1
|
Singh M, Ali H, Renuka Jyothi S, Kaur I, Kumar S, Sharma N, Siva Prasad GV, Pramanik A, Hassan Almalki W, Imran M. Tau proteins and senescent Cells: Targeting aging pathways in Alzheimer's disease. Brain Res 2024; 1844:149165. [PMID: 39155034 DOI: 10.1016/j.brainres.2024.149165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by abnormal accumulation of tau proteins and amyloid-β, leading to neuronal death and cognitive impairment. Recent studies have implicated aging pathways, including dysregulation of tau and cellular senescence in AD pathogenesis. In AD brains, tau protein, which normally stabilizes microtubules, becomes hyperphosphorylated and forms insoluble neurofibrillary tangles. These tau aggregates impair neuronal function and are propagated across the brain's neurocircuitry. Meanwhile, the number of senescent cells accumulating in the aging brain is rising, releasing a pro-inflammatory SASP responsible for neuroinflammation and neurodegeneration. This review explores potential therapeutic interventions for AD targeting tau protein and senescent cells, and tau -directed compounds, senolytics, eliminating senescent cells, and agents that modulate the SASP-senomodulators. Ultimately, a combined approach that incorporates tau-directed medications and targeted senescent cell-based therapies holds promise for reducing the harmful impact of AD's shared aging pathways.
Collapse
Affiliation(s)
- Mahaveer Singh
- School of Pharmacy and Technology Management, SVKMs NMIMS University, Shirpur campus, Maharastra India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali 140307, Punjab, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
2
|
Perry JC, Vann SD. Reduction in neurons immunoreactive for calcium-binding proteins in the anteroventral thalamic nuclei of individuals with Down syndrome. Neuroscience 2024; 557:56-66. [PMID: 39127343 DOI: 10.1016/j.neuroscience.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/26/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
The anterior thalamic nuclei are important for cognition, and memory in particular. However, little is known about how the anterior thalamic nuclei are affected in many neurological disorders partly due to difficulties in selective segmentation in in vivo scans, due to their size and location. Post-mortem studies, therefore, remain a valuable source of information about the status of the anterior thalamic nuclei. We used post-mortem tissue to assess the status of the anteroventral thalamic nucleus in Down syndrome using samples from males and females ranging from 22-65 years in age and comparing to tissue from age matched controls. As expected, there was increased beta-amyloid plaque expression in the Down syndrome group. While there was a significant increase in neuronal density in the Down syndrome group, the values showed more variation consistent with a heterogeneous population. The surface area of the anteroventral thalamic nucleus was smaller in the Down syndrome group suggesting the increased neuronal density was due to greater neuronal packing but likely fewer overall neurons. There was a marked reduction in the proportion of neurons immunoreactive for the calcium-binding proteins calbindin, calretinin, and parvalbumin in individuals with Down syndrome. These findings highlight the vulnerability of calcium-binding proteins in the anteroventral nucleus in Down syndrome, which could both be driven by, and exacerbate, Alzheimer-related pathology in this region.
Collapse
Affiliation(s)
- James C Perry
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| | - Seralynne D Vann
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK.
| |
Collapse
|
3
|
Sárkány B, Dávid C, Hortobágyi T, Gombás P, Somogyi P, Acsády L, Viney TJ. Early and selective localization of tau filaments to glutamatergic subcellular domains within the human anterodorsal thalamus. Acta Neuropathol 2024; 147:98. [PMID: 38861157 PMCID: PMC11166832 DOI: 10.1007/s00401-024-02749-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/21/2024] [Accepted: 06/01/2024] [Indexed: 06/12/2024]
Abstract
Widespread cortical accumulation of misfolded pathological tau proteins (ptau) in the form of paired helical filaments is a major hallmark of Alzheimer's disease. Subcellular localization of ptau at various stages of disease progression is likely to be informative of the cellular mechanisms involving its spread. Here, we found that the density of ptau within several distinct rostral thalamic nuclei in post-mortem human tissue (n = 25 cases) increased with the disease stage, with the anterodorsal nucleus (ADn) consistently being the most affected. In the ADn, ptau-positive elements were present already in the pre-cortical (Braak 0) stage. Tau pathology preferentially affected the calretinin-expressing subpopulation of glutamatergic neurons in the ADn. At the subcellular level, we detected ptau immunoreactivity in ADn cell bodies, dendrites, and in a specialized type of presynaptic terminal that expresses vesicular glutamate transporter 2 (vGLUT2) and likely originates from the mammillary body. The ptau-containing terminals displayed signs of degeneration, including endosomal/lysosomal organelles. In contrast, corticothalamic axon terminals lacked ptau. The data demonstrate the involvement of a specific cell population in ADn at the onset of the disease. The presence of ptau in subcortical glutamatergic presynaptic terminals supports hypotheses about the transsynaptic spread of tau selectively affecting specialized axonal pathways.
Collapse
Affiliation(s)
- Barbara Sárkány
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK.
| | - Csaba Dávid
- Lendület Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, 1083, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Tibor Hortobágyi
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Péter Gombás
- Department of Pathology, Szt. Borbála Hospital, Tatabánya, 2800, Hungary
| | - Peter Somogyi
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - László Acsády
- Lendület Laboratory of Thalamus Research, Institute of Experimental Medicine, Budapest, 1083, Hungary.
| | - Tim J Viney
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK.
