1
|
Cindrić A, Vučković F, Murray A, Klarić TS, Alić I, Krištić J, Nižetić D, Lauc G. Total cell N-glycosylation is altered during differentiation of induced pluripotent stem cells to neural stem cells and is disturbed by trisomy 21. BBA ADVANCES 2024; 7:100137. [PMID: 39845703 PMCID: PMC11751427 DOI: 10.1016/j.bbadva.2024.100137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025] Open
Abstract
Down syndrome (DS), a genetic condition caused by trisomy 21 (T21), manifests various neurological symptoms, including intellectual disability, early neurodegeneration, and early-onset dementia. N-glycosylation is a protein modification that plays a critical role in numerous neurobiological processes and whose dysregulation is associated with a range of neurological disorders. However, whether N-glycosylation of neural glycoproteins is affected in DS has not been studied. To better understand how T21 affects N-glycosylation during neural differentiation, we utilized an isogenic in vitro induced pluripotent stem cell (iPSC) model of T21 in which both T21 and euploid disomic karyotype (D21) clones were obtained from a single individual with mosaic DS. We comprehensively characterized and compared the total N-glycomes of iPSCs and their neural stem cell (NSC) derivatives. N-glycomics analysis of whole cell lysates was performed using liquid chromatography coupled with tandem mass spectrometry to determine N-glycan structures. Our results show that neural differentiation of iPSCs to NSCs is characterized by an increase in the abundance of complex N-glycans at the expense of minimally processed mannosidic N-glycans. Moreover, we found differences in N-glycosylation patterns between D21 and T21 cells. Notably, the abundance of pseudohybrid N-glycans was significantly higher in T21 cells which also exhibited a significantly lower abundance of a specific hybrid monoantennary fucosylated N-glycan (H6N3F1). Overall, our data define the total N-glycome of both D21 and T21 iPSCs and NSCs and show that T21 already impacts N-glycosylation patterns in the stem cell state in a manner consistent with aberrantly premature neural differentiation of T21 cells.
Collapse
Affiliation(s)
- Ana Cindrić
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | - Frano Vučković
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
| | - Aoife Murray
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
| | | | - Ivan Alić
- Faculty of Veterinary Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Dean Nižetić
- The Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, E1 2AT, UK
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, 10000 Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
2
|
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: 10] [Impact Index Per Article: 5.0] [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
|
3
|
Martini AC, Gross TJ, Head E, Mapstone M. Beyond amyloid: Immune, cerebrovascular, and metabolic contributions to Alzheimer disease in people with Down syndrome. Neuron 2022; 110:2063-2079. [PMID: 35472307 PMCID: PMC9262826 DOI: 10.1016/j.neuron.2022.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/16/2022]
Abstract
People with Down syndrome (DS) have increased risk of Alzheimer disease (AD), presumably conferred through genetic predispositions arising from trisomy 21. These predispositions necessarily include triplication of the amyloid precursor protein (APP), but also other Ch21 genes that confer risk directly or through interactions with genes on other chromosomes. We discuss evidence that multiple genes on chromosome 21 are associated with metabolic dysfunction in DS. The resulting dysregulated pathways involve the immune system, leading to chronic inflammation; the cerebrovascular system, leading to disruption of the blood brain barrier (BBB); and cellular energy metabolism, promoting increased oxidative stress. In combination, these disruptions may produce a precarious biological milieu that, in the presence of accumulating amyloid, drives the pathophysiological cascade of AD in people with DS. Critically, mechanistic drivers of this dysfunction may be targetable in future clinical trials of pharmaceutical and/or lifestyle interventions.
Collapse
Affiliation(s)
- Alessandra C Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Thomas J Gross
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
4
|
Kirstein M, Cambrils A, Segarra A, Melero A, Varea E. Cholinergic Senescence in the Ts65Dn Mouse Model for Down Syndrome. Neurochem Res 2022; 47:3076-3092. [PMID: 35767135 PMCID: PMC9470680 DOI: 10.1007/s11064-022-03659-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022]
Abstract
Down syndrome (DS) induces a variable phenotype including intellectual disabilities and early development of Alzheimer’s disease (AD). Moreover, individuals with DS display accelerated aging that affects diverse organs, among them the brain. The Ts65Dn mouse is the most widely used model to study DS. Progressive loss of cholinergic neurons is one of the hallmarks of AD present in DS and in the Ts65Dn model. In this study, we quantify the number of cholinergic neurons in control and Ts65Dn mice, observing a general reduction in their number with age but in particular, a greater loss in old Ts65Dn mice. Increased expression of the m1 muscarinic receptor in the hippocampus counteracts this loss. Cholinergic neurons in the Ts65Dn mice display overexpression of the early expression gene c-fos and an increase in the expression of β-galactosidase, a marker of senescence. A possible mechanism for senescence induction could be phosphorylation of the transcription factor FOXO1 and its retention in the cytoplasm, which we are able to confirm in the Ts65Dn model. In our study, using Ts65Dn mice, we observe increased cholinergic activity, which induces a process of early senescence that culminates in the loss of these neurons.
Collapse
Affiliation(s)
- Martina Kirstein
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Alba Cambrils
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Ana Segarra
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Ana Melero
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain
| | - Emilio Varea
- Cell Biology Department, Universitat de València, Dr. Moliner, 50, Burjassot, 46100, València, Spain.
| |
Collapse
|
5
|
Hamadelseed O, Elkhidir IH, Skutella T. Psychosocial Risk Factors for Alzheimer's Disease in Patients with Down Syndrome and Their Association with Brain Changes: A Narrative Review. Neurol Ther 2022; 11:931-953. [PMID: 35596914 PMCID: PMC9338203 DOI: 10.1007/s40120-022-00361-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/28/2022] [Indexed: 12/02/2022] Open
Abstract
Several recent epidemiological studies attempted to identify risk factors for Alzheimer’s disease. Age, family history, genetic factors (APOE genotype, trisomy 21), physical activity, and a low level of schooling are significant risk factors. In this review, we summarize the known psychosocial risk factors for the development of Alzheimer’s disease in patients with Down syndrome and their association with neuroanatomical changes in the brains of people with Down syndrome. We completed a comprehensive review of the literature on PubMed, Google Scholar, and Web of Science about psychosocial risk factors for Alzheimer’s disease, for Alzheimer’s disease in Down syndrome, and Alzheimer’s disease in Down syndrome and their association with neuroanatomical changes in the brains of people with Down syndrome. Alzheimer’s disease causes early pathological changes in individuals with Down syndrome, especially in the hippocampus and corpus callosum. People with Down syndrome living with dementia showed reduced volumes of brain areas affected by Alzheimer’s disease as the hippocampus and corpus callosum in association with cognitive decline. These changes occur with increasing age, and the presence or absence of psychosocial risk factors impacts the degree of cognitive function. Correlating Alzheimer’s disease biomarkers in Down syndrome and cognitive function scores while considering the effect of psychosocial risk factors helps us identify the mechanisms leading to Alzheimer’s disease at an early age. Also, this approach enables us to create more sensitive and relevant clinical, memory, and reasoning assessments for people with Down syndrome.
Collapse
Affiliation(s)
- Osama Hamadelseed
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany.
| | - Ibrahim H Elkhidir
- Faculty of Medicine, University of Khartoum, Alqasr St., Khartoum, Sudan
| | - Thomas Skutella
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany
| |
Collapse
|
6
|
Norins LC. Down syndrome and Alzheimer’s disease: Same infectious cause, same preventive? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2021.110745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
7
|
Windsperger K, Hoehl S. Development of Down Syndrome Research Over the Last Decades-What Healthcare and Education Professionals Need to Know. Front Psychiatry 2021; 12:749046. [PMID: 34970162 PMCID: PMC8712441 DOI: 10.3389/fpsyt.2021.749046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Down syndrome (DS) is the most prevalent neurodevelopmental disorder, with a known genetic cause. Besides facial dysmorphologies and congenital and/or acquired medical conditions, the syndrome is characterized by intellectual disability, accelerated aging, and an increased likelihood of an early onset Alzheimer's disease in adulthood. These common patterns of DS are derived from the long-held standard in the field of DS research, that describes individuals with DS as a homogeneous group and compares phenotypic outcomes with either neurotypical controls or other neurodevelopmental disorders. This traditional view has changed, as modern research pinpoints a broad variability in both the occurrence and severity of symptoms across DS, arguing for DS heterogeneity and against a single "DS profile." Nevertheless, prenatal counseling does not often prioritize the awareness of potential within-group variations of DS, portraying only a vague picture of the developmental outcomes of children with DS to expectant parents. This mini-review provides a concise update on existent information about the heterogeneity of DS from a full-spectrum developmental perspective, within an interdisciplinary context. Knowledge on DS heterogeneity will not only enable professionals to enhance the quality of prenatal counseling, but also help parents to set targeted early interventions, to further optimize daily functions and the quality of life of their children.
Collapse
Affiliation(s)
- Karin Windsperger
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Stefanie Hoehl
- Research Unit Developmental Psychology, Department of Developmental and Educational Psychology, Faculty of Psychology, University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Jojoa-Acosta MF, Signo-Miguel S, Garcia-Zapirain MB, Gimeno-Santos M, Méndez-Zorrilla A, Vaidya CJ, Molins-Sauri M, Guerra-Balic M, Bruna-Rabassa O. Executive Functioning in Adults with Down Syndrome: Machine-Learning-Based Prediction of Inhibitory Capacity. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182010785. [PMID: 34682531 PMCID: PMC8536074 DOI: 10.3390/ijerph182010785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022]
Abstract
The study of executive function decline in adults with Down syndrome (DS) is important, because it supports independent functioning in real-world settings. Inhibitory control is posited to be essential for self-regulation and adaptation to daily life activities. However, cognitive domains that most predict the capacity for inhibition in adults with DS have not been identified. The aim of this study was to identify cognitive domains that predict the capacity for inhibition, using novel data-driven techniques in a sample of adults with DS (n = 188; 49.47% men; 33.6 ± 8.8 years old), with low and moderate levels of intellectual disability. Neuropsychological tests, including assessment of memory, attention, language, executive functions, and praxis, were submitted to Random Forest, support vector machine, and logistic regression algorithms for the purpose of predicting inhibition capacity, assessed with the Cats-and-Dogs test. Convergent results from the three algorithms show that the best predictors for inhibition capacity were constructive praxis, verbal memory, immediate memory, planning, and written verbal comprehension. These results suggest the minimum set of neuropsychological assessments and potential intervention targets for individuals with DS and ID, which may optimize potential for independent living.
Collapse
Affiliation(s)
- Mario Fernando Jojoa-Acosta
- eVIDA—Lab, Faculty of Engineering, Deusto University, 48007 Bilbao, Spain; (M.F.J.-A.); (M.B.G.-Z.); (A.M.-Z.)
| | - Sara Signo-Miguel
- Faculty of Psychology, Education and Sports Sciences Blanquerna, Ramon Llull University, 08022 Barcelona, Spain; (S.S.-M.); (M.G.-S.); (M.M.-S.); (M.G.-B.)
| | | | - Mercè Gimeno-Santos
- Faculty of Psychology, Education and Sports Sciences Blanquerna, Ramon Llull University, 08022 Barcelona, Spain; (S.S.-M.); (M.G.-S.); (M.M.-S.); (M.G.-B.)
| | - Amaia Méndez-Zorrilla
- eVIDA—Lab, Faculty of Engineering, Deusto University, 48007 Bilbao, Spain; (M.F.J.-A.); (M.B.G.-Z.); (A.M.-Z.)
| | - Chandan J. Vaidya
- Department of Psychology, Georgetown University, Washington, DC 20057, USA;
| | - Marta Molins-Sauri
- Faculty of Psychology, Education and Sports Sciences Blanquerna, Ramon Llull University, 08022 Barcelona, Spain; (S.S.-M.); (M.G.-S.); (M.M.-S.); (M.G.-B.)
| | - Myriam Guerra-Balic
- Faculty of Psychology, Education and Sports Sciences Blanquerna, Ramon Llull University, 08022 Barcelona, Spain; (S.S.-M.); (M.G.-S.); (M.M.-S.); (M.G.-B.)
| | - Olga Bruna-Rabassa
- Faculty of Psychology, Education and Sports Sciences Blanquerna, Ramon Llull University, 08022 Barcelona, Spain; (S.S.-M.); (M.G.-S.); (M.M.-S.); (M.G.-B.)
