis, with brief discussions of the possible role of cellular senescence in other neurodegenerative diseases including Down syndrome, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Kristopher Holloway
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Kashfia Neherin
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Kha Uyen Dam
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Hong Zhang
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA.
| |
Collapse
|
46
|
Chen Y, Xiao Y, Zhang Y, Wang R, Wang F, Gao H, Liu Y, Zhang R, Sun H, Zhou Z, Wang S, Chen K, Sun Y, Tu M, Li J, Luo Q, Wu Y, Zhu L, Huang Y, Sun X, Guo G, Zhang D. Single-cell landscape analysis reveals systematic senescence in mammalian Down syndrome. Clin Transl Med 2023; 13:e1310. [PMID: 37461266 PMCID: PMC10352595 DOI: 10.1002/ctm2.1310] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/28/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Down syndrome (DS), which is characterized by various malfunctions, is the most common chromosomal disorder. As the DS population continues to grow and most of those with DS live beyond puberty, early-onset health problems have become apparent. However, the cellular landscape and molecular alterations have not been thoroughly studied. METHODS This study utilized single-cell resolution techniques to examine DS in humans and mice, spanning seven distinct organs. A total of 71 934 mouse and 98 207 human cells were analyzed to uncover the molecular alterations occurring in different cell types and organs related to DS, specifically starting from the fetal stage. Additionally, SA-β-Gal staining, western blot, and histological study were employed to verify the alterations. RESULTS In this study, we firstly established the transcriptomic profile of the mammalian DS, deciphering the cellular map and molecular mechanism. Our analysis indicated that DS cells across various types and organs experienced senescence stresses from as early as the fetal stage. This was marked by elevated SA-β-Gal activity, overexpression of cell cycle inhibitors, augmented inflammatory responses, and a loss of cellular identity. Furthermore, we found evidence of mitochondrial disturbance, an increase in ribosomal protein transcription, and heightened apoptosis in fetal DS cells. This investigation also unearthed a regulatory network driven by an HSA21 gene, which leads to genome-wide expression changes. CONCLUSION The findings from this study offer significant insights into the molecular alterations that occur in DS, shedding light on the pathological processes underlying this disorder. These results can potentially guide future research and treatment development for DS.
Collapse
Affiliation(s)
- Yao Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanyu Xiao
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanye Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Renying Wang
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Feixia Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huajing Gao
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifeng Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runju Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiyu Sun
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziming Zhou
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Siwen Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yixi Sun
- Department of Reproductive Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mixue Tu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyi Li
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Qiong Luo
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Yiqing Wu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Linling Zhu
- Department of Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| | - Yun Huang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| | - Xiao Sun
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Birth Defect Control and Prevention Research Center of Zhejiang Province, Hangzhou, China
| |
Collapse
|
47
|
Curtis ME, Smith T, Nenov M, Blass BE, Praticò D. Retromer Stabilization Improves Cognitive Function and Synaptic Plasticity in a Mouse Model of Down Syndrome. J Alzheimers Dis 2023:JAD230205. [PMID: 37334603 PMCID: PMC10357169 DOI: 10.3233/jad-230205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Retromer complex proteins are decreased in postmortem brain tissues from Down syndrome subjects and inversely correlate with the Alzheimer's disease-like neuropathology. However, whether targeting in vivo the retromer system affects cognitive deficits and synaptic function in Down syndrome remains unknown. OBJECTIVE The aim of the current study was to examine the effects of pharmacological retromer stabilization on cognitive and synaptic functions in a mouse model of Down syndrome. METHODS Ts65dn mice were administered the pharmacological chaperone, TPT-172, or vehicle from 4 to 9 months of age and then assessed for changes in cognitive function. To assess the effects of TPT-172 on synaptic plasticity, hippocampal slices from Ts65dn mice were incubated in TPT-172 and used for field potential recordings. RESULTS Chronic TPT-172 treatment improved performance in cognitive function tests, its incubation with hippocampal slices ameliorated synaptic function response. CONCLUSION Pharmacological stabilization of the retromer complex improves synaptic plasticity and memory in a mouse model of Down syndrome. These results support the therapeutic potential of pharmacological retromer stabilization for individual with Down syndrome.