| |
Collapse
|
4
|
Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Bryukhovetskiy I, Manzhulo I, Wiklund L, Sharma HS. Sleep deprivation enhances amyloid beta peptide, p-tau and serotonin in the brain: Neuroprotective effects of nanowired delivery of cerebrolysin with monoclonal antibodies to amyloid beta peptide, p-tau and serotonin. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 171:125-162. [PMID: 37783554 DOI: 10.1016/bs.irn.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Sleep deprivation is quite frequent in military during combat, intelligence gathering or peacekeeping operations. Even one night of sleep deprivation leads to accumulation of amyloid beta peptide burden that would lead to precipitation of Alzheimer's disease over the years. Thus, efforts are needed to slow down or neutralize accumulation of amyloid beta peptide (AβP) and associated Alzheimer's disease brain pathology including phosphorylated tau (p-tau) within the brain fluid environment. Sleep deprivation also alters serotonin (5-hydroxytryptamine) metabolism in the brain microenvironment and impair upregulation of several neurotrophic factors. Thus, blockade or neutralization of AβP, p-tau and serotonin in sleep deprivation may attenuate brain pathology. In this investigation this hypothesis is examined using nanodelivery of cerebrolysin- a balanced composition of several neurotrophic factors and active peptide fragments together with monoclonal antibodies against AβP, p-tau and serotonin (5-hydroxytryptamine, 5-HT). Our observations suggest that sleep deprivation induced pathophysiology is significantly reduced following nanodelivery of cerebrolysin together with monoclonal antibodies to AβP, p-tau and 5-HT, not reported earlier.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston MA, USA
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
5
|
Allwright M, Mundell HD, McCorkindale AN, Lindley RI, Austin PJ, Guennewig B, Sutherland GT. Ranking the risk factors for Alzheimer's disease; findings from the UK Biobank study. AGING BRAIN 2023; 3:100081. [PMID: 37384134 PMCID: PMC10293768 DOI: 10.1016/j.nbas.2023.100081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 06/30/2023] Open
Abstract
Background The cause of the most common form of dementia, sporadic Alzheimer's disease (AD), remains unknown. This may reflect insufficiently powered studies to date for this multi-factorial disorder. The UK Biobank dataset presents a unique opportunity to rank known risk factors and determine novel variables. Methods A custom machine learning approach for high dimensionality data was applied to explore prospectively associations between AD in a sub-cohort of 156,209 UK Biobank participants aged 60-70 including more than 2,090 who were subsequently diagnosed with AD. Results After the possession of the APOE4 allele, the next highest ranked risk factors were other genetic variants within the TOMM40-APOE-APOC1 locus. When stratified by their apolipoprotein epsilon 4 (APOE4) carrier status, the most prominent risk factors in carriers were AST:ALT ratio, the "number of treatments/ medications" taken as well as "time spent in hospital" while protection was conferred by "Sleeplessness/Insomnia". In non-APOE carriers, lower socioeconomic status and fewer years of education were highly ranked but effect sizes were small relative to APOE4 carriers. Conclusions Possession of the APOE4 allele was confirmed as the most important risk factor in AD. Other TOMM40-APOE-APOC1 locus variants further moderate the risk of AD in APOE4 carriers. Liver pathology is a novel risk factor in APOE4 carriers while "Sleeplessness/Insomnia" is protective in AD irrespective of APOE4 status. Other factors such as "Number of treatments/ medications" suggest that multimorbidity is an important risk factor for AD. Future treatments aimed at co-morbidities, including liver disease, may concomitantly lower the risk of sporadic AD.
Collapse
Affiliation(s)
- Michael Allwright
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Hamish D Mundell
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - Andrew N McCorkindale
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - Richard I. Lindley
- Westmead Applied Research Centre, Sydney Medical School, University of Sydney, NSW 2006 and George Institute for Global Health, Newtown, NSW 2042, Australia
| | - Paul J. Austin
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Boris Guennewig
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Greg T Sutherland
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
6
|
Semikasev E, Ahlemeyer B, Acker T, Schänzer A, Baumgart-Vogt E. Rise and fall of peroxisomes during Alzheimer´s disease: a pilot study in human brains. Acta Neuropathol Commun 2023; 11:80. [PMID: 37170361 PMCID: PMC10176950 DOI: 10.1186/s40478-023-01567-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Peroxisomes are eukaryotic organelles that rapidly change in number depending on the metabolic requirement of distinct cell types and tissues. In the brain, these organelles are essential for neuronal migration and myelination during development and their dysfunction is associated with age-related neurodegenerative diseases. Except for one study analysing ABCD3-positive peroxisomes in neurons of the frontal neocortex of Alzheimer disease (AD) patients, no data on other brain regions or peroxisomal proteins are available. In the present morphometric study, we quantified peroxisomes labelled with PEX14, a metabolism-independent peroxisome marker, in 13 different brain areas of 8 patients each either with low, intermediate or high AD neuropathological changes compared to 10 control patients. Classification of patient samples was based on the official ABC score. During AD-stage progression, the peroxisome density decreased in the area entorhinalis, parietal/occipital neocortex and cerebellum, it increased and in later AD-stage patients decreased in the subiculum and hippocampal CA3 region, frontal neocortex and pontine gray and it remained unchanged in the gyrus dentatus, temporal neocortex, striatum and inferior olive. Moreover, we investigated the density of catalase-positive peroxisomes in a subset of patients (> 80 years), focussing on regions with significant alterations of PEX14-positive peroxisomes. In hippocampal neurons, only one third of all peroxisomes contained detectable levels of catalase exhibiting constant density at all AD stages. Whereas the density of all peroxisomes in neocortical neurons was only half of the one of the hippocampus, two thirds of them were catalase-positive exhibiting increased levels at higher ABC scores. In conclusion, we observed spatiotemporal differences in the response of peroxisomes to different stages of AD-associated pathologies.
Collapse
Affiliation(s)
- Eugen Semikasev
- Division of Medical Cell Biology, Institute for Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, 35385, Giessen, Germany
- Department of Neurosurgery, University Hospital of Giessen, Klinikstr. 33, 35392, Giessen, Germany
| | - Barbara Ahlemeyer
- Division of Medical Cell Biology, Institute for Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, 35385, Giessen, Germany.
| | - Till Acker
- Institute of Neuropathology, Justus-Liebig University, Arndtstr. 16, 35392, Giessen, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig University, Arndtstr. 16, 35392, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Division of Medical Cell Biology, Institute for Anatomy and Cell Biology, Justus-Liebig University, Aulweg 123, 35385, Giessen, Germany.
| |
Collapse
|
7
|
Neumann P, Lenz DE, Streit WJ, Bechmann I. Is microglial dystrophy a form of cellular senescence? An analysis of senescence markers in the aged human brain. Glia 2023; 71:377-390. [PMID: 36286188 DOI: 10.1002/glia.24282] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 01/08/2023]
Abstract
Aging can cause morphological transformation in human microglia indicative of cell senescence, termed microglial dystrophy. However, cellular senescence is characterized by additional changes, such as an irregular cell cycle arrest, and a variety of metabolic and molecular changes including a senescence-associated secretory phenotype, dysfunction of degradation mechanisms, and altered DNA damage response. Here, we tested whether dystrophic microglia display customary markers of cell senescence by performing double and triple staining in sections of the temporal lobe and brain stem from 14 humans. We found that markers related to oxidative damage, such as upregulation of 8-hydroxy-2'-deoxyguanosine (8-OHdG), hemeoxygenase-1 (HO-1), and y-H2AX, as well as inclusion of lipofuscin, do not or only exceptionally colocalize with dystrophic microglia. Further, we did not observe a decline in lamin B1 around nuclear laminae in either dystrophic or ramified microglia within the same microscopic field. Only ferritin expression, which is known to increase with aging in CNS microglia, was frequently observed in dystrophic, but rarely in ramified microglial cells. We conclude that neither dystrophic nor ramified microglia in human brain exhibit significant expression of conventional senescence markers associated with oxidative stress, and that ferritin is the dominant immunophenotypic change related to microglial aging. We suggest that multiple pathogenic mechanisms other than those driving cellular senescence contribute to dystrophic transformation of microglia.