- Correspondence:
| |
Collapse
|
9
|
The Association between Sex and Risk of Alzheimer's Disease in Adults with Down Syndrome. J Clin Med 2021; 10:jcm10132966. [PMID: 34279450 PMCID: PMC8268850 DOI: 10.3390/jcm10132966] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Sex differences in the risk of Alzheimer’s Disease (AD) in adults with Down Syndrome (DS) have not been extensively investigated, and existing studies have found conflicting results. This study examined the effect of sex on the risk of AD in adults with DS, adjusted for covariates. Methods: Adults with DS were assessed longitudinally for the development of AD. Competing risk survival analyses were used to determine the effect of sex alone and after adjustment for APOE-ε4 status, ethnicity, and level of intellectual disability (ID). Results: Sex differences were significant only in adults over 60 years of age, where men with DS were 6.32 (95% CI: 2.11–18.96, p < 0.001) times more likely to develop AD compared with age-matched women with DS. Conclusions: There is an age-associated effect of sex on the risk of AD, with men over 60 years old having six times the risk of AD compared with age-matched women, independent of APOE-ε4 status, ethnicity, and level of ID.
Collapse
|
10
|
Ramírez-Toraño F, García-Alba J, Bruña R, Esteba-Castillo S, Vaquero L, Pereda E, Maestú F, Fernández A. Hypersynchronized Magnetoencephalography Brain Networks in Patients with Mild Cognitive Impairment and Alzheimer's Disease in Down Syndrome. Brain Connect 2021; 11:725-733. [PMID: 33858203 DOI: 10.1089/brain.2020.0897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Introduction: The majority of individuals with Down syndrome (DS) show signs of Alzheimer's disease (AD) neuropathology in their fourth decade. However, there is a lack of specific markers for characterizing the disease stages while considering this population's differential features. Methods: Forty-one DS individuals participated in the study, and were classified into three groups according to their clinical status: Alzheimer's disease (AD-DS), mild cognitive impairment (MCI-DS), and controls (CN-DS). We performed an exhaustive neuropsychological evaluation and assessed brain functional connectivity (FC) from magnetoencephalographic recordings. Results: Compared with CN-DS, both MCI-DS and AD-DS showed a pattern of increased FC within the high alpha band. The neuropsychological assessment showed a generalized cognitive impairment, especially affecting mnestic functions, in MCI-DS and, more pronouncedly, in AD-DS. Discussion: These findings might help to characterize the AD-continuum in DS. In addition, they support the role of the excitatory/inhibitory imbalance as a key pathophysiological factor in AD. Impact statement The pattern of functional connectivity (FC) hypersynchronization found in this study resembles the largely reported Alzheimer's disease (AD) FC evolution pattern in population with typical development. This study supports the hypothesis of the excitatory/inhibitory imbalance as a key pathophysiological factor in AD, and its conclusions could help in the characterization and prediction of Down syndrome individuals with a greater likelihood of converting to dementia.
Collapse
Affiliation(s)
- Federico Ramírez-Toraño
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Department of Experimental Psychology, Universidad Complutense de Madrid, Madrid, Spain
| | - Javier García-Alba
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Research and Psychology in Education Department, Complutense University of Madrid, Madrid, Spain
| | - Ricardo Bruña
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Department of Experimental Psychology, Universidad Complutense de Madrid, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Susanna Esteba-Castillo
- Specialized Department in Mental Health and Intellectual Disability, Parc Hospitalari Martí i Julià-Institut 'd'Assistència Sanitària, Institut 'd'Assistència Sanitària (IAS), Girona, Spain
| | - Lucía Vaquero
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Department of Legal Medicine, Psychiatry and Pathology, Complutense University of Madrid, Spain
| | - Ernesto Pereda
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Electrical Engineering and Bioengineering Group, Department of Industrial Engineering and IUNE and ITB Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Fernando Maestú
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Department of Experimental Psychology, Universidad Complutense de Madrid, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Alberto Fernández
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Department of Legal Medicine, Psychiatry and Pathology, Complutense University of Madrid, Spain.,Institute of Sanitary Investigation (IdISSC), San Carlos University Hospital, Madrid, Spain
| |
Collapse
|
11
|
Musaeus CS, Salem LC, Kjaer TW, Waldemar G. Electroencephalographic functional connectivity is altered in persons with Down syndrome and Alzheimer's disease. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2021; 65:236-245. [PMID: 33336867 DOI: 10.1111/jir.12803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/06/2020] [Accepted: 11/21/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Persons with Down syndrome (DS) are at increased risk of developing Alzheimer's dementia (DS-AD). Due to heterogeneity in the functioning in persons with DS, it is difficult to use cognitive testing to assess whether a person with DS has developed dementia due to AD. Electroencephalography (EEG) functional connectivity has shown promising results as a diagnostic tool for AD in persons without DS. In the current exploratory study, we investigated whether EEG functional connectivity could be used as a diagnostic marker of AD in persons with DS and the association with symptoms. METHODS Electroencephalography from 12 persons with DS and 16 persons with DS-AD were analysed, and both coherence and weighted phase lag index were calculated. In addition, we calculated the average coherence for fronto-parietal and temporo-parietal connections. Lastly, we investigated the correlation between the informant-based Dementia Screening Questionnaire in Intellectual Disability (DSQIID) and total alpha coherence. RESULTS Decreased alpha and increased delta coherence and weighted phase lag index were observed in DS-AD as compared with DS. The decrease in alpha coherence was more marked in the fronto-parietal connections as compared with the temporo-parietal connections. No significant correlation was found between DSQIID and total alpha coherence (P value = 0.095, rho = -0.335). CONCLUSION The decreased alpha coherence and weighted phase lag index have previously been found in AD. The increased delta coherence and weighted phase lag index may indicate a different initial neurophysiological presentation as compared with patients with AD or may be a sign of more advanced disease. Larger studies are needed to confirm the current findings.
Collapse
Affiliation(s)
- C S Musaeus
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - L C Salem
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - T W Kjaer
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Neurophysiology Center, Zealand University Hospital, Roskilde, Denmark
| | - G Waldemar
- Department of Neurology, Danish Dementia Research Centre (DDRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Lanzillotta C, Di Domenico F. Stress Responses in Down Syndrome Neurodegeneration: State of the Art and Therapeutic Molecules. Biomolecules 2021; 11:biom11020266. [PMID: 33670211 PMCID: PMC7916967 DOI: 10.3390/biom11020266] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Down syndrome (DS) is the most common genomic disorder characterized by the increased incidence of developing early Alzheimer’s disease (AD). In DS, the triplication of genes on chromosome 21 is intimately associated with the increase of AD pathological hallmarks and with the development of brain redox imbalance and aberrant proteostasis. Increasing evidence has recently shown that oxidative stress (OS), associated with mitochondrial dysfunction and with the failure of antioxidant responses (e.g., SOD1 and Nrf2), is an early signature of DS, promoting protein oxidation and the formation of toxic protein aggregates. In turn, systems involved in the surveillance of protein synthesis/folding/degradation mechanisms, such as the integrated stress response (ISR), the unfolded stress response (UPR), and autophagy, are impaired in DS, thus exacerbating brain damage. A number of pre-clinical and clinical studies have been applied to the context of DS with the aim of rescuing redox balance and proteostasis by boosting the antioxidant response and/or inducing the mechanisms of protein re-folding and clearance, and at final of reducing cognitive decline. So far, such therapeutic approaches demonstrated their efficacy in reverting several aspects of DS phenotype in murine models, however, additional studies aimed to translate these approaches in clinical practice are still needed.
Collapse
|
13
|
Thomas MSC, Ojinaga Alfageme O, D'Souza H, Patkee PA, Rutherford MA, Mok KY, Hardy J, Karmiloff-Smith A. A multi-level developmental approach to exploring individual differences in Down syndrome: genes, brain, behaviour, and environment. RESEARCH IN DEVELOPMENTAL DISABILITIES 2020; 104:103638. [PMID: 32653761 PMCID: PMC7438975 DOI: 10.1016/j.ridd.2020.103638] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 05/06/2023]
Abstract
In this article, we focus on the causes of individual differences in Down syndrome (DS), exemplifying the multi-level, multi-method, lifespan developmental approach advocated by Karmiloff-Smith (1998, 2009, 2012, 2016). We evaluate the possibility of linking variations in infant and child development with variations in the (elevated) risk for Alzheimer's disease (AD) in adults with DS. We review the theoretical basis for this argument, considering genetics, epigenetics, brain, behaviour and environment. In studies 1 and 2, we focus on variation in language development. We utilise data from the MacArthur-Bates Communicative Development Inventories (CDI; Fenson et al., 2007), and Mullen Scales of Early Learning (MSEL) receptive and productive language subscales (Mullen, 1995) from 84 infants and children with DS (mean age 2;3, range 0;7 to 5;3). As expected, there was developmental delay in both receptive and expressive vocabulary and wide individual differences. Study 1 examined the influence of an environmental measure (socio-economic status as measured by parental occupation) on the observed variability. SES did not predict a reliable amount of the variation. Study 2 examined the predictive power of a specific genetic measure (apolipoprotein APOE genotype) which modulates risk for AD in adulthood. There was no reliable effect of APOE genotype, though weak evidence that development was faster for the genotype conferring greater AD risk (ε4 carriers), consistent with recent observations in infant attention (D'Souza, Mason et al., 2020). Study 3 considered the concerted effect of the DS genotype on early brain development. We describe new magnetic resonance imaging methods for measuring prenatal and neonatal brain structure in DS (e.g., volumes of supratentorial brain, cortex, cerebellar volume; Patkee et al., 2019). We establish the methodological viability of linking differences in early brain structure to measures of infant cognitive development, measured by the MSEL, as a potential early marker of clinical relevance. Five case studies are presented as proof of concept, but these are as yet too few to discern a pattern.
Collapse
Affiliation(s)
- Michael S C Thomas
- Centre for Brain and Cognitive Development, Birkbeck, University of London, London WC1E 7HX, United Kingdom.
| | - Olatz Ojinaga Alfageme
- Centre for Brain and Cognitive Development, Birkbeck, University of London, London WC1E 7HX, United Kingdom; Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas's Hospital, London, SE1 7EH, United Kingdom
| | - Hana D'Souza
- Centre for Brain and Cognitive Development, Birkbeck, University of London, London WC1E 7HX, United Kingdom; Department of Psychology & Newnham College, University of Cambridge, Cambridge CB3 9DF, United Kingdom
| | - Prachi A Patkee
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas's Hospital, London, SE1 7EH, United Kingdom
| | - Mary A Rutherford
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas's Hospital, London, SE1 7EH, United Kingdom
| | - Kin Y Mok
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, United Kingdom
| | - John Hardy
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, United Kingdom
| | - Annette Karmiloff-Smith
- Centre for Brain and Cognitive Development, Birkbeck, University of London, London WC1E 7HX, United Kingdom
| |
Collapse
|
14
|
Kamer AR, Craig RG, Niederman R, Fortea J, de Leon MJ. Periodontal disease as a possible cause for Alzheimer's disease. Periodontol 2000 2020; 83:242-271. [PMID: 32385876 DOI: 10.1111/prd.12327] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/23/2019] [Indexed: 12/13/2022]
Abstract
Approximately 47 million people worldwide have been diagnosed with dementia, 60%-80% of whom have dementia of the Alzheimer's disease type. Unfortunately, there is no cure in sight. Defining modifiable risk factors for Alzheimer's disease may have a significant impact on its prevalence. An increasing body of evidence suggests that chronic inflammation and microbial dysbiosis are risk factors for Alzheimer's disease. Periodontal disease is a chronic inflammatory disease that develops in response to response to microbial dysbiosis. Many studies have shown an association between periodontal disease and Alzheimer's disease. The intent of this paper was to review the existing literature and determine, using the Bradford Hill criteria, whether periodontal disease is causally related to Alzheimer's disease.