Collapse
Affiliation(s)
- Mary Elizabeth Curtis
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Tiffany Smith
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Miroslav Nenov
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | | | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
48
|
Tristão RM, Scafutto Marengo LA, Costa JFDD, Pires ALDS, Boato EM. The use of the cambridge neuropsychological test automated battery for people born with Down syndrome and those born premature: A comparative systematic review. JOURNAL OF INTELLECTUAL DISABILITIES : JOID 2023; 27:539-567. [PMID: 35166595 DOI: 10.1177/17446295211050460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
This review aimed to investigate the use of the Cambridge Neuropsychological Automated Testing Battery (CANTAB) for people at risk of cognitive impairment, especially those born with Down syndrome and those born preterm. Six databases were searched according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standards, in addition to the bibliography index listed in the CANTAB site. Twenty four studies regarding Down syndrome and 17 regarding prematurity were reviewed and are here described. Both cognitive profiles were described, and their performance was compared on specific tasks and CANTAB tests. In this battery of tests, people with Down syndrome usually present impaired key cognitive domains, such as episodic memory and recognition memory. Results were presented considering general aspects described in the studies, specific findings such as dementia, the role of genetics, and cognitive profile, among other descriptions. Comparability between both populations in future studies is discussed.
Collapse
Affiliation(s)
- Rosana M Tristão
- Faculty of Medicine and University Hospital, Medicine of the Child and Adolescent, University of Brasilia, Brasilia, Brazil
| | | | | | - Ana Luísa Dos Santos Pires
- Faculty of Medicine and University Hospital, Medicine of the Child and Adolescent, University of Brasilia, Brasilia, Brazil
| | - Elvio M Boato
- Center for Science and Technology, CogniAction Lab, Catholic University of Brasilia, Brasilia, Brazil
| |
Collapse
|
49
|
Redhead Y, Gibbins D, Lana-Elola E, Watson-Scales S, Dobson L, Krause M, Liu KJ, Fisher EMC, Green JBA, Tybulewicz VLJ. Craniofacial dysmorphology in Down syndrome is caused by increased dosage of Dyrk1a and at least three other genes. Development 2023; 150:dev201077. [PMID: 37102702 PMCID: PMC10163349 DOI: 10.1242/dev.201077] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 03/21/2023] [Indexed: 04/28/2023]
Abstract
Down syndrome (DS), trisomy of human chromosome 21 (Hsa21), occurs in 1 in 800 live births and is the most common human aneuploidy. DS results in multiple phenotypes, including craniofacial dysmorphology, which is characterised by midfacial hypoplasia, brachycephaly and micrognathia. The genetic and developmental causes of this are poorly understood. Using morphometric analysis of the Dp1Tyb mouse model of DS and an associated mouse genetic mapping panel, we demonstrate that four Hsa21-orthologous regions of mouse chromosome 16 contain dosage-sensitive genes that cause the DS craniofacial phenotype, and identify one of these causative genes as Dyrk1a. We show that the earliest and most severe defects in Dp1Tyb skulls are in bones of neural crest (NC) origin, and that mineralisation of the Dp1Tyb skull base synchondroses is aberrant. Furthermore, we show that increased dosage of Dyrk1a results in decreased NC cell proliferation and a decrease in size and cellularity of the NC-derived frontal bone primordia. Thus, DS craniofacial dysmorphology is caused by an increased dosage of Dyrk1a and at least three other genes.
Collapse
Affiliation(s)
- Yushi Redhead
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | | | | | | | - Lisa Dobson
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Matthias Krause
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Karen J. Liu
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
| | | | - Jeremy B. A. Green
- Centre for Craniofacial Biology and Regenerative Biology, King's College London, London SE1 9RT, UK
| | | |
Collapse
|
50
|
Koul B, Farooq U, Yadav D, Song M. Phytochemicals: A Promising Alternative for the Prevention of Alzheimer's Disease. Life (Basel) 2023; 13:life13040999. [PMID: 37109528 PMCID: PMC10144079 DOI: 10.3390/life13040999] [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: 03/20/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a neurological condition that worsens with ageing and affects memory and cognitive function. Presently more than 55 million individuals are affected by AD all over the world, and it is a leading cause of death in old age. The main purpose of this paper is to review the phytochemical constituents of different plants that are used for the treatment of AD. A thorough and organized review of the existing literature was conducted, and the data under the different sections were found using a computerized bibliographic search through the use of databases such as PubMed, Web of Science, Google Scholar, Scopus, CAB Abstracts, MEDLINE, EMBASE, INMEDPLAN, NATTS, and numerous other websites. Around 360 papers were screened, and, out of that, 258 papers were selected on the basis of keywords and relevant information that needed to be included in this review. A total of 55 plants belonging to different families have been reported to possess different bioactive compounds (galantamine, curcumin, silymarin, and many more) that play a significant role in the treatment of AD. These plants possess anti-inflammatory, antioxidant, anticholinesterase, and anti-amyloid properties and are safe for consumption. This paper focuses on the taxonomic details of the plants, the mode of action of their phytochemicals, their safety, future prospects, limitations, and sustainability criteria for the effective treatment of AD.
Collapse
Affiliation(s)
- Bhupendra Koul
- Department of Biotechnology, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Usma Farooq
- Department of Botany, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|