Collapse
Affiliation(s)
| | - Dana E Lenz
- Institute of Anatomy, Universität Leipzig, Leipzig, Germany
| | - Wolfgang J Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Ingo Bechmann
- Institute of Anatomy, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
8
|
Otero-Garcia M, Mahajani SU, Wakhloo D, Tang W, Xue YQ, Morabito S, Pan J, Oberhauser J, Madira AE, Shakouri T, Deng Y, Allison T, He Z, Lowry WE, Kawaguchi R, Swarup V, Cobos I. Molecular signatures underlying neurofibrillary tangle susceptibility in Alzheimer's disease. Neuron 2022; 110:2929-2948.e8. [PMID: 35882228 PMCID: PMC9509477 DOI: 10.1016/j.neuron.2022.06.021] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 03/08/2022] [Accepted: 06/27/2022] [Indexed: 01/01/2023]
Abstract
Tau aggregation in neurofibrillary tangles (NFTs) is closely associated with neurodegeneration and cognitive decline in Alzheimer's disease (AD). However, the molecular signatures that distinguish between aggregation-prone and aggregation-resistant cell states are unknown. We developed methods for the high-throughput isolation and transcriptome profiling of single somas with NFTs from the human AD brain, quantified the susceptibility of 20 neocortical subtypes for NFT formation and death, and identified both shared and cell-type-specific signatures. NFT-bearing neurons shared a marked upregulation of synaptic transmission-related genes, including a core set of 63 genes enriched for synaptic vesicle cycling. Oxidative phosphorylation and mitochondrial dysfunction were highly cell-type dependent. Apoptosis was only modestly enriched, and the susceptibilities of NFT-bearing and NFT-free neurons for death were highly similar. Our analysis suggests that NFTs represent cell-type-specific responses to stress and synaptic dysfunction. We provide a resource for biomarker discovery and the investigation of tau-dependent and tau-independent mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Marcos Otero-Garcia
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sameehan U Mahajani
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Debia Wakhloo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weijing Tang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yue-Qiang Xue
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Samuel Morabito
- Mathematical, Computational and Systems Biology Program, University of California, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Jie Pan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jane Oberhauser
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Angela E Madira
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tamara Shakouri
- Department of Pathology, University of California, Los Angeles, CA 90095, USA
| | - Yongning Deng
- Department of Pathology, University of California, Los Angeles, CA 90095, USA; Department of Neurology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Thomas Allison
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095, USA
| | - Zihuai He
- Department Neurology and Neurological Sciences and Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - William E Lowry
- Department of Molecular Cell and Developmental Biology, Broad Center for Regenerative Medicine and Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Riki Kawaguchi
- Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Vivek Swarup
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA; Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Inma Cobos
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
9
|
Kuchcinski G, Patin L, Lopes R, Leroy M, Delbeuck X, Rollin-Sillaire A, Lebouvier T, Wang Y, Spincemaille P, Tourdias T, Hacein-Bey L, Devos D, Pasquier F, Leclerc X, Pruvo JP, Verclytte S. Quantitative susceptibility mapping demonstrates different patterns of iron overload in subtypes of early-onset Alzheimer's disease. Eur Radiol 2022; 33:184-195. [PMID: 35881183 DOI: 10.1007/s00330-022-09014-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES We aimed to define brain iron distribution patterns in subtypes of early-onset Alzheimer's disease (EOAD) by the use of quantitative susceptibility mapping (QSM). METHODS EOAD patients prospectively underwent MRI on a 3-T scanner and concomitant clinical and neuropsychological evaluation, between 2016 and 2019. An age-matched control group was constituted of cognitively healthy participants at risk of developing AD. Volumetry of the hippocampus and cerebral cortex was performed on 3DT1 images. EOAD subtypes were defined according to the hippocampal to cortical volume ratio (HV:CTV). Limbic-predominant atrophy (LPMRI) is referred to HV:CTV ratios below the 25th percentile, hippocampal-sparing (HpSpMRI) above the 75th percentile, and typical-AD between the 25th and 75th percentile. Brain iron was estimated using QSM. QSM analyses were made voxel-wise and in 7 regions of interest within deep gray nuclei and limbic structures. Iron distribution in EOAD subtypes and controls was compared using an ANOVA. RESULTS Sixty-eight EOAD patients and 43 controls were evaluated. QSM values were significantly higher in deep gray nuclei (p < 0.001) and limbic structures (p = 0.04) of EOAD patients compared to controls. Among EOAD subtypes, HpSpMRI had the highest QSM values in deep gray nuclei (p < 0.001) whereas the highest QSM values in limbic structures were observed in LPMRI (p = 0.005). QSM in deep gray nuclei had an AUC = 0.92 in discriminating HpSpMRI and controls. CONCLUSIONS In early-onset Alzheimer's disease patients, we observed significant variations of iron distribution reflecting the pattern of brain atrophy. Iron overload in deep gray nuclei could help to identify patients with atypical presentation of Alzheimer's disease. KEY POINTS • In early-onset AD patients, QSM indicated a significant brain iron overload in comparison with age-matched controls. • Iron load in limbic structures was higher in participants with limbic-predominant subtype. • Iron load in deep nuclei was more important in participants with hippocampal-sparing subtype.