Collapse
Affiliation(s)
- Angela R Kamer
- Department of Periodontology and Implant Dentistry, New York University, College of Dentistry, New York, New York, USA
| | - Ronald G Craig
- Department of Periodontology and Implant Dentistry, New York University, College of Dentistry, New York, New York, USA.,Department of Basic Sciences and Craniofacial Biology, New York University, College of Dentistry, New York, New York, USA
| | - Richard Niederman
- Department of Epidemiology and Health Promotion, New York University, College of Dentistry, New York, New York, USA
| | - Juan Fortea
- Alzheimer Down Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau- Biomedical Research Institute Sant Pau- Universitat Autònoma de Barcelona and Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain.,Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain
| | - Mony J de Leon
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
15
|
de Campos Gomes F, de Melo-Neto JS, Goloni-Bertollo EM, Pavarino ÉC. Trends and predictions for survival and mortality in individuals with Down syndrome in Brazil: A 21-year analysis. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2020; 64:551-560. [PMID: 32378275 DOI: 10.1111/jir.12735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 04/03/2020] [Accepted: 04/09/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Regional heterogeneities and sociodemographic characteristics affect mortality and population survival in Brazil. However, for individuals with Down syndrome (DS) this information remains unknown. In this study, we analysed survival and mortality rates among DS individuals in the five Brazilian geographic regions. In addition, we investigated whether there is an association between mortality and sociodemographic factors across administrative regions. METHODS Data between 1996 and 2016, comprising 10 028 records of deaths of individuals with DS, were collected from database records of the Department of Informatics of the Unified Health System. Data on race/ethnicity, sex, age and years of schooling were defined for the association analyses. Survival data were analysed according to the Kaplan-Meier method and Cox regression model. RESULTS The number of deaths among people with DS has increased in recent years. Children are more susceptible to death, especially in the first years of life. Individuals living in the northern region, Indigenous women and people with no years of schooling have higher mortality. In the Southeast and South region, for White and Yellow, survival is related to a higher level of education. Ethnic factors and years of schooling influence risk for mortality across the administrative regions. CONCLUSIONS These findings show that sociodemographic characteristics affect survival and are associated with the risk of mortality for people with DS. In addition, this suggests that differences in access to health services among Brazilian regions, especially in the first years of life, may affect the survival of individuals with DS.
Collapse
Affiliation(s)
- F de Campos Gomes
- Genetics and Molecular Biology Research Unit (UPGEM), São José do Rio Preto Medical School (FAMERP), São José do Rio Preto, Brazil
| | - J S de Melo-Neto
- Institute of Health Sciences, Federal University of Pará (UFPA), Belém, Brazil
| | - E M Goloni-Bertollo
- Genetics and Molecular Biology Research Unit (UPGEM), São José do Rio Preto Medical School (FAMERP), São José do Rio Preto, Brazil
| | - É C Pavarino
- Genetics and Molecular Biology Research Unit (UPGEM), São José do Rio Preto Medical School (FAMERP), São José do Rio Preto, Brazil
| |
Collapse
|
16
|
Carfì A, Romano A, Zaccaria G, Villani ER, Manes Gravina E, Vetrano DL, Bernabei R, Onder G. The burden of chronic disease, multimorbidity, and polypharmacy in adults with Down syndrome. Am J Med Genet A 2020; 182:1735-1743. [PMID: 32449279 DOI: 10.1002/ajmg.a.61636] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/02/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Data on clinical characteristics of adults with Down syndrome (DS) are limited and the clinical phenotype of these persons is poorly described. This study aimed to describe the occurrence of chronic diseases and pattern of medication use in a population of adults with DS. Participants were 421 community dwelling adults with DS, aged 18 years or older. Individuals were assessed through a standardized clinical protocol. Multimorbidity was defined as the occurrence of two or more chronic conditions and polypharmacy as the concomitant use of five or more medications. The mean age of study participants was 38.3 ± 12.8 years and 214 (51%) were women. Three hundred and seventy-four participants (88.8%) presented with multimorbidity. The most prevalent condition was visual impairment (72.9%), followed by thyroid disease (50.1%) and hearing impairment (26.8%). Chronic diseases were more prevalent among participants aged >40 years. The mean number of medications used was 2.09 and polypharmacy was observed in 10.5% of the study sample. Psychotropic medications were used by a mean of 0.7 individuals of the total sample. The high prevalence of multimorbidity and the common use of multiple medications contributes to a high level of clinical complexity, which appears to be similar to the degree of complexity of the older non-trisomic population. A comprehensive and holistic approach, commonly adopted in geriatric medicine, may provide the most appropriate care to persons with DS as they grow into adulthood.
Collapse
Affiliation(s)
- Angelo Carfì
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Allegra Romano
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Zaccaria
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emanuele Rocco Villani
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Manes Gravina
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Liborio Vetrano
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Neurobiology, Care Sciences and Society, Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Roberto Bernabei
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| | - Graziano Onder
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, and Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
17
|
Lee NR, Nayak A, Irfanoglu MO, Sadeghi N, Stoodley CJ, Adeyemi E, Clasen LS, Pierpaoli C. Hypoplasia of cerebellar afferent networks in Down syndrome revealed by DTI-driven tensor based morphometry. Sci Rep 2020; 10:5447. [PMID: 32214129 PMCID: PMC7096514 DOI: 10.1038/s41598-020-61799-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
Quantitative magnetic resonance imaging (MRI) investigations of brain anatomy in children and young adults with Down syndrome (DS) are limited, with no diffusion tensor imaging (DTI) studies covering that age range. We used DTI-driven tensor based morphometry (DTBM), a novel technique that extracts morphometric information from diffusion data, to investigate brain anatomy in 15 participants with DS and 15 age- and sex-matched typically developing (TD) controls, ages 6-24 years (mean age ~17 years). DTBM revealed marked hypoplasia of cerebellar afferent systems in DS, including fronto-pontine (middle cerebellar peduncle) and olivo-cerebellar (inferior cerebellar peduncle) connections. Prominent gray matter hypoplasia was observed in medial frontal regions, the inferior olives, and the cerebellum. Very few abnormalities were detected by classical diffusion MRI metrics, such as fractional anisotropy and mean diffusivity. Our results highlight the potential importance of cerebro-cerebellar networks in the clinical manifestations of DS and suggest a role for DTBM in the investigation of other brain disorders involving white matter hypoplasia or atrophy.
Collapse
Affiliation(s)
- Nancy Raitano Lee
- Drexel University, Department of Psychology, Philadelphia, PA, 19104, USA.
| | - Amritha Nayak
- National Institute of Biomedical Imaging and Bioengineering, NIH, Quantitative Medical Imaging Section, Bethesda, MD, 20892, USA
| | - M Okan Irfanoglu
- National Institute of Biomedical Imaging and Bioengineering, NIH, Quantitative Medical Imaging Section, Bethesda, MD, 20892, USA
| | - Neda Sadeghi
- National Institute of Biomedical Imaging and Bioengineering, NIH, Quantitative Medical Imaging Section, Bethesda, MD, 20892, USA
| | | | | | - Liv S Clasen
- National Institute of Mental Health, NIH, Developmental Neurogenomics Unit, Human Genetics Branch, Bethesda, MD, 20892, USA
| | - Carlo Pierpaoli
- National Institute of Biomedical Imaging and Bioengineering, NIH, Quantitative Medical Imaging Section, Bethesda, MD, 20892, USA
| |
Collapse
|
18
|
Musaeus CS, Salem LC, Kjaer TW, Waldemar G. Microstate Changes Associated With Alzheimer's Disease in Persons With Down Syndrome. Front Neurosci 2019; 13:1251. [PMID: 31849579 PMCID: PMC6892825 DOI: 10.3389/fnins.2019.01251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/05/2019] [Indexed: 11/13/2022] Open
Abstract
Down syndrome (DS) is associated with development of dementia due to Alzheimer’s disease (AD). However, due to considerable heterogeneity in intellectual function among persons with DS, it is difficult to assess whether a person with DS has developed dementia due to AD (DS-AD). EEG spectral power has previously shown very promising results with increased slowing in DS-AD compared to DS. However, another technique called microstates may be used to assess whole-brain dynamics and has to our knowledge not previously been investigated in either DS or DS-AD. The aim of the current study was to assess whether microstates could be used to differentiate between adults with DS, and DS-AD. We included EEGs from 10 persons with DS and 15 persons with DS-AD in the analysis. For the microstate analyses, we calculated four global maps, which were then back-fitted to all the EEGs. Lastly, we extracted the duration, occurrence, and coverage for each of the microstates. Here, we found the four archetypical maps as has previously been reported in the literature. We did not find any significant difference between DS and DS-AD but the largest difference in microstate duration between DS and DS-AD was found in microstate A and D. These findings are in line with structural MR studies showing that both the frontal and temporal lobes are affected in persons with DS-AD. Microstates may potentially serve as a diagnostic marker, but larger studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Christian Sandøe Musaeus
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lise Cronberg Salem
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Troels Wesenberg Kjaer
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Neurophysiology Center, Zealand University Hospital, Roskilde, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Yu YE, Xing Z, Do C, Pao A, Lee EJ, Krinsky-McHale S, Silverman W, Schupf N, Tycko B. Genetic and epigenetic pathways in Down syndrome: Insights to the brain and immune system from humans and mouse models. PROGRESS IN BRAIN RESEARCH 2019; 251:1-28. [PMID: 32057305 PMCID: PMC7286740 DOI: 10.1016/bs.pbr.2019.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The presence of an extra copy of human chromosome 21 (Hsa21) leads to a constellation of phenotypic manifestations in Down syndrome (DS), including prominent effects on the brain and immune system. Intensive efforts to unravel the molecular mechanisms underlying these phenotypes may help developing effective therapies, both in DS and in the general population. Here we review recent progress in genetic and epigenetic analysis of trisomy 21 (Ts21). New mouse models of DS based on syntenic conservation of segments of the mouse and human chromosomes are starting to clarify the contributions of chromosomal subregions and orthologous genes to specific phenotypes in DS. The expression of genes on Hsa21 is regulated by epigenetic mechanisms, and with recent findings of highly recurrent gene-specific changes in DNA methylation patterns in brain and immune system cells with Ts21, the epigenomics of DS has become an active research area. Here we highlight the value of combining human studies with mouse models for defining DS critical genes and understanding the trans-acting effects of a simple chromosomal aneuploidy on genome-wide epigenetic patterning. These genetic and epigenetic studies are starting to uncover fundamental biological mechanisms, leading to insights that may soon become therapeutically relevant.
Collapse
Affiliation(s)
- Y Eugene Yu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States; Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY, United States.
| | - Zhuo Xing
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Catherine Do
- Department of Biomedical Research, Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation and Hackensack-Meridian Health School of Medicine at Seton Hall University, Nutley, NJ, United States
| | - Annie Pao
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Eun Joon Lee
- Department of Biomedical Research, Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation and Hackensack-Meridian Health School of Medicine at Seton Hall University, Nutley, NJ, United States
| | - Sharon Krinsky-McHale
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Wayne Silverman
- Kennedy Krieger Institute and Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pediatrics, University of California at Irvine, Irvine, CA, United States
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States
| | - Benjamin Tycko
- Department of Biomedical Research, Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation and Hackensack-Meridian Health School of Medicine at Seton Hall University, Nutley, NJ, United States.
| |
Collapse
|
20
|
Choi C, Kim T, Chang KT, Min K. DSCR1-mediated TET1 splicing regulates miR-124 expression to control adult hippocampal neurogenesis. EMBO J 2019; 38:e101293. [PMID: 31304631 PMCID: PMC6627232 DOI: 10.15252/embj.2018101293] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 11/09/2022] Open
Abstract
Whether epigenetic factors such as DNA methylation and microRNAs interact to control adult hippocampal neurogenesis is not fully understood. Here, we show that Down syndrome critical region 1 (DSCR1) protein plays a key role in adult hippocampal neurogenesis by modulating two epigenetic factors: TET1 and miR-124. We find that DSCR1 mutant mice have impaired adult hippocampal neurogenesis. DSCR1 binds to TET1 introns to regulate splicing of TET1, thereby modulating TET1 level. Furthermore, TET1 controls the demethylation of the miRNA-124 promoter to modulate miR-124 expression. Correcting the level of TET1 in DSCR1 knockout mice is sufficient to prevent defective adult neurogenesis. Importantly, restoring DSCR1 level in a Down syndrome mouse model effectively rescued adult neurogenesis and learning and memory deficits. Our study reveals that DSCR1 plays a critical upstream role in epigenetic regulation of adult neurogenesis and provides insights into potential therapeutic strategy for treating cognitive defects in Down syndrome.
Collapse
Affiliation(s)
- Chiyeol Choi
- Department of Biological SciencesSchool of Life SciencesUlsan National Institute of Science and TechnologyUlsanKorea
- National Creative Research Initiative Center for ProteostasisUlsan National Institute of Science and TechnologyUlsanKorea
| | - Taehoon Kim
- Department of Biological SciencesSchool of Life SciencesUlsan National Institute of Science and TechnologyUlsanKorea
- National Creative Research Initiative Center for ProteostasisUlsan National Institute of Science and TechnologyUlsanKorea
| | - Karen T Chang
- Zilkha Neurogenetic InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Kyung‐Tai Min
- Department of Biological SciencesSchool of Life SciencesUlsan National Institute of Science and TechnologyUlsanKorea
- National Creative Research Initiative Center for ProteostasisUlsan National Institute of Science and TechnologyUlsanKorea
| |
Collapse
|
21
|
Sherzai D, Sherzai A. Preventing Alzheimer's: Our Most Urgent Health Care Priority. Am J Lifestyle Med 2019; 13:451-461. [PMID: 31523210 DOI: 10.1177/1559827619843465] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dementia is the fastest growing epidemic in the developed nations, and if not curtailed, it will single handedly collapse our health care system. The prevalence of dementia is 1 in 10 individuals older than 65 years and increases to 50% of all individuals older than 85 years. The prevalence of Alzheimer's dementia (AD), the most common form of dementia, has been increasing rapidly and is projected to reach 16 million individuals by the year 2050. Several prevailing myths about the science of dementia are discussed, such as that AD is inevitable and that it is exclusively a genetic disease. The fact is that AD is dependent on a multitude of genetic, epigenetic, and environmental factors that interact with one another. In fact, 4 core drivers represent 90% of what determines disease progression in AD. These are (1) glucose or energy dysregulation, (2) lipid dysregulation, (3) inflammation, and (4) oxidation. Lifestyle change can significantly alter the course of AD. The authors have created an acronym-NEURO-to help lifestyle practitioners and the public remember the most important lifestyle elements in the treatment and prevention of AD based on the evidence. "N" is for Nutrition, "E" for Exercise, "U" for Unwind (stress management), "R" for Restorative Sleep, and "O" for Optimizing mental and social activity. The evidence base for each of the components is reviewed.
Collapse
Affiliation(s)
- Dean Sherzai
- Department of Neurology, Alzheimer's Prevention Program, Loma Linda University Health, California
| | - Ayesha Sherzai
- Department of Neurology, Alzheimer's Prevention Program, Loma Linda University Health, California
| |
Collapse
|
22
|
Fick DM. Aging and Disabilities: How to Age Well With Down Syndrome and Leave No One Behind. J Gerontol Nurs 2019; 45:2-4. [PMID: 31026325 DOI: 10.3928/00989134-20190410-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Zhao X, Bhattacharyya A. Human Models Are Needed for Studying Human Neurodevelopmental Disorders. Am J Hum Genet 2018; 103:829-857. [PMID: 30526865 DOI: 10.1016/j.ajhg.2018.10.009] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 10/09/2018] [Indexed: 12/19/2022] Open
Abstract
The analysis of animal models of neurological disease has been instrumental in furthering our understanding of neurodevelopment and brain diseases. However, animal models are limited in revealing some of the most fundamental aspects of development, genetics, pathology, and disease mechanisms that are unique to humans. These shortcomings are exaggerated in disorders that affect the brain, where the most significant differences between humans and animal models exist, and could underscore failures in targeted therapeutic interventions in affected individuals. Human pluripotent stem cells have emerged as a much-needed model system for investigating human-specific biology and disease mechanisms. However, questions remain regarding whether these cell-culture-based models are sufficient or even necessary. In this review, we summarize human-specific features of neurodevelopment and the most common neurodevelopmental disorders, present discrepancies between animal models and human diseases, demonstrate how human stem cell models can provide meaningful information, and discuss the challenges that exist in our pursuit to understand distinctively human aspects of neurodevelopment and brain disease. This information argues for a more thoughtful approach to disease modeling through consideration of the valuable features and limitations of each model system, be they human or animal, to mimic disease characteristics.
Collapse
Affiliation(s)
- Xinyu Zhao
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA.
| | - Anita Bhattacharyya
- Waisman Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison WI 53705, USA.
| |
Collapse
|
24
|
White matter involvement in young non-demented Down's syndrome subjects: a tract-based spatial statistic analysis. Neuroradiology 2018; 60:1335-1341. [PMID: 30264168 DOI: 10.1007/s00234-018-2102-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/17/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE Cognitive decline in Down syndrome generally shows neurodegenerative aspects similar to what is observed in Alzheimer's disease. Few studies reported information on white matter integrity. The aim of this study was to evaluate white matter alterations in a cohort of young Down subjects, without dementia, by means of DTI technique, compared to a normal control group. METHODS The study group consisted of 17 right-handed subjects with DS and many control subjects. All individuals participating in this study were examined by MR exam including DTI acquisition (32 non-coplanar directions); image processing and analysis were performed using FMRIB Software Library (FSL version 4.1.9, http://www.fmrib.ox.ac.uk/fsl )) software package. Finally, the diffusion tensor was estimated voxel by voxel and the FA map derived from the tensor. A two-sample t test was performed to assess differences between DS and control subjects. RESULTS The FA is decreased in DS subjects, compared to control subjects, in the region of the anterior thalamic radiation, the inferior fronto-occipital fasciculum, the inferior longitudinal fasciculum, and the cortico-spinal tract, bilaterally. CONCLUSIONS The early white matter damage visible in our DS subjects could have great impact in the therapeutic management, in particular in better adapting the timing of therapies to counteract the toxic effect of the deposition of amyloid that leads to oxidative stress.
Collapse
|
25
|
Nguyen TL, Duchon A, Manousopoulou A, Loaëc N, Villiers B, Pani G, Karatas M, Mechling AE, Harsan LA, Limanton E, Bazureau JP, Carreaux F, Garbis SD, Meijer L, Herault Y. Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological inhibitor of DYRK1A. Dis Model Mech 2018; 11:dmm035634. [PMID: 30115750 PMCID: PMC6176987 DOI: 10.1242/dmm.035634] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Growing evidence supports the implication of DYRK1A in the development of cognitive deficits seen in Down syndrome (DS) and Alzheimer's disease (AD). We here demonstrate that pharmacological inhibition of brain DYRK1A is able to correct recognition memory deficits in three DS mouse models with increasing genetic complexity [Tg(Dyrk1a), Ts65Dn, Dp1Yey], all expressing an extra copy of Dyrk1a Overexpressed DYRK1A accumulates in the cytoplasm and at the synapse. Treatment of the three DS models with the pharmacological DYRK1A inhibitor leucettine L41 leads to normalization of DYRK1A activity and corrects the novel object cognitive impairment observed in these models. Brain functional magnetic resonance imaging reveals that this cognitive improvement is paralleled by functional connectivity remodelling of core brain areas involved in learning/memory processes. The impact of Dyrk1a trisomy and L41 treatment on brain phosphoproteins was investigated by a quantitative phosphoproteomics method, revealing the implication of synaptic (synapsin 1) and cytoskeletal components involved in synaptic response and axonal organization. These results encourage the development of DYRK1A inhibitors as drug candidates to treat cognitive deficits associated with DS and AD.
Collapse
Affiliation(s)
- Thu Lan Nguyen
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 67400 Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67400 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67400 Illkirch, France
- Université de Strasbourg, 67400 Illkirch, France
- ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Arnaud Duchon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 67400 Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67400 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67400 Illkirch, France
- Université de Strasbourg, 67400 Illkirch, France
| | - Antigoni Manousopoulou
- Faculty of Medicine/Cancer Sciences & Clinical and Experimental Medicine, University of Southampton, Center for Proteomic Research, Life Sciences Building 85, Highfield, Southampton SO17 1BJ, UK
| | - Nadège Loaëc
- ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Benoît Villiers
- ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Guillaume Pani
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 67400 Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67400 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67400 Illkirch, France
- Université de Strasbourg, 67400 Illkirch, France
| | - Meltem Karatas
- Laboratory of Engineering, Informatics and Imaging (ICube), Integrative multimodal imaging in healthcare (IMIS), UMR 7357, and University Hospital Strasbourg, Department of Biophysics and Nuclear Medicine, University of Strasbourg, 67400 Illkirch, France
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Breisacher Strasse 60a, 79106 Freiburg, Germany
| | - Anna E Mechling
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Breisacher Strasse 60a, 79106 Freiburg, Germany
| | - Laura-Adela Harsan
- Laboratory of Engineering, Informatics and Imaging (ICube), Integrative multimodal imaging in healthcare (IMIS), UMR 7357, and University Hospital Strasbourg, Department of Biophysics and Nuclear Medicine, University of Strasbourg, 67400 Illkirch, France
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Breisacher Strasse 60a, 79106 Freiburg, Germany
| | - Emmanuelle Limanton
- Université de Rennes 1, ISCR (Institut des sciences chimiques de Rennes)-UMR, 6226, 35000 Rennes, France
| | - Jean-Pierre Bazureau
- Université de Rennes 1, ISCR (Institut des sciences chimiques de Rennes)-UMR, 6226, 35000 Rennes, France
| | - François Carreaux
- Université de Rennes 1, ISCR (Institut des sciences chimiques de Rennes)-UMR, 6226, 35000 Rennes, France
| | - Spiros D Garbis
- Faculty of Medicine/Cancer Sciences & Clinical and Experimental Medicine, University of Southampton, Center for Proteomic Research, Life Sciences Building 85, Highfield, Southampton SO17 1BJ, UK
| | - Laurent Meijer
- ManRos Therapeutics, Perharidy Research Center, 29680 Roscoff, Bretagne, France
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 67400 Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67400 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67400 Illkirch, France
- Université de Strasbourg, 67400 Illkirch, France
| |
Collapse
|
26
|
McKenzie K, Metcalfe D, Michie A, Murray G. Service provision in Scotland for people with an intellectual disability who have, or are at risk of developing, dementia. DEMENTIA 2018; 19:736-749. [PMID: 29958499 DOI: 10.1177/1471301218785795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This research aimed to identify current national provision by health services in Scotland in relation to proactive screening and reactive assessment for people with an intellectual disability in Scotland who have, or are at risk of developing, dementia. Staff from 12 intellectual disability services, representing the 11 health board areas in Scotland, completed an online questionnaire which asked about proactive screening and reactive assessment for people with intellectual disability who had, or were at risk of developing, dementia as well as suggested areas for improvement. All of the areas provided services for people with intellectual disability who have, or are at risk of developing, dementia, but differed as to whether this was reactive, proactive or both. Nine services offered intervention following diagnosis. The most common elements used across both proactive screening and reactive assessment were conducting a health check, using a general dementia questionnaire designed for people with an intellectual disability and direct assessment with the person. Clinical psychology and community learning disability nurses were the professions most likely to be involved routinely in both proactive screening and reactive assessments. The psychometric properties of the most commonly used assessments of cognitive and behavioural functioning were mixed. The areas of improvement suggested by practitioners mainly related to ways of improving existing pathways. This research represents the first step in providing an overview of service provision in Scotland. There was some inconsistency in relation to the general and specific components which were involved in proactive screening and reactive assessment. Implications for service provision are discussed.