Collapse
Affiliation(s)
- Grégory Kuchcinski
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France. .,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France. .,Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France.
| | - Lucas Patin
- Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France
| | - Renaud Lopes
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France
| | - Mélanie Leroy
- Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France
| | - Xavier Delbeuck
- Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France
| | - Adeline Rollin-Sillaire
- Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France.,Department of Neurology, CHU Lille, F-59000, Lille, France
| | - Thibaud Lebouvier
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France.,Department of Neurology, CHU Lille, F-59000, Lille, France
| | - Yi Wang
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Thomas Tourdias
- Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, F-33000, Bordeaux, France.,Neurocentre Magendie, Inserm, U1215, Université de Bordeaux, F-33000, Bordeaux, France
| | - Lotfi Hacein-Bey
- Radiology Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - David Devos
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,Department of Pharmacology, CHU Lille, F-59000, Lille, France
| | - Florence Pasquier
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France.,Department of Neurology, CHU Lille, F-59000, Lille, France
| | - Xavier Leclerc
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France.,Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France
| | - Jean-Pierre Pruvo
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France.,Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France
| | - Sébastien Verclytte
- Department of Imaging, Lille Catholic Hospitals, Lille Catholic University, F-59000, Lille, France
| |
Collapse
|
10
|
Antonazzo B, Marano G, Romagnoli E, Ronzoni S, Frati G, Sani G, Janiri L, Mazza M. Impact of arterial hypertension and its management strategies on cognitive function and dementia: a comprehensive umbrella review. Minerva Cardiol Angiol 2022; 70:285-297. [PMID: 33258570 DOI: 10.23736/s2724-5683.20.05452-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Cognitive decline and dementia recognize multiple risk factors and pathophysiological mechanisms, often involved simultaneously with complex interactions. Several studies have shown that both arterial hypertension and hypotension are associated with a greater risk of cognitive decline and dementia, but clinical evidence on this point is conflicting. Our aim was to conduct an umbrella review on cognitive function, dementia, and blood pressure, with particular attention to epidemiological, prognostic and therapeutic aspects. EVIDENCE ACQUISITION We conducted a dedicated literature search on PubMed for systematic reviews and meta-analyses that focused on arterial pressure, hypertension, hypotension and similar conditions, and cognitive function, cognitive decline and dementia. The internal validity of systematic reviews and meta-analyses was formally analyzed using the OQAQ tool. The umbrella review was planned in accordance with current international recommendations and was described as specified by the PRISMA guidelines. EVIDENCE SYNTHESIS Seventeen systematic reviews (including 13 meta-analyses) were included, for a total of 675 clinical studies and over 1 million patients. Hypertension results to be associated with a lower risk of Alzheimer's dementia, greater risk of vascular dementia and greater risk of cognitive decline. Orthostatic hypotension seems to be associated with greater risk of Alzheimer's dementia, vascular dementia and dementia of Parkinson's disease. Therapy with acetylcholinesterase inhibitors produces lower risk of cardiovascular events, greater risk of hypertension and greater risk of bradycardia, while the anti-hypertensive therapy leads to a lower risk of dementia of all types and lower risk of cognitive decline. CONCLUSIONS To date, the evidence on the relationship between blood pressure, cognitive decline and dementia provides somewhat heterogeneous data. Further studies are clearly needed, with explicit inclusion criteria as objective as possible, adequate follow-up and precise characterization of implemented cardiovascular and cognitive treatments.
Collapse
Affiliation(s)
| | - Giuseppe Marano
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Enrico Romagnoli
- Department of Cardiovascular and Thoracic Sciences, Institute of Cardiology, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Rome, Italy
| | | | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University, Rome, Italy
- IRCCS NEUROMED, Pozzilli, Isernia, Italy
| | - Gabriele Sani
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Luigi Janiri
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Marianna Mazza
- Department of Geriatrics, Institute of Psychiatry and Psychology, Neuroscience and Orthopedics, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy -
| |
Collapse
|
11
|
Delpech JC, Pathak D, Varghese M, Kalavai SV, Hays EC, Hof PR, Johnson WE, Ikezu S, Medalla M, Luebke JI, Ikezu T. Wolframin-1-expressing neurons in the entorhinal cortex propagate tau to CA1 neurons and impair hippocampal memory in mice. Sci Transl Med 2021; 13:eabe8455. [PMID: 34524859 PMCID: PMC8763211 DOI: 10.1126/scitranslmed.abe8455] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Abnormally phosphorylated tau, an early neuropathologic marker of Alzheimer’s disease (AD), first occurs in the brain’s entorhinal cortex layer II (ECII) and then spreads to the CA1 field of the hippocampus. Animal models of tau propagation aiming to recapitulate this phenomenon mostly show tau transfer from ECII stellate neurons to the dentate gyrus, but tau pathology in the dentate gyrus does not appear until advanced stages of AD. Wolframin-1–expressing (Wfs1+) pyramidal neurons have been shown functionally to modulate hippocampal CA1 neurons in mice. Here, we report that Wfs1+ pyramidal neurons are conserved in the ECII of postmortem human brain tissue and that Wfs1 colocalized with abnormally phosphorylated tau in brains from individuals with early AD. Wfs1+ neuron–specific expression of human P301L mutant tau in mouse ECII resulted in transfer of tau to hippocampal CA1 pyramidal neurons, suggesting spread of tau pathology as observed in the early Braak stages of AD. In mice expressing human mutant tau specifically in the ECII brain region, electrophysiological recordings of CA1 pyramidal neurons showed reduced excitability. Multielectrode array recordings of optogenetically stimulated Wfs1+ ECII axons resulted in reduced CA1 neuronal firing. Chemogenetic activation of CA1 pyramidal neurons showed a reduction in c-fos+ cells in the CA1. Last, a fear conditioning task revealed deficits in trace and contextual memory in mice overexpressing human mutant tau in the ECII. This work demonstrates tau transfer from the ECII to CA1 in mouse brain and provides an early Braak stage preclinical model of AD. Wolframin-1–positive neurons in the entorhinal cortex of mouse brain propagate tau to the hippocampal CA1 region resulting in memory impairment.