Collapse
Affiliation(s)
| | - Dale Metcalfe
- Northumbria University, Newcastle upon Tyne, UK.,Northumbria University, Newcastle upon Tyne, UK
| | - Amanda Michie
- NHS Lothian, Edinburgh, UK.,Northumbria University, Newcastle upon Tyne, UK
| | | |
Collapse
|
27
|
Head E, Powell DK, Schmitt FA. Metabolic and Vascular Imaging Biomarkers in Down Syndrome Provide Unique Insights Into Brain Aging and Alzheimer Disease Pathogenesis. Front Aging Neurosci 2018; 10:191. [PMID: 29977201 PMCID: PMC6021507 DOI: 10.3389/fnagi.2018.00191] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/06/2018] [Indexed: 12/26/2022] Open
Abstract
People with Down syndrome (DS) are at high risk for developing Alzheimer disease (AD). Neuropathology consistent with AD is present by 40 years of age and dementia may develop up to a decade later. In this review, we describe metabolic and vascular neuroimaging studies in DS that suggest these functional changes are a key feature of aging, linked to cognitive decline and AD in this vulnerable cohort. FDG-PET imaging in DS suggests systematic reductions in glucose metabolism in posterior cingulate and parietotemporal cortex. Magentic resonance spectroscopy studies show consistent decreases in neuronal health and increased myoinositol, suggesting inflammation. There are few vascular imaging studies in DS suggesting a gap in our knowledge. Future studies would benefit from longitudinal measures and combining various imaging approaches to identify early signs of dementia in DS that may be amenable to intervention.
Collapse
Affiliation(s)
- Elizabeth Head
- Department of Pharmacology & Nutritional Sciences, Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - David K Powell
- Magnetic Resonance Imaging and Spectroscopy Center, Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Frederick A Schmitt
- Department of Neurology, Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
28
|
Manti S, Cutrupi MC, Cuppari C, Ferro E, Dipasquale V, Di Rosa G, Chimenz R, La Rosa MA, Valenti A, Salpietro V. Inflammatory biomarkers and intellectual disability in patients with Down syndrome. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2018; 62:382-390. [PMID: 29349839 DOI: 10.1111/jir.12470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 11/22/2017] [Accepted: 12/15/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Intellectual disability (ID) is part of the Down syndrome (DS) phenotypic spectrum, but the exact molecular pathophysiology of ID in individuals with DS is not yet fully understood, with many research hypotheses still unproven. Basing on previous studies (which suggested a possible role of altered inflammatory response in DS-related ID), we assessed the serum levels of a number of inflammatory biomarkers [serum amyloid A (SAA), C-reactive protein (C-RP), high mobility group box-1 (HMGB1)] in a cohort of individuals with DS and healthy controls. METHODS In total, 24 children diagnosed with DS and 12 healthy controls were enrolled, and all underwent detailed cognitive assessment. Also, serum SAA, C-RP and HMGB1 levels were measured in all recruited subjects and correlated to the severity of ID in the DS group. RESULTS Serum SAA, C-RP and HMGB1 values were found to be significantly higher in the DS group compared with the healthy subjects (P = 0.001). In addition, serum HMGB1 levels positively correlated with C-RP and SAA in the DS group but not in the healthy controls. Only serum C-RP levels resulted inversely correlated (P < 0.01) with intelligence quotient (IQ); conversely, significant statistical correlations between serum SAA levels and IQ (as well as between HMGB1 and IQ) have been not found (P > 0.05). CONCLUSIONS The levels of the determined markers were higher in DS individuals compared with (cognitively) healthy subjects, and CRP showed a negative correlation with IQ in children with DS.
Collapse
Affiliation(s)
- S Manti
- Department of Human Pathology of Adult and Developmental Age 'Gaetano Barresi', University Hospital of Messina, Italy
| | - M C Cutrupi
- Department of Human Pathology of Adult and Developmental Age 'Gaetano Barresi', University Hospital of Messina, Italy
| | - C Cuppari
- Department of Human Pathology of Adult and Developmental Age 'Gaetano Barresi', University Hospital of Messina, Italy
| | - E Ferro
- Department of Human Pathology of Adult and Developmental Age 'Gaetano Barresi', University Hospital of Messina, Italy
| | - V Dipasquale
- Department of Human Pathology of Adult and Developmental Age 'Gaetano Barresi', University Hospital of Messina, Italy
| | - G Di Rosa
- Department of Human Pathology of the Adult and Developmental Age 'Gaetano Barresi', Unit of Child Neurology and Psychiatry, University of Messina, Messina, Italy
| | - R Chimenz
- Department of Human Pathology of Adult and Developmental Age 'Gaetano Barresi', University Hospital of Messina, Italy
| | - M A La Rosa
- Department of Human Pathology of Adult and Developmental Age 'Gaetano Barresi', University Hospital of Messina, Italy
| | - A Valenti
- Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - V Salpietro
- Department of Molecular Neurosciences, University College of London, London, UK
| |
Collapse
|
29
|
Lwanga A, Herrera W, Cruz Madrid K, Irungu A. Outpatient Interventions That May Enhance the Care of a Patient with Co-existing Moyamoya and Down Syndromes. Cureus 2018; 10:e2336. [PMID: 29774174 PMCID: PMC5955713 DOI: 10.7759/cureus.2336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Moyamoya vasculopathy is a condition of chronic, progressive occlusion of the distal internal carotid arteries and the Circle of Willis. The resultant ischemia produces compensatory angiogenesis and the growth of a network of collateral blood vessels, which on angiography resemble a “puff of smoke” or “moyamoya” in Japanese. The objective of this case report is to describe the clinical course of a patient with Down and moyamoya syndromes and to enlighten clinicians about strategies that can be taken to enhance the care of similar patients. A 55-year-old African American female presented to the hospital with complaints of headache, vision loss, dysarthria, and ataxia. She had a past medical history of Down syndrome and a stroke with residual lower extremity weakness. At her baseline, the patient was able to perform her activities of daily living but required assistance with independent activities of daily living. Computed tomography of the brain showed hypodense areas at the right occipital, temporal, and parietal lobes. Computed tomography angiography of the head and neck identified occlusion bilaterally at the supraclinoid internal carotid arteries and right posterior cerebral artery; there was collateral arterial flow within the right middle cerebral and anterior cerebral arteries that was consistent with moyamoya vasculopathy. Patients with Down syndrome experience premature accelerated aging and suffer from comorbidities seen in geriatric patients by the time they reach their 40s. Patients with moyamoya vasculopathy experience neurocognitive and neuropsychiatric deficits that correspond to the regions of the brain that are affected. This patient with Down and moyamoya syndromes had impaired neurocognitive and functional status, and we believe that she would have benefited from receiving a comprehensive geriatric assessment and neuropsychiatric testing.
Collapse
Affiliation(s)
- Anita Lwanga
- Department of Academic Internal Medicine and Geriatrics, University of Illinois at Chicago
| | - Waldo Herrera
- Internal Medicine/division of Hoapital Medicine, NorthShore University Health System
| | - Katya Cruz Madrid
- Department of Academic Internal Medicine and Geriatrics, University of Illinois In Chicago
| | | |
Collapse
|
30
|
Caraci F, Iulita MF, Pentz R, Flores Aguilar L, Orciani C, Barone C, Romano C, Drago F, Cuello AC. Searching for new pharmacological targets for the treatment of Alzheimer's disease in Down syndrome. Eur J Pharmacol 2017; 817:7-19. [DOI: 10.1016/j.ejphar.2017.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/26/2017] [Accepted: 10/04/2017] [Indexed: 11/26/2022]
|
31
|
Lee JH, Lee AJ, Dang LH, Pang D, Kisselev S, Krinsky-McHale SJ, Zigman WB, Luchsinger JA, Silverman W, Tycko B, Clark LN, Schupf N. Candidate gene analysis for Alzheimer's disease in adults with Down syndrome. Neurobiol Aging 2017; 56:150-158. [PMID: 28554490 DOI: 10.1016/j.neurobiolaging.2017.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 03/17/2017] [Accepted: 04/21/2017] [Indexed: 11/16/2022]
Abstract
Individuals with Down syndrome (DS) overexpress many genes on chromosome 21 due to trisomy and have high risk of dementia due to the Alzheimer's disease (AD) neuropathology. However, there is a wide range of phenotypic differences (e.g., age at onset of AD, amyloid β levels) among adults with DS, suggesting the importance of factors that modify risk within this particularly vulnerable population, including genotypic variability. Previous genetic studies in the general population have identified multiple genes that are associated with AD. This study examined the contribution of polymorphisms in these genes to the risk of AD in adults with DS ranging from 30 to 78 years of age at study entry (N = 320). We used multiple logistic regressions to estimate the likelihood of AD using single-nucleotide polymorphisms (SNPs) in candidate genes, adjusting for age, sex, race/ethnicity, level of intellectual disability and APOE genotype. This study identified multiple SNPs in APP and CST3 that were associated with AD at a gene-wise level empirical p-value of 0.05, with odds ratios in the range of 1.5-2. SNPs in MARK4 were marginally associated with AD. CST3 and MARK4 may contribute to our understanding of potential mechanisms where CST3 may contribute to the amyloid pathway by inhibiting plaque formation, and MARK4 may contribute to the regulation of the transition between stable and dynamic microtubules.
Collapse
Affiliation(s)
- Joseph H Lee
- Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA.
| | - Annie J Lee
- Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Lam-Ha Dang
- Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA
| | - Deborah Pang
- Department of Psychology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Sergey Kisselev
- Department of Pathology & Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Sharon J Krinsky-McHale
- Department of Psychology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Warren B Zigman
- Department of Psychology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - José A Luchsinger
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Wayne Silverman
- Kennedy Krieger Institute and Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin Tycko
- Department of Pathology & Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Lorraine N Clark
- Taub Institute, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Pathology & Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Nicole Schupf
- Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Epidemiology, School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
32
|
Shirao T, Hanamura K, Koganezawa N, Ishizuka Y, Yamazaki H, Sekino Y. The role of drebrin in neurons. J Neurochem 2017; 141:819-834. [PMID: 28199019 DOI: 10.1111/jnc.13988] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 12/27/2016] [Accepted: 01/11/2017] [Indexed: 01/13/2023]
Abstract
Drebrin is an actin-binding protein that changes the helical pitch of actin filaments (F-actin), and drebrin-decorated F-actin shows slow treadmilling and decreased rate of depolymerization. Moreover, the characteristic morphology of drebrin-decorated F-actin enables it to respond differently to the same signals from other actin cytoskeletons. Drebrin consists of two major isoforms, drebrin E and drebrin A. In the developing brain, drebrin E appears in migrating neurons and accumulates in the growth cones of axons and dendrites. Drebrin E-decorated F-actin links lamellipodium F-actin to microtubules in the growth cones. Then drebrin A appears at nascent synapses and drebrin A-decorated F-actin facilitates postsynaptic molecular assembly. In the adult brain, drebrin A-decorated F-actin is concentrated in the central region of dendritic spines. During long-term potentiation initiation, NMDA receptor-mediated Ca2+ influx induces the transient exodus of drebrin A-decorated F-actin via myosin II ATPase activation. Because of the unique physical characteristics of drebrin A-decorated F-actin, this exodus likely contributes to the facilitation of F-actin polymerization and spine enlargement. Additionally, drebrin reaccumulation in dendritic spines is observed after the exodus. In our drebrin exodus model of structure-based synaptic plasticity, reestablishment of drebrin A-decorated F-actin is necessary to keep the enlarged spine size during long-term potentiation maintenance. In this review, we introduce the genetic and biochemical properties of drebrin and the roles of drebrin in early stage of brain development, synaptic formation and synaptic plasticity. Further, we discuss the pathological relevance of drebrin loss in Alzheimer's disease. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".