Collapse
Affiliation(s)
- Jean-Christophe Delpech
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Dhruba Pathak
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald C. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Srinidhi Venkatesan Kalavai
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Emma C Hays
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald C. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W Evan Johnson
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Seiko Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Maria Medalla
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Jennifer I Luebke
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Center for Systems Neuroscience, Boston University, Boston, MA, USA.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
12
|
Perez-Gonzalez J, Jiménez-Ángeles L, Rojas Saavedra K, Barbará Morales E, Medina-Bañuelos V. Mild cognitive impairment classification using combined structural and diffusion imaging biomarkers. Phys Med Biol 2021; 66. [PMID: 34167090 DOI: 10.1088/1361-6560/ac0e77] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/24/2021] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease is a multifactorial neurodegenerative disorder preceded by a prodromal stage called mild cognitive impairment (MCI). Early diagnosis of MCI is crucial for delaying the progression and optimizing the treatment. In this study we propose a random forest (RF) classifier to distinguish between MCI and healthy control subjects (HC), identifying the most relevant features computed from structural T1-weighted and diffusion-weighted magnetic resonance images (sMRI and DWI), combined with neuro-psychological scores. To train the RF we used a set of 60 subjects (HC = 30, MCI = 30) drawn from the Alzheimer's disease neuroimaging initiative database, while testing with unseen data was carried out on a 23-subjects Mexican cohort (HC = 12, MCI = 11). Features from hippocampus, thalamus and amygdala, for left and right hemispheres were fed to the RF, with the most relevant being previously selected by applying extra trees classifier and the mean decrease in impurity index. All the analyzed brain structures presented changes in sMRI and DWI features for MCI, but those computed from sMRI contribute the most to distinguish from HC. However, sMRI+DWI improves classification performance in training area under the receiver operating characteristic curve (AUROC = 93.5 ± 8%, accuracy = 88.8 ± 9%) and testing with unseen data (AUROC = 93.79%, accuracy = 91.3%), having a better performance when neuro-psychological scores were included. Compared to other classifiers the proposed RF provide the best performance for HC/MCI discrimination and the application of a feature selection step improves its performance. These findings imply that multimodal analysis gives better results than unimodal analysis and hence may be a useful tool to assist in early MCI diagnosis.
Collapse
Affiliation(s)
- Jorge Perez-Gonzalez
- Unidad Académica del Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas en el Estado de Yucatán, UNAM, Yucatán, México
| | - Luis Jiménez-Ángeles
- Department of Biomedical Systems Engineering, Engineering Faculty, UNAM, Mexico City, México
| | - Karla Rojas Saavedra
- Health Sciences Department, Universidad del Valle de México, Mexico City, México
| | | | | |
Collapse
|
13
|
Bernstein AS, Rapcsak SZ, Hornberger M, Saranathan M. Structural Changes in Thalamic Nuclei Across Prodromal and Clinical Alzheimer's Disease. J Alzheimers Dis 2021; 82:361-371. [PMID: 34024824 DOI: 10.3233/jad-201583] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Increasing evidence suggests that thalamic nuclei may atrophy in Alzheimer's disease (AD). We hypothesized that there will be significant atrophy of limbic thalamic nuclei associated with declining memory and cognition across the AD continuum. OBJECTIVE The objective of this work was to characterize volume differences in thalamic nuclei in subjects with early and late mild cognitive impairment (MCI) as well as AD when compared to healthy control (HC) subjects using a novel MRI-based thalamic segmentation technique (THOMAS). METHODS MPRAGE data from the ADNI database were used in this study (n = 540). Healthy control (n = 125), early MCI (n = 212), late MCI (n = 114), and AD subjects (n = 89) were selected, and their MRI data were parcellated to determine the volumes of 11 thalamic nuclei for each subject. Volumes across the different clinical subgroups were compared using ANCOVA. RESULTS There were significant differences in thalamic nuclei volumes between HC, late MCI, and AD subjects. The anteroventral, mediodorsal, pulvinar, medial geniculate, and centromedian nuclei were significantly smaller in subjects with late MCI and AD when compared to HC subjects. Furthermore, the mediodorsal, pulvinar, and medial geniculate nuclei were significantly smaller in early MCI when compared to HC subjects. CONCLUSION This work highlights nucleus specific atrophy within the thalamus in subjects with early and late MCI and AD. This is consistent with the hypothesis that memory and cognitive changes in AD are mediated by damage to a large-scale integrated neural network that extends beyond the medial temporal lobes.
Collapse
Affiliation(s)
- Adam S Bernstein
- Department of Medical Imaging, University of Arizona, Tuscon, AZ, USA
| | | | | | | | | |
Collapse
|
14
|
Qing Z, Chen F, Lu J, Lv P, Li W, Liang X, Wang M, Wang Z, Zhang X, Zhang B. Causal structural covariance network revealing atrophy progression in Alzheimer's disease continuum. Hum Brain Mapp 2021; 42:3950-3962. [PMID: 33978292 PMCID: PMC8288084 DOI: 10.1002/hbm.25531] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 04/10/2021] [Accepted: 04/26/2021] [Indexed: 01/24/2023] Open
Abstract
The structural covariance network (SCN) has provided a perspective on the large‐scale brain organization impairment in the Alzheimer's Disease (AD) continuum. However, the successive structural impairment across brain regions, which may underlie the disrupted SCN in the AD continuum, is not well understood. In the current study, we enrolled 446 subjects with AD, mild cognitive impairment (MCI) or normal aging (NA) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. The SCN as well as a casual SCN (CaSCN) based on Granger causality analysis were applied to the T1‐weighted structural magnetic resonance images of the subjects. Compared with that of the NAs, the SCN was disrupted in the MCI and AD subjects, with the hippocampus and left middle temporal lobe being the most impaired nodes, which is in line with previous studies. In contrast, according to the 194 subjects with records on CSF amyloid and Tau, the CaSCN revealed that during AD progression, the CaSCN was enhanced. Specifically, the hippocampus, thalamus, and precuneus/posterior cingulate cortex (PCC) were identified as the core regions in which atrophy originated and could predict atrophy in other brain regions. Taken together, these findings provide a comprehensive view of brain atrophy in the AD continuum and the relationships among the brain atrophy in different regions, which may provide novel insight into the progression of AD.
Collapse
Affiliation(s)
- Zhao Qing
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Institute of Brain Science, Nanjing University, Nanjing, China
| | - Feng Chen
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiaming Lu
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Pin Lv
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Weiping Li
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xue Liang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Maoxue Wang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhengge Wang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xin Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Bing Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Institute of Brain Science, Nanjing University, Nanjing, China
| | | |
Collapse
|
15
|
Abey A, Davies D, Goldsbury C, Buckland M, Valenzuela M, Duncan T. Distribution of tau hyperphosphorylation in canine dementia resembles early Alzheimer's disease and other tauopathies. Brain Pathol 2020; 31:144-162. [PMID: 32810333 PMCID: PMC8018065 DOI: 10.1111/bpa.12893] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Some aged community dogs acquire a degenerative syndrome termed Canine Cognitive Dysfunction (CCD) that resembles human dementia because of Alzheimer’s Disease (AD), with comparable cognitive and behavioral deficits. Dogs also have similar neuroanatomy, share our domestic environment and develop amyloid‐β plaques, making them likely a valuable ecological model of AD. However, prior investigations have demonstrated a lack of neurofibrillary tau pathology in aged dogs, an important hallmark of AD, though elevated phosphorylated tau (p‐tau) at the Serine 396 (S396) epitope has been reported in CCD. Here using enhanced immunohistochemical methods, we investigated p‐tau in six CCD brains and six controls using the AT8 antibody (later stage neurofibrillary pathology), and an antibody against S396 p‐tau (earlier stage tau dysfunction). For the first time, we systematically assessed the Papez circuit and regions associated with Braak staging and found that all CCD dogs displayed elevated S396 p‐tau labeling throughout the circuit. The limbic thalamus was particularly implicated, with a similar labeling pattern to that reported for AD neurofibrillary pathology, especially the anterior nuclei, while the hippocampus exhibited dysfunction confined to synaptic layers and efferent pathways. The cingulate and temporal lobes displayed significantly greater tauopathy than the frontal and occipital cortices, also reflective of early Braak staging patterns in AD. Immunofluorescence confirmed that S396 was accumulating within neuronal axons, somata and oligodendrocytes. We also observed AT8 labeling in one CCD brain, near the transentorhinal cortex in layer II neurons, one of the first regions to be affected in AD. Together, these data demonstrate a concordance in regional distribution of tauopathy between CCD and AD, most evident in the limbic thalamus, an important step in further validating CCD as a translational model for human AD and understanding early AD pathogenic mechanisms.
Collapse
Affiliation(s)
- Ajantha Abey
- Regenerative Neuroscience Group, The Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia.,Discipline of Anatomy and Histology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Danielle Davies
- Discipline of Anatomy and Histology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Alzheimer's Disease Cell Biology Research Lab, The Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Claire Goldsbury
- Discipline of Anatomy and Histology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Alzheimer's Disease Cell Biology Research Lab, The Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Michael Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Sydney Medical School, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Michael Valenzuela
- Regenerative Neuroscience Group, The Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia.,Discipline of Anatomy and Histology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,School of Psychiatry, Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Thomas Duncan
- Regenerative Neuroscience Group, The Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia.,Discipline of Anatomy and Histology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.,Department of Anatomy, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
16
|
miR-16-5p is upregulated by amyloid β deposition in Alzheimer's disease models and induces neuronal cell apoptosis through direct targeting and suppression of BCL-2. Exp Gerontol 2020; 136:110954. [PMID: 32320719 DOI: 10.1016/j.exger.2020.110954] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with irreversible neurodegeneration. Accumulation of amyloid beta (Aβ) in the brain is considered to be a major cause of neuronal cell death in AD, but the neurotoxic mechanism of Aβ is not yet fully understood. Here, we focused on the role of microRNAs (miRNAs) in Aβ-induced neuronal cell death. In microarray and RT-qPCR analysis of plasma miRNAs obtained from 5 familiar AD mutations (5xFAD) and wild-type (WT) mice of various ages, miR-16-5p showed a significant age-related change that was accompanied by neuronal cell death in the brain tissue of 5xFAD mice. In addition, increased miR-16-5p was prominent near Aβ plaque-deposition sites in 5xFAD mouse brains. Aβ treatment induced miR-16-5p upregulation and apoptosis in primary cultured mouse cortical neurons and the SH-SY5Y human neuroblastoma cell line. In silico analysis and reporter gene assays indicated that miR-16-5p directly targets the mRNA encoding the anti-apoptotic factor, B cell lymphoma-2 (BCL-2), in the neuronal cell line. Overexpression of miR-16-5p in SH-SY5Y cells downregulated BCL-2 expression and induced apoptosis. These results collectively suggest that the miR-16-5p/BCL-2 axis plays an important role for neuronal cell apoptosis in AD.
Collapse
|
17
|
Zhu K, Wang X, Sun B, Wu J, Lu H, Zhang X, Liang H, Zhang D, Liu C. Primary Age-Related Tauopathy in Human Subcortical Nuclei. Front Neurosci 2019; 13:529. [PMID: 31191227 PMCID: PMC6549797 DOI: 10.3389/fnins.2019.00529] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/07/2019] [Indexed: 12/29/2022] Open
Abstract
The present study aimed to determine the spatial distribution patterns of hyperphosphorylated tau-immunoreactive cells in subcortical nuclei of post-mortem human brain with primary age-related tauopathy (PART). Subcortical tauopathy has important pathological and clinical implications. Expression of tau was examined in different subcortical regions of definite PART cases with a Braak neurofibrillary tangle stage >0 and ≤IV, and with a Thal phase 0 (no beta-amyloid present). Post-mortem brain tissue of PART was studied using immunohistochemistry and subsequent semi-quantitative assessment with Braak NFT stage -matched pre-Alzheimer’s disease (AD) and AD cases as a control. Expression of tau was frequently found in subcortical nuclei including the substantia nigra, inferior colliculus, locus coeruleus, medulla oblongata in the brainstem, the caudate, putamen, nucleus globus pallidus in the striatum, the hypothalamus, thalamus, subthalamus in the diencephalon, and the cervical spinal cord in both PART and AD, but not in the dentate nucleus of the cerebellum. A positive correlation was found between the Braak NFT stage and the tau distribution (qualitative)/tau density (quantitative) in PART and AD. Brainstem nuclei were commonly involved in early PART with NFT Braak stage I/II, there was no preference among the substantia nigra, inferior colliculus, locus caeruleus and medulla oblongata. The prevalence and severity of tau pathology in subcortical nuclei of PART and AD were positively correlated with NFT Braak stage, suggesting that these nuclei were increasingly involved as PART and AD progressed. Subcortical nuclei were likely the sites initially affected by aging associated tau pathology, especially the brainstem nuclei including the substantia nigra, inferior colliculus, locus caeruleus and medulla oblongata.