Collapse
Affiliation(s)
- Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kenji Hanamura
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Noriko Koganezawa
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yuta Ishizuka
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yuko Sekino
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.,Division of Pharmacology, National Institute of Health Sciences, Tokyo, Japan
| |
Collapse
|
33
|
Tramutola A, Di Domenico F, Barone E, Arena A, Giorgi A, di Francesco L, Schininà ME, Coccia R, Head E, Butterfield DA, Perluigi M. Polyubiquitinylation Profile in Down Syndrome Brain Before and After the Development of Alzheimer Neuropathology. Antioxid Redox Signal 2017; 26:280-298. [PMID: 27627691 PMCID: PMC5327052 DOI: 10.1089/ars.2016.6686] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIMS Among the putative mechanisms proposed to be common factors in Down syndrome (DS) and Alzheimer's disease (AD) neuropathology, deficits in protein quality control (PQC) have emerged as a unifying mechanism of neurodegeneration. Considering that disturbance of protein degradation systems is present in DS and that oxidized/misfolded proteins require polyubiquitinylation for degradation via the ubiquitin proteasome system, this study investigated if dysregulation of protein polyubiquitinylation is associated with AD neurodegeneration in DS. RESULTS Postmortem brains from DS cases before and after development of AD neuropathology and age-matched controls were analyzed. By selectively isolating polyubiquitinated proteins, we were able to identify specific proteins with an altered pattern of polyubiquitinylation as a function of age. Interestingly, we found that oxidation is coupled with polyubiquitinylation for most proteins mainly involved in PQC and energy metabolism. INNOVATION This is the first study showing alteration of the polyubiquitinylation profile as a function of aging in DS brain compared with healthy controls. Understanding the onset of the altered ubiquitome profile in DS brain may contribute to identification of key molecular regulators of age-associated cognitive decline. CONCLUSIONS Disturbance of the polyubiquitinylation machinery may be a key feature of aging and neurodegeneration. In DS, age-associated deficits of the proteolytic system may further exacerbate the accumulation of oxidized/misfolded/polyubiquitinated proteins, which is not efficiently degraded and may become harmful to neurons and contribute to AD neuropathology. Antioxid. Redox Signal. 26, 280-298.
Collapse
Affiliation(s)
- Antonella Tramutola
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| | - Fabio Di Domenico
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| | - Eugenio Barone
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| | - Andrea Arena
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| | - Alessandra Giorgi
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| | - Laura di Francesco
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| | | | - Raffaella Coccia
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| | - Elizabeth Head
- 2 Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky.,3 Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington, Kentucky
| | - D Allan Butterfield
- 2 Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky.,4 Department of Chemistry, University of Kentucky , Lexington, Kentucky
| | - Marzia Perluigi
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Italy, Rome
| |
Collapse
|
34
|
Asai M, Kinjo A, Kimura S, Mori R, Kawakubo T, Shirotani K, Yagishita S, Maruyama K, Iwata N. Perturbed Calcineurin-NFAT Signaling Is Associated with the Development of Alzheimer's Disease. Biol Pharm Bull 2017; 39:1646-1652. [PMID: 27725441 DOI: 10.1248/bpb.b16-00350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Down syndrome (DS), the most common genetic disorder, is caused by trisomy 21. DS is accompanied by heart defects, hearing and vision problems, obesity, leukemia, and other conditions, including Alzheimer's disease (AD). In comparison, most cancers are rare in people with DS. Overexpression of dual specificity tyrosine-phosphorylation-regulated kinase 1A and a regulator of calcineurin 1 located on chromosome 21 leads to excessive suppression of the calcineurin-nuclear factor of activated T cells (NFAT) signaling pathway, resulting in reduced expression of a critical angiogenic factor. However, it is unclear whether the calcineurin-NFAT signaling pathway is involved in AD pathology in DS patients. Here, we investigated the association between the calcineurin-NFAT signaling pathway and AD using neuronal cells. Short-term pharmacological stimulation decreased gene expression of tau and neprilysin, and long-term inhibition of the signaling pathway decreased that of amyloid precursor protein. Moreover, a calcineurin inhibitor, cyclosporine A, also decreased neprilysin activity, leading to increases in amyloid-β peptide levels. Taken together, our results suggest that a dysregulation in calcineurin-NFAT signaling may contribute to the early onset of AD in people with DS.
Collapse
Affiliation(s)
- Masashi Asai
- School of Pharmaceutical Sciences, Nagasaki University
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ishizuka Y, Hanamura K. Drebrin in Alzheimer’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:203-223. [DOI: 10.1007/978-4-431-56550-5_12] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
36
|
Kawakubo T, Mori R, Shirotani K, Iwata N, Asai M. Neprilysin Is Suppressed by Dual-Specificity Tyrosine-Phosphorylation Regulated Kinase 1A (DYRK1A) in Down-Syndrome-Derived Fibroblasts. Biol Pharm Bull 2017; 40:327-333. [PMID: 28250274 DOI: 10.1248/bpb.b16-00825] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Amyloid-β peptide (Aβ) accumulation is a triggering event leading to the Alzheimer's disease (AD) pathological cascade. Almost all familial AD-linked gene mutations increase Aβ production and accelerate the onset of AD. The Swedish mutation of amyloid precursor protein (APP) affects β-secretase activity and increases Aβ production up to ca. 6-fold in cultured cells; the onset age is around 50. Down syndrome (DS) patients with chromosome 21 trisomy present AD-like pathologies at earlier ages (40s) compared with sporadic AD patients, because APP gene expression is 1.5-fold higher than that in healthy people, thus causing a 1.5-fold increase in Aβ production. However, when comparing the causal relationship of Aβ accumulation with the onset age between the above two populations, early DS pathogenesis does not appear to be accounted for by the increased Aβ production alone. In this study, we found that neprilysin, a major Aβ-degrading enzyme, was downregulated in DS patient-derived fibroblasts, compared with healthy people-derived fibroblasts. Treatment with harmine, an inhibitor of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), which is located in the DS critical region of chromosome 21, and gene knockdown of DYRK1A, upregulated neprilysin in fibroblasts. These results suggest that a decrease in the Aβ catabolic rate may be, at least in part, one of the causes for accelerated AD-like pathogenesis in DS patients if a similar event occurs in the brains, and that neprilysin activity may be regulated directly or indirectly by DYRK1A-mediated phosphorylation. DYRK1A inhibition may be a promising disease-modifying therapy for AD via neprilysin upregulation.
Collapse
Affiliation(s)
- Takashi Kawakubo
- Department of Genome-based Drug Discovery, Graduation School of Biomedical Sciences, Nagasaki University
| | | | | | | | | |
Collapse
|
37
|
Hamlett ED, Goetzl EJ, Ledreux A, Vasilevko V, Boger HA, LaRosa A, Clark D, Carroll SL, Carmona-Iragui M, Fortea J, Mufson EJ, Sabbagh M, Mohammed AH, Hartley D, Doran E, Lott IT, Granholm AC. Neuronal exosomes reveal Alzheimer's disease biomarkers in Down syndrome. Alzheimers Dement 2016; 13:541-549. [PMID: 27755974 DOI: 10.1016/j.jalz.2016.08.012] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/18/2016] [Accepted: 08/26/2016] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Individuals with Down syndrome (DS) exhibit Alzheimer's disease (AD) neuropathology and dementia early in life. Blood biomarkers of AD neuropathology would be valuable, as non-AD intellectual disabilities of DS and AD dementia overlap clinically. We hypothesized that elevations of amyloid β (Aβ) peptides and phosphorylated-tau in neuronal exosomes may document preclinical AD. METHODS AD neuropathogenic proteins Aβ1-42, P-T181-tau, and P-S396-tau were quantified by enzyme-linked immunosorbent assays in extracts of neuronal exosomes purified from blood of individuals with DS and age-matched controls. RESULTS Neuronal exosome levels of Aβ1-42, P-T181-tau, and P-S396-tau were significantly elevated in individuals with DS compared with age-matched controls at all ages beginning in childhood. No significant gender differences were observed. DISCUSSION These early increases in Aβ1-42, P-T181-tau, and P-S396-tau in individuals with DS may provide a basis for early intervention as targeted treatments become available.
Collapse
Affiliation(s)
- Eric D Hamlett
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; The Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Edward J Goetzl
- Geriatric Research Center of the Jewish Home of San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA
| | - Aurélie Ledreux
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; The Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Vitaly Vasilevko
- University of California, Irvine Institute for Memory Impairment and Neurological Disorders, Irvine, CA, USA
| | - Heather A Boger
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; The Center on Aging, Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Angela LaRosa
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - David Clark
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - María Carmona-Iragui
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau, Barcelona, Spain; Down Medical Center, Fundacío Catalana Síndrome de Down, Barcelona, Spain
| | - Juan Fortea
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau-Biomedical Research Institute Sant Pau, Barcelona, Spain; Down Medical Center, Fundacío Catalana Síndrome de Down, Barcelona, Spain
| | - Elliott J Mufson
- Barrow Neurological Institute, Department of Neurobiology, Phoenix, AZ, USA
| | - Marwan Sabbagh
- Barrow Neurological Institute, Department of Neurobiology, Phoenix, AZ, USA
| | - Abdul H Mohammed
- Department of Psychology, Linnaeus University, Växjo, Sweden; Center for Alzheimer Research, Karolinska Institutet, Huddinge, Sweden
| | | | - Eric Doran
- Department of Pediatrics, School of Medicine, University of California, Irvine, Orange, CA, USA
| | - Ira T Lott
- Department of Pediatrics, School of Medicine, University of California, Irvine, Orange, CA, USA
| | - Ann-Charlotte Granholm
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; The Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA; The Center on Aging, Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Center for Alzheimer Research, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
38
|
Arvio M, Luostarinen L. Down syndrome in adults: a 27-year follow-up of adaptive skills. Clin Genet 2016; 90:456-460. [PMID: 27067497 DOI: 10.1111/cge.12787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 11/29/2022]
Abstract
In 1988, we assessed the adaptive skills of 45 adults with Down syndrome (DS) (21 women and 24 men, age 20-58) with the Portage scale. Since then, we have followed them and also screened for signs of clinical dementia with the Present Psychiatric State - Learning Disabilities assessment. The mean adaptive age (AA) of the study group decreased with increasing age; the age of 35 being the turning point in the clinical course of DS. The mean AA was 4.4 years between ages 20 and 34, 3.4 years between ages 35 and 49, and 2.4 years between ages 50 and 66. Inter-individual variation was, however, large. Between ages 20 and 25, the AA of the study subjects ranged from 2.3 to 6 years; and after the age of 50, from 0.3 to 4.8 years. By the end of the study, all subjects showed signs of clinical dementia. These appeared most frequently as reduced self-care skills, loss of energy, forgetfulness, and impaired understanding. We found no connection between apolipoprotein E genotype and the clinical course of DS. We recommend follow-up of adaptive skills and screening for dementia signs in adults with DS.