Collapse
Affiliation(s)
- Keqing Zhu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Wang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Sun
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Juanli Wu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Lu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoling Zhang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Huazheng Liang
- Brain Structure and Function, Neuroscience Research Australia, Randwick, NSW, Australia.,Department of Neurology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chong Liu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Fredericks CA, Brown JA, Deng J, Kramer A, Ossenkoppele R, Rankin K, Kramer JH, Miller BL, Rabinovici GD, Seeley WW. Intrinsic connectivity networks in posterior cortical atrophy: A role for the pulvinar? Neuroimage Clin 2018; 21:101628. [PMID: 30528957 PMCID: PMC6411779 DOI: 10.1016/j.nicl.2018.101628] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/26/2018] [Accepted: 12/01/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Posterior cortical atrophy (PCA) is a clinical variant of Alzheimer's disease (AD) that presents with progressive visuospatial symptoms. While amnestic AD is characterized by disrupted default mode network (DMN) connectivity with corresponding increases in salience network (SN) connectivity, a visuospatial network appears to be disrupted early in PCA. Based on PCA patients' clinical features, we hypothesized that, in addition to early decreased integrity within the visuospatial network, patients with PCA would show increases in SN connectivity despite relative preservation of DMN. As the lateral pulvinar nucleus of the thalamus has direct anatomical connections with striate and extrastriate cortex and DMN, and the medial pulvinar is anatomically interconnected with SN, we further hypothesized that lateral and medial pulvinar nuclei might be implicated in intrinsic connectivity changes in PCA. METHODS 26 patients with PCA and 64 matched controls were recruited through UCSF Memory and Aging Center research programs. Each completed a standardized neuropsychological battery, structural MRI, and task-free fMRI. Seed-based functional correlations were used to probe networks of interest, including those seeded by the medial and lateral pulvinar thalamic nuclei, across the whole brain, and functional data analyses were adjusted for brain atrophy. RESULTS Patients with PCA showed disproportionate deficits in the visuospatial domain; they also showed preserved social sensitivity and endorsed more depressive symptoms than HCs. PCA patients had significant parietooccipital atrophy accompanied by widespread connectivity decreases within the visuospatial network, enhanced connectivity between some structures in SN, and enhanced connectivity between key nodes of the DMN compared to controls. Increased SN connectivity correlated with a measure of social sensitivity, and increased DMN connectivity correlated with short-term memory performance. Medial pulvinar connectivity increases in PCA were topographically similar to SN (anterior insula) connectivity increases, while lateral pulvinar connectivity increases were similar to DMN (posterior cingulate) connectivity increases. CONCLUSIONS PCA is characterized by preserved to heightened connectivity in the SN and DMN despite decreased visuospatial network connectivity. The spatial similarity of medial and lateral pulvinar connectivity changes to those seen in the SN and DMN suggests a role for the pulvinar in intrinsic connectivity network changes in PCA.
Collapse
Affiliation(s)
- Carolyn A Fredericks
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - Jesse A Brown
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - Jersey Deng
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA
| | - Abigail Kramer
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - Rik Ossenkoppele
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - Katherine Rankin
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - Joel H Kramer
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - Bruce L Miller
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| | - William W Seeley
- Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA 94143, USA.
| |
Collapse
|
19
|
Perry JC, Pakkenberg B, Vann SD. Striking reduction in neurons and glial cells in anterior thalamic nuclei of older patients with Down syndrome. Neurobiol Aging 2018; 75:54-61. [PMID: 30550978 PMCID: PMC6357872 DOI: 10.1016/j.neurobiolaging.2018.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 02/09/2023]
Abstract
The anterior thalamic nuclei are important for spatial and episodic memory, however, surprisingly little is known about the status of these nuclei in neurological conditions that present with memory impairments, such as Down syndrome. We quantified neurons and glial cells in the anterior thalamic nuclei of four older patients with Down syndrome. There was a striking reduction in the volume of the anterior thalamic nuclei and this appeared to reflect the loss of approximately 70% of neurons. The number of glial cells was also reduced but to a lesser degree than neurons. The anterior thalamic nuclei appear to be particularly sensitive to effects of aging in Down syndrome and the pathology in this region likely contributes to the memory impairments observed. These findings reaffirm the importance of examining the status of the anterior thalamic nuclei in conditions where memory impairments have been principally assigned to pathology in the medial temporal lobe. Volume of anterior thalamus is markedly reduced in older patients with Down syndrome. Number of neurons in anterior thalamus are substantially reduced. Number of glial cells in anterior thalamus are substantially reduced.
Collapse
Affiliation(s)
- James C Perry
- School of Psychology, Cardiff University, Cardiff, UK
| | - Bente Pakkenberg
- Research Laboratory for Stereology and Neuroscience, Copenhagen University Hospital, Denmark and Institute of Clinical Medicine, Faculty of Health, University of Copenhagen, Bispebjerg, Copenhagen, Denmark
| | | |
Collapse
|
20
|
Jazvinšćak Jembrek M, Slade N, Hof PR, Šimić G. The interactions of p53 with tau and Aß as potential therapeutic targets for Alzheimer’s disease. Prog Neurobiol 2018; 168:104-127. [DOI: 10.1016/j.pneurobio.2018.05.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/04/2018] [Accepted: 05/01/2018] [Indexed: 12/24/2022]
|
21
|
A Closer Look into the Role of Protein Tau in the Identification of Promising Therapeutic Targets for Alzheimer's Disease. Brain Sci 2018; 8:brainsci8090162. [PMID: 30149687 PMCID: PMC6162660 DOI: 10.3390/brainsci8090162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 01/09/2023] Open
Abstract
One of the most commonly known chronic neurodegenerative disorders, Alzheimer's disease (AD), manifests the common type of dementia in 60⁻80% of cases. From a clinical standpoint, a patent cognitive decline and a severe change in personality, as caused by a loss of neurons, is usually evident in AD with about 50 million people affected in 2016. The disease progression in patients is distinguished by a gradual plummet in cognitive functions, eliciting symptoms such as memory loss, and eventually requiring full-time medical care. From a histopathological standpoint, the defining characteristics are intracellular aggregations of hyper-phosphorylated tau protein, known as neurofibrillary tangles (NFT), and depositions of amyloid β-peptides (Aβ) in the brain. The abnormal phosphorylation of tau protein is attributed to a wide gamut of neurological disorders known as tauopathies. In addition to the hyperphosphorylated tau lesions, neuroinflammatory processes could occur in a sustained manner through astro-glial activation, resulting in the disease progression. Recent findings have suggested a strong interplay between the mechanism of Tau phosphorylation, disruption of microtubules, and synaptic loss and pathology of AD. The mechanisms underlying these interactions along with their respective consequences in Tau pathology are still ill-defined. Thus, in this review: (1) we highlight the interplays existing between Tau pathology and AD; and (2) take a closer look into its role while identifying some promising therapeutic advances including state of the art imaging techniques.
Collapse
|
22
|
Yuan H, Du L, Ge P, Wang X, Xia Q. Association of microtubule-associated protein tau gene polymorphisms with the risk of sporadic Alzheimer's disease: a meta-analysis. Int J Neurosci 2018; 128:577-585. [PMID: 29098924 DOI: 10.1080/00207454.2017.1400972] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Hai Yuan
- Department of Rehabilitation Medicine, The Second People's Hospital of Hefei City, Hefei, China
| | - Lingling Du
- Department of Rehabilitation Medicine, The Second People's Hospital of Hefei City, Hefei, China
- Department of Rehabilitation Medicine, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, China
| | - Pingping Ge
- Department of Rehabilitation Medicine, The Second People's Hospital of Hefei City, Hefei, China
| | - Xiaotong Wang
- Department of Neurology, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qing Xia
- Department of Rehabilitation Medicine, The Second People's Hospital of Hefei City, Hefei, China
| |
Collapse
|
23
|
Rüb U, Stratmann K, Heinsen H, Seidel K, Bouzrou M, Korf HW. Alzheimer's Disease: Characterization of the Brain Sites of the Initial Tau Cytoskeletal Pathology Will Improve the Success of Novel Immunological Anti-Tau Treatment Approaches. J Alzheimers Dis 2017; 57:683-696. [PMID: 28269779 DOI: 10.3233/jad-161102] [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] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) represents the most frequent neurodegenerative disease of the human brain worldwide. Currently practiced treatment strategies for AD only include some less effective symptomatic therapeutic interventions, which unable to counteract the disease course of AD. New therapeutic attempts aimed to prevent, reduce, or remove the extracellular depositions of the amyloid-β protein did not elicit beneficial effects on cognitive deficits or functional decline of AD. In view of the failure of these amyloid-β-based therapeutic trials and the close correlation between the brain pathology of the cytoskeletal tau protein and clinical AD symptoms, therapeutic attention has since shifted to the tau cytoskeletal protein as a novel drug target. The abnormal hyperphosphorylation and intraneuronal aggregation of this protein are early events in the evolution of the AD-related neurofibrillary pathology, and the brain spread of the AD-related tau aggregation pathology may possibly follow a corruptive protein templating and seeding-like mechanism according to the prion hypothesis. Accordingly, immunotherapeutic targeting of the tau aggregation pathology during the very early pre-tangle phase is currently considered to represent an effective and promising therapeutic approach for AD. Recent studies have shown that the initial immunoreactive tau aggregation pathology already prevails in several subcortical regions in the absence of any cytoskeletal changes in the cerebral cortex. Thus, it may be hypothesized that the subcortical brain regions represent the "port of entry" for the pathogenetic agent from which the disease ascends anterogradely as an "interconnectivity pathology".
Collapse
Affiliation(s)
- Udo Rüb
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Katharina Stratmann
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Helmut Heinsen
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany.,Department of Pathology, Ageing Brain Study Group, University of São Paulo Medical School, São Paulo, Brazil
| | - Kay Seidel
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Mohamed Bouzrou
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| | - Horst-Werner Korf
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-University, Frankfurt/Main, Germany
| |
Collapse
|
24
|
Aggleton JP, Pralus A, Nelson AJD, Hornberger M. Thalamic pathology and memory loss in early Alzheimer's disease: moving the focus from the medial temporal lobe to Papez circuit. Brain 2016; 139:1877-90. [PMID: 27190025 PMCID: PMC4939698 DOI: 10.1093/brain/aww083] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/26/2016] [Indexed: 11/13/2022] Open
Abstract
It is widely assumed that incipient protein pathology in the medial temporal lobe instigates the loss of episodic memory in Alzheimer’s disease, one of the earliest cognitive deficits in this type of dementia. Within this region, the hippocampus is seen as the most vital for episodic memory. Consequently, research into the causes of memory loss in Alzheimer’s disease continues to centre on hippocampal dysfunction and how disease-modifying therapies in this region can potentially alleviate memory symptomology. The present review questions this entrenched notion by bringing together findings from post-mortem studies, non-invasive imaging (including studies of presymptomatic, at-risk cases) and genetically modified animal models. The combined evidence indicates that the loss of episodic memory in early Alzheimer’s disease reflects much wider neurodegeneration in an extended mnemonic system (Papez circuit), which critically involves the limbic thalamus. Within this system, the anterior thalamic nuclei are prominent, both for their vital contributions to episodic memory and for how these same nuclei appear vulnerable in prodromal Alzheimer’s disease. As thalamic abnormalities occur in some of the earliest stages of the disease, the idea that such changes are merely secondary to medial temporal lobe dysfunctions is challenged. This alternate view is further strengthened by the interdependent relationship between the anterior thalamic nuclei and retrosplenial cortex, given how dysfunctions in the latter cortical area provide some of the earliest
in vivo
imaging evidence of prodromal Alzheimer’s disease. Appreciating the importance of the anterior thalamic nuclei for memory and attention provides a more balanced understanding of Alzheimer’s disease. Furthermore, this refocus on the limbic thalamus, as well as the rest of Papez circuit, would have significant implications for the diagnostics, modelling, and experimental treatment of cognitive symptoms in Alzheimer’s disease.
Collapse
Affiliation(s)
- John P Aggleton
- School of Psychology, Cardiff University, Park Place, Cardiff, CF10 3AT, UK
| | - Agathe Pralus
- Master of Biosciences, ENS de Lyon, 46 allée d'Italie, 69007 Lyon, France
| | - Andrew J D Nelson
- School of Psychology, Cardiff University, Park Place, Cardiff, CF10 3AT, UK
| | | |
Collapse
|