Collapse
Affiliation(s)
- M Arvio
- Päijät-Häme Joint Municipal Authority, Lahti, Finland. .,Turku University Hospital, Turku, Finland.
| | - L Luostarinen
- Päijät-Häme Joint Municipal Authority, Lahti, Finland
| |
Collapse
|
39
|
Anderson-Mooney AJ, Schmitt FA, Head E, Lott IT, Heilman KM. Gait dyspraxia as a clinical marker of cognitive decline in Down syndrome: A review of theory and proposed mechanisms. Brain Cogn 2016; 104:48-57. [PMID: 26930369 PMCID: PMC4801771 DOI: 10.1016/j.bandc.2016.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 02/13/2016] [Accepted: 02/21/2016] [Indexed: 12/15/2022]
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual disability in children. With aging, DS is associated with an increased risk for Alzheimer's disease (AD). The development of AD neuropathology in individuals with DS can result in further disturbances in cognition and behavior and may significantly exacerbate caregiver burden. Early detection may allow for appropriate preparation by caregivers. Recent literature suggests that declines in gait may serve as an early marker of AD-related cognitive disorders; however, this relationship has not been examined in individuals with DS. The theory regarding gait dyspraxia and cognitive decline in the general population is reviewed, and potential applications to the population with individuals with DS are highlighted. Challenges and benefits in the line of inquiry are discussed. In particular, it appears that gait declines in aging individuals with DS may be associated with known declines in frontoparietal gray matter, development of AD-related pathology, and white matter losses in tracts critical to motor control. These changes are also potentially related to the cognitive and functional changes often observed during the same chronological period as gait declines in adults with DS. Gait declines may be an early marker of cognitive change, related to the development of underlying AD-related pathology, in individuals with DS. Future investigations in this area may provide insight into the clinical changes associated with development of AD pathology in both the population with DS and the general population, enhancing efforts for optimal patient and caregiver support and propelling investigations regarding safety/quality of life interventions and disease-modifying interventions.
Collapse
Affiliation(s)
- Amelia J Anderson-Mooney
- University of Kentucky College of Medicine, Department of Neurology, 740 S. Limestone, Suite B-101, Lexington, KY 40536, United States.
| | - Frederick A Schmitt
- University of Kentucky College of Medicine, Department of Neurology and Sanders-Brown Center on Aging, 800 S. Limestone, Room 312, Lexington, KY 40536, United States.
| | - Elizabeth Head
- University of Kentucky, Department of Molecular & Biomedical Pharmacology and Sanders-Brown Center on Aging, 800 S. Limestone, Room 203, Lexington, KY 40536, United States.
| | - Ira T Lott
- University of California - Irvine School of Medicine, Department of Pediatrics, Bldg 2 3rd Floor Rt 81, 101 The City Drive, Mail Code: 4482, Orange, CA 92668, United States.
| | - Kenneth M Heilman
- University of Florida College of Medicine, Department of Neurology, Room L3-100, McKnight Brain Institute, 1149 Newell Drive, Gainesville, FL 32611, United States.
| |
Collapse
|
40
|
Belichenko PV, Madani R, Rey-Bellet L, Pihlgren M, Becker A, Plassard A, Vuillermot S, Giriens V, Nosheny RL, Kleschevnikov AM, Valletta JS, Bengtsson SKS, Linke GR, Maloney MT, Hickman DT, Reis P, Granet A, Mlaki D, Lopez-Deber MP, Do L, Singhal N, Masliah E, Pearn ML, Pfeifer A, Muhs A, Mobley WC. An Anti-β-Amyloid Vaccine for Treating Cognitive Deficits in a Mouse Model of Down Syndrome. PLoS One 2016; 11:e0152471. [PMID: 27023444 PMCID: PMC4811554 DOI: 10.1371/journal.pone.0152471] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/15/2016] [Indexed: 11/18/2022] Open
Abstract
In Down syndrome (DS) or trisomy of chromosome 21, the β-amyloid (Aβ) peptide product of the amyloid precursor protein (APP) is present in excess. Evidence points to increased APP gene dose and Aβ as playing a critical role in cognitive difficulties experienced by people with DS. Particularly, Aβ is linked to the late-life emergence of dementia as associated with neuropathological markers of Alzheimer's disease (AD). At present, no treatment targets Aβ-related pathogenesis in people with DS. Herein we used a vaccine containing the Aβ 1-15 peptide embedded into liposomes together with the adjuvant monophosphoryl lipid A (MPLA). Ts65Dn mice, a model of DS, were immunized with the anti-Aβ vaccine at 5 months of age and were examined for cognitive measures at 8 months of age. The status of basal forebrain cholinergic neurons and brain levels of APP and its proteolytic products were measured. Immunization of Ts65Dn mice resulted in robust anti-Aβ IgG titers, demonstrating the ability of the vaccine to break self-tolerance. The vaccine-induced antibodies reacted with Aβ without detectable binding to either APP or its C-terminal fragments. Vaccination of Ts65Dn mice resulted in a modest, but non-significant reduction in brain Aβ levels relative to vehicle-treated Ts65Dn mice, resulting in similar levels of Aβ as diploid (2N) mice. Importantly, vaccinated Ts65Dn mice showed resolution of memory deficits in the novel object recognition and contextual fear conditioning tests, as well as reduction of cholinergic neuron atrophy. No treatment adverse effects were observed; vaccine did not result in inflammation, cellular infiltration, or hemorrhage. These data are the first to show that an anti-Aβ immunotherapeutic approach may act to target Aβ-related pathology in a mouse model of DS.
Collapse
Affiliation(s)
- Pavel V. Belichenko
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | | | | | - Ann Becker
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | | | | | - Rachel L. Nosheny
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Alexander M. Kleschevnikov
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Janice S. Valletta
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Sara K. S. Bengtsson
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Gordon R. Linke
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Michael T. Maloney
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, California, United States of America
| | | | | | | | | | | | - Long Do
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Nishant Singhal
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Eliezer Masliah
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Matthew L. Pearn
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | | | - William C. Mobley
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
41
|
Matthews DC, Lukic AS, Andrews RD, Marendic B, Brewer J, Rissman RA, Mosconi L, Strother SC, Wernick MN, Mobley WC, Ness S, Schmidt ME, Rafii MS. Dissociation of Down syndrome and Alzheimer's disease effects with imaging. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2016. [PMID: 28642933 PMCID: PMC5477635 DOI: 10.1016/j.trci.2016.02.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction Down Syndrome (DS) adults experience accumulation of Alzheimer's disease (AD)–like amyloid plaques and tangles and a high incidence of dementia and could provide an enriched population to study AD-targeted treatments. However, to evaluate effects of therapeutic intervention, it is necessary to dissociate the contributions of DS and AD from overall phenotype. Imaging biomarkers offer the potential to characterize and stratify patients who will worsen clinically but have yielded mixed findings in DS subjects. Methods We evaluated 18F fluorodeoxyglucose positron emission tomography (PET), florbetapir PET, and structural magnetic resonance (sMR) image data from 12 nondemented DS adults using advanced multivariate machine learning methods. Results Our results showed distinctive patterns of glucose metabolism and brain volume enabling dissociation of DS and AD effects. AD-like pattern expression corresponded to amyloid burden and clinical measures. Discussion These findings lay groundwork to enable AD clinical trials with characterization and disease-specific tracking of DS adults.
Collapse
Affiliation(s)
| | | | | | | | - James Brewer
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Robert A Rissman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Lisa Mosconi
- Department of Psychiatry, New York University Langone School of Medicine, New York, NY, USA
| | - Stephen C Strother
- ADM Diagnostics, Northbrook, IL, USA.,Rotman Research Institute, Baycrest Hospital and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Miles N Wernick
- ADM Diagnostics, Northbrook, IL, USA.,Departments of Electrical and Computer Engineering and Biomedical Engineering, Medical Imaging Research Center, Illinois Institute of Technology, Chicago, IL, USA
| | - William C Mobley
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Seth Ness
- Janssen Research and Development LLC, Raritan, NJ, USA
| | | | - Michael S Rafii
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
42
|
Kamer AR, Fortea JO, Videla S, Mayoral A, Janal M, Carmona-Iragui M, Benejam B, Craig RG, Saxena D, Corby P, Glodzik L, Annam KRC, Robbins M, de Leon MJ. Periodontal disease's contribution to Alzheimer's disease progression in Down syndrome. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2016; 2:49-57. [PMID: 27239536 PMCID: PMC4879643 DOI: 10.1016/j.dadm.2016.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
People with Down syndrome (DS) are at an increased risk for Alzheimer's disease (AD). After 60 years of age, >50% of DS subjects acquire dementia. Nevertheless, the age of onset is highly variable possibly because of both genetic and environmental factors. Genetics cannot be modified, but environmental risk factors present a potentially relevant intervention for DS persons at risk for AD. Among them, inflammation, important in AD of DS type, is potential target. Consistent with this hypothesis, chronic peripheral inflammation and infections may contribute to AD pathogenesis in DS. People with DS have an aggressive form of periodontitis characterized by rapid progression, significant bacterial and inflammatory burden, and an onset as early as 6 years of age. This review offers a hypothetical mechanistic link between periodontitis and AD in the DS population. Because periodontitis is a treatable condition, it may be a readily modifiable risk factor for AD.
Collapse
Affiliation(s)
- Angela R Kamer
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, New York, NY, USA; Department of Psychiatry, Center for Brain Health, School of Medicine, New York, NY, USA
| | - Juan O Fortea
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autónoma de Barcelona, Barcelona, Spain; Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Sebastià Videla
- Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Angela Mayoral
- Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Dentistry School Universitat Internacional de Catalunya, Sant Cugat del Vallés, Barcelona, Spain
| | - Malvin Janal
- Department of Epidemiology, College of Dentistry, New York University, New York, NY, USA
| | - Maria Carmona-Iragui
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autónoma de Barcelona, Barcelona, Spain; Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Bessy Benejam
- Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Ronald G Craig
- Department of Basic Sciences and Craniofacial Biology, College of Dentistry, New York University, New York, NY, USA
| | - Deepak Saxena
- Department of Basic Sciences and Craniofacial Biology, College of Dentistry, New York University, New York, NY, USA
| | - Patricia Corby
- Department of Psychiatry, Center for Brain Health, School of Medicine, New York, NY, USA
| | - Lidia Glodzik
- Department of Psychiatry, Center for Brain Health, School of Medicine, New York, NY, USA
| | - Kumar Raghava Chowdary Annam
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, New York, NY, USA
| | - Miriam Robbins
- Department of Dental Medicine, Winthrop University Hospital, Mineola, NY, USA
| | - Mony J de Leon
- Department of Psychiatry, Center for Brain Health, School of Medicine, New York, NY, USA
| |
Collapse
|
43
|
Hamlett ED, Boger HA, Ledreux A, Kelley CM, Mufson EJ, Falangola MF, Guilfoyle DN, Nixon RA, Patterson D, Duval N, Granholm ACE. Cognitive Impairment, Neuroimaging, and Alzheimer Neuropathology in Mouse Models of Down Syndrome. Curr Alzheimer Res 2016; 13:35-52. [PMID: 26391050 PMCID: PMC5034871 DOI: 10.2174/1567205012666150921095505] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 08/08/2015] [Accepted: 08/20/2015] [Indexed: 11/22/2022]
Abstract
Down syndrome (DS) is the most common non-lethal genetic condition that affects approximately 1 in 700 births in the United States of America. DS is characterized by complete or segmental chromosome 21 trisomy, which leads to variable intellectual disabilities, progressive memory loss, and accelerated neurodegeneration with age. During the last three decades, people with DS have experienced a doubling of life expectancy due to progress in treatment of medical comorbidities, which has allowed this population to reach the age when they develop early onset Alzheimer's disease (AD). Individuals with DS develop cognitive and pathological hallmarks of AD in their fourth or fifth decade, and are currently lacking successful prevention or treatment options for dementia. The profound memory deficits associated with DS-related AD (DS-AD) have been associated with degeneration of several neuronal populations, but mechanisms of neurodegeneration are largely unexplored. The most successful animal model for DS is the Ts65Dn mouse, but several new models have also been developed. In the current review, we discuss recent findings and potential treatment options for the management of memory loss and AD neuropathology in DS mouse models. We also review agerelated neuropathology, and recent findings from neuroimaging studies. The validation of appropriate DS mouse models that mimic neurodegeneration and memory loss in humans with DS can be valuable in the study of novel preventative and treatment interventions, and may be helpful in pinpointing gene-gene interactions as well as specific gene segments involved in neurodegeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ann-Charlotte E Granholm
- Department Neurosciences, Director, Center on Aging, Medical Univ. South Carolina, Basic Science Bldg, Room 403, 173 Ashley Avenue, Charleston, SC 29425, USA.
| |
Collapse
|
44
|
Nizetic D, Chen CL, Hong W, Koo EH. Inter-Dependent Mechanisms Behind Cognitive Dysfunction, Vascular Biology and Alzheimer's Dementia in Down Syndrome: Multi-Faceted Roles of APP. Front Behav Neurosci 2015; 9:299. [PMID: 26648852 PMCID: PMC4664698 DOI: 10.3389/fnbeh.2015.00299] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/27/2015] [Indexed: 01/05/2023] Open
Abstract
People with Down syndrome (DS) virtually all develop intellectual disability (ID) of varying degree of severity, and also have a high risk of early Alzheimer's disease (AD). ID prior to the onset of dementia, and its relationship to the onset of dementia in DS is a complex phenomenon influenced by many factors, and scarcely understood. Unraveling the causative factors and modulators of these processes remains a challenge, with potential to be informative for both ID and AD, for the development of early biomarkers and/or therapeutic approaches. We review the potential relative and inter-connected roles of the chromosome 21 gene for amyloid precursor protein (APP), in both pathological conditions. Rare non-DS people with duplication of APP (dupAPP) get familial early onset AD (FEOAD) with virtually 100% penetrance and prominent cerebrovascular pathology, but don't suffer from ID before dementia onset. All of these features appear to be radically different in DS. On the other hand, rare individuals with partial trisomy 21 (T21) (with APP, but not DS-critical region in trisomy) have been described having ID. Likewise, partial T21 DS (without APP trisomy) show a range of ID, but no AD pathology. We review the multi-faceted roles of APP that might affect cognitive functioning. Given the fact that both Aβ secretion and synaptic maturation/plasticity are dependent on neuronal activity, we explore how this conflicting inter-dependency might affect cognitive pathogenesis in a dynamic way in DS, throughout the lifespan of an individual.
Collapse
Affiliation(s)
- Dean Nizetic
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore Singapore ; The LonDownS Consortium, Wellcome Trust London, UK ; The Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London London, UK
| | - Christopher L Chen
- Department of Psychological Medicine and Memory Aging and Cognition Centre, National University Health System, Singapore Singapore ; Department of Pharmacology, National University of Singapore, Singapore Singapore
| | - Wanjin Hong
- Agency for Science, Technology and Research (AStar), Institute of Molecular Cell Biology, Singapore Singapore
| | - Edward H Koo
- Departments of Medicine and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore Singapore ; Department of Neurosciences, University of California, San Diego San Diego, CA, USA
| |
Collapse
|
45
|
Cerebrovascular contributions to aging and Alzheimer's disease in Down syndrome. Biochim Biophys Acta Mol Basis Dis 2015; 1862:909-14. [PMID: 26593849 DOI: 10.1016/j.bbadis.2015.11.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 11/22/2022]
Abstract
Down syndrome (DS) is a common cause of intellectual disability and is also associated with early age of onset of Alzheimer's disease (AD). Due to an extra copy of chromosome 21, most adults over 40years old with DS have beta-amyloid plaques as a result of overexpression of the amyloid precursor protein. Cerebrovascular pathology may also be a significant contributor to neuropathology observed in the brains of adults with DS. This review describes the features of cardiovascular dysfunction and cerebrovascular pathology in DS that may be modifiable risk factors and thus targets for interventions. We will describe cerebrovascular pathology, the role of co-morbidities, imaging studies indicating vascular pathology and the possible consequences. It is clear that our understanding of aging and AD in people with DS will benefit from further studies to determine the role that cerebrovascular dysfunction contributes to cognitive health. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
|
46
|
Covelli V, Meucci P, Raggi A, Fontana G, Leonardi M. A Pilot Study on Function and Disability of Aging People with Down Syndrome in Italy. JOURNAL OF POLICY AND PRACTICE IN INTELLECTUAL DISABILITIES 2015. [DOI: 10.1111/jppi.12129] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
| | - Paolo Meucci
- Fondazione IRCCS Istituto Neurologico Carlo Besta; Milan Italy
| | - Alberto Raggi
- Fondazione IRCCS Istituto Neurologico Carlo Besta; Milan Italy
| | | | | |
Collapse
|
47
|
Schupf N, Lee A, Park N, Dang LH, Pang D, Yale A, Oh DKT, Krinsky-McHale SJ, Jenkins EC, Luchsinger JA, Zigman WB, Silverman W, Tycko B, Kisselev S, Clark L, Lee JH. Candidate genes for Alzheimer's disease are associated with individual differences in plasma levels of beta amyloid peptides in adults with Down syndrome. Neurobiol Aging 2015; 36:2907.e1-10. [PMID: 26166206 DOI: 10.1016/j.neurobiolaging.2015.06.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 06/08/2015] [Accepted: 06/14/2015] [Indexed: 01/08/2023]
Abstract
We examined the contribution of candidates genes for Alzheimer's disease (AD) to individual differences in levels of beta amyloid peptides in adults with Down syndrom, a population at high risk for AD. Participants were 254 non-demented adults with Down syndrome, 30-78 years of age. Genomic deoxyribonucleic acid was genotyped using an Illumina GoldenGate custom array. We used linear regression to examine differences in levels of Aβ peptides associated with the number of risk alleles, adjusting for age, sex, level of intellectual disability, race and/or ethnicity, and the presence of the APOE ε4 allele. For Aβ42 levels, the strongest gene-wise association was found for a single nucleotide polymorphism (SNP) on CAHLM1; for Aβ40 levels, the strongest gene-wise associations were found for SNPs in IDE and SOD1, while the strongest gene-wise associations with levels of the Aβ42/Aβ40 ratio were found for SNPs in SORCS1. Broadly classified, variants in these genes may influence amyloid precursor protein processing (CALHM1, IDE), vesicular trafficking (SORCS1), and response to oxidative stress (SOD1).
Collapse
Affiliation(s)
- Nicole Schupf
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; G.H. Sergievsky Center, New York, NY, USA; Department of Epidemiology, Columbia University Medical Center, New York, NY, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY, USA.
| | - Annie Lee
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Naeun Park
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Lam-Ha Dang
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Deborah Pang
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Alexander Yale
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - David Kyung-Taek Oh
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sharon J Krinsky-McHale
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Edmund C Jenkins
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - José A Luchsinger
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Warren B Zigman
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wayne Silverman
- Kennedy Krieger Institute and Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin Tycko
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sergey Kisselev
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Lorraine Clark
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Joseph H Lee
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; G.H. Sergievsky Center, New York, NY, USA; Department of Epidemiology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
48
|
Romano A, Cornia R, Moraschi M, Bozzao A, Chiacchiararelli L, Coppola V, Iani C, Stella G, Albertini G, Pierallini A. Age-Related Cortical Thickness Reduction in Non-Demented Down's Syndrome Subjects. J Neuroimaging 2015; 26:95-102. [DOI: 10.1111/jon.12259] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 03/09/2015] [Accepted: 04/04/2015] [Indexed: 11/27/2022] Open
Affiliation(s)
- Andrea Romano
- San Raffaele Foundation, Rehabilitation Facility; Merit Project RBNE08E8CZ
| | - Riccardo Cornia
- University of Modena and Reggio Emilia, Departments of Biomedical; Metabolic and Neurosciences
| | - Marta Moraschi
- San Raffaele Foundation, Rehabilitation Facility; Merit Project RBNE08E8CZ
| | - Alessandro Bozzao
- University Sapienza, NESMOS, Department of Neuroradiology; S.Andrea Hospital
| | | | - Valeria Coppola
- University Sapienza, NESMOS, Department of Neuroradiology; S.Andrea Hospital
| | - Cristina Iani
- University of Modena and Reggio Emilia; Department of Communication and Economy
| | - Giacomo Stella
- University of Modena and Reggio Emilia, Departments of Biomedical; Metabolic and Neurosciences
| | | | | |
Collapse
|
49
|
Gruszecka A, Kopczyński P, Cudziło D, Lipińska N, Romaniuk A, Barczak W, Rozwadowska N, Totoń E, Rubiś B. Telomere shortening in Down syndrome patients--when does it start? DNA Cell Biol 2015; 34:412-7. [PMID: 25786194 DOI: 10.1089/dna.2014.2746] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Down syndrome (DS) is one of the most common aneuploidy. In general population, its prevalence is 1:600-1:800 live births. It is caused by a trisomy of chromosome 21. DS is phenotypically manifested by premature aging, upward slant to the eyes, epicanthus, flattened face, and poor muscle tone. In addition to physical changes, this syndrome is characterized by early onset of diseases specific to old age, such as Alzheimer's disease, vision and hearing problems, and precocious menopause. Since DS symptoms include premature aging, the shortening of telomeres might be one of the markers of cellular aging. Consequently, the aim of the study was to determine the length of the telomeres in leukocytes from the blood of juvenile patients with DS (n=68) compared to an age-matched control group (n=56) and also to determine the diagnostic or predictive value for this parameter. We show that, for the first time, in juveniles, the average relative telomere length in studied subjects is significantly longer than in the control group (50.46 vs. 40.56, respectively arbitrary units [AU]; p=0.0026). The results provide interesting basis for further research to determine the causes and consequences of telomere maintaining and the dynamics of this process in patients with DS.
Collapse
Affiliation(s)
- Aleksandra Gruszecka
- 1Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan, Poland
| | - Przemysław Kopczyński
- 2Centre for Orthodontic Mini-implants at the Department and Clinic of Maxillofacial Orthopedics and Orthodontics, Poznan University of Medical Sciences, Poznan, Poland
| | - Dorota Cudziło
- 3Orthodontic Department, Institute of Mother and Child, Warsaw, Poland
| | - Natalia Lipińska
- 1Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan, Poland
| | - Aleksandra Romaniuk
- 1Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Barczak
- 4Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan, Poland.,5Radiobiology Laboratory, Department of Medical Physics, The Greater Poland Cancer Centre, Poznan, Poland
| | | | - Ewa Totoń
- 1Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan, Poland
| | - Błażej Rubiś
- 1Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
50
|
Romano A, Moraschi M, Cornia R, Bozzao A, Gagliardo O, Chiacchiararelli L, Iani C, Stella G, Albertini G, Pierallini A. Age effects on cortical thickness in young Down's syndrome subjects: a cross-sectional gender study. Neuroradiology 2015; 57:401-11. [PMID: 25560246 DOI: 10.1007/s00234-014-1482-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/15/2014] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The aim of this study was to determine differences in the characteristic pattern of age-related cortical thinning in men and women with Down's syndrome (DS) by means of MRI and automatic cortical thickness measurements and a cross-sectional design, in a large cohort of young subjects. METHODS Eighty-four subjects with DS, 30 females (11-35 years, mean age ± SD = 22.8 ± 5.9) and 54 males (11-35 years, mean age ± SD = 21.5 ± 6.5), were examined using a 1.5-T scanner. MRI-based quantification of cortical thickness was performed using FreeSurfer software package. For all subjects participating in the study, the Pearson product-moment correlation coefficient between age and mean cortical thickness values has been evaluated. RESULTS A significant negative correlation between cortical thickness and age was found in female DS subjects, predominantly in frontal and parietal lobes, bilaterally. In male DS subjects, a significant negative correlation between cortical thickness and age was found in the right fronto-temporal lobes and cingulate regions. Whole brain mean cortical thickness values were significantly negative correlated with age only in female DS subjects. CONCLUSIONS Females with Down's syndrome showed a strong correlation between cortical thickness and age, already in early age. We suggest that the cognitive impairment due to hormonal deficit in the postmenopausal period could be emphasized by the early structural decline of gray matter in female DS subjects.
Collapse
Affiliation(s)
- Andrea Romano
- San Raffaele Foundation Rome, Rehabilitation Facility Ceglie Messapica, Merit Project RBNE08E8CZ, Via di Grottarossa 1035, 00189, Rome, Italy,
| | | | | | | | | | | | | | | | | | | |
Collapse
|