1
|
Liou JJ, Li J, Berardinelli J, Jin H, Santini T, Noh J, Farhat N, Wu M, Aizenstein HJ, Mettenburg JM, Yong WH, Head E, Ikonomovic MD, Ibrahim TS, Kofler JK. Correlating hippocampal and amygdala volumes with neuropathological burden in Down syndrome and Alzheimer's disease and related neurodegenerative pathologies using 7T postmortem MRI. J Neuropathol Exp Neurol 2025; 84:364-378. [PMID: 40063697 PMCID: PMC12012357 DOI: 10.1093/jnen/nlaf010] [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] [Indexed: 03/19/2025] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC), is common in elderly brains and often seen in conjunction with Alzheimer's disease neuropathologic change (ADNC). LATE-NC typically begins in the amygdala and spreads to the hippocampus and neocortex. Whether it contributes to hippocampal and amygdala atrophy in Down syndrome (DS) remains unexplored. We analyzed amygdala and hippocampal volumes and neuropathological burden in 12 DS cases and 54 non-DS cases with AD and related neurodegenerative pathologies (ADRNP) using 7 Tesla (7T) postmortem ex vivo MRI. Postmortem and antemortem hippocampal volumes were significantly correlated in a subset of 17 cases with available antemortem MRI scans. DS cases had smaller hippocampal and amygdala volumes than ADRNP cases; these correlated with more severe Braak stage but not with Thal phase. LATE-NC and hippocampal sclerosis (HS) were uncommon in DS cases. In ADRNP cases, lower hippocampal volumes associated with dementia duration, advanced Thal phase, Braak NFT stage, C score, LATE-NC stage, HS and arteriolosclerosis severity; reduced amygdala volumes correlated with severe LATE-NC stage, HS, and arteriolosclerosis severity, but not with Thal phase or Braak NFT stage. Lewy body pathology did not affect hippocampal or amygdala volume in either cohort. Thus, hippocampal volumes in ADRNP were influenced by both ADNC and LATE-NC, and amygdala volumes were primarily influenced by LATE-NC. In DS, hippocampal and amygdala volumes were primarily influenced by tau pathology.
Collapse
Affiliation(s)
- Jr-Jiun Liou
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jinghang Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jacob Berardinelli
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hecheng Jin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tales Santini
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jaehoon Noh
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nadim Farhat
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Minjie Wu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Howard J Aizenstein
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph M Mettenburg
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - William H Yong
- Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, United States
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, United States
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tamer S Ibrahim
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Julia K Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Peng W, Ma Y, Li C, Dai W, Fu X, Liu L, Liu L, Liu J. Fusion of brain imaging genetic data for alzheimer's disease diagnosis and causal factors identification using multi-stream attention mechanisms and graph convolutional networks. Neural Netw 2025; 184:107020. [PMID: 39721106 DOI: 10.1016/j.neunet.2024.107020] [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: 06/01/2024] [Revised: 11/03/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
Correctly diagnosing Alzheimer's disease (AD) and identifying pathogenic brain regions and genes play a vital role in understanding the AD and developing effective prevention and treatment strategies. Recent works combine imaging and genetic data, and leverage the strengths of both modalities to achieve better classification results. In this work, we propose MCA-GCN, a Multi-stream Cross-Attention and Graph Convolutional Network-based classification method for AD patients. It first constructs a brain region-gene association network based on brain region fMRI time series and gene SNP data. Then it integrates the absolute and relative positions of the brain region time series to obtain a new brain region time series containing temporal information. Then long-range and local association features between brain regions and genes are sequentially aggregated by multi-stream cross-attention and graph convolutional networks. Finally, the learned brain region and gene features are input to the fully connected network to predict AD types. Experimental results on the ADNI dataset show that our model outperforms other methods in AD classification tasks. Moreover, MCA-GCN designed a multi-stage feature scoring process to extract high-risk genes and brain regions related to disease classification.
Collapse
Affiliation(s)
- Wei Peng
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology; Kunming 650500, PR China; Computer Technology Application Key Lab of Yunnan Province; Kunming 650500, PR China.
| | - Yanhan Ma
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology; Kunming 650500, PR China; Computer Technology Application Key Lab of Yunnan Province; Kunming 650500, PR China
| | - Chunshan Li
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology; Kunming 650500, PR China; Computer Technology Application Key Lab of Yunnan Province; Kunming 650500, PR China
| | - Wei Dai
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology; Kunming 650500, PR China; Computer Technology Application Key Lab of Yunnan Province; Kunming 650500, PR China
| | - Xiaodong Fu
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology; Kunming 650500, PR China; Computer Technology Application Key Lab of Yunnan Province; Kunming 650500, PR China
| | - Li Liu
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology; Kunming 650500, PR China; Computer Technology Application Key Lab of Yunnan Province; Kunming 650500, PR China
| | - Lijun Liu
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology; Kunming 650500, PR China; Computer Technology Application Key Lab of Yunnan Province; Kunming 650500, PR China
| | - Jin Liu
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, PR China
| |
Collapse
|
3
|
Richter N, Breidenbach L, Schmieschek MH, Heiss WD, Fink GR, Onur OA. Alzheimer-typical temporo-parietal atrophy and hypoperfusion are associated with a more significant cholinergic impairment in amnestic neurodegenerative syndromes. J Alzheimers Dis 2025; 104:1290-1300. [PMID: 40116674 DOI: 10.1177/13872877251324080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
BackgroundTo date, cholinomimetics remain central in the pharmacotherapy of Alzheimer's disease (AD) dementia. However, postmortem investigations indicate that the AD-typical progressive amnestic syndrome may also result from predominantly limbic non-AD neuropathology such as TDP-43 proteinopathy and argyrophilic grain disease. Experimental evidence links a beneficial response to cholinomimetics in early AD to reduced markers of cholinergic neurotransmission. However, the cholinergic impairment varies among patients with a clinical AD presentation, likely due to non-AD (co)-pathologies.ObjectiveThis study examines whether AD-typical atrophy and hypoperfusion can provide information about the cholinergic system in clinically diagnosed AD.MethodsThirty-two patients with amnestic mild cognitive impairment or mild dementia due to AD underwent positron emission tomography (PET) with the tracer N-methyl-4-piperidyl-acetate (MP4A) to estimate acetylcholinesterase (AChE) activity, neurological examinations, cerebral magnetic resonance imaging (MRI) and neuropsychological assessment. The 'cholinergic deficit' was computed as the deviation of AChE activity from cognitively normal controls across the cerebral cortex and correlated gray matter (GM) and perfusion of temporo-parietal cortices typically affected by AD and basal forebrain (BF) GM.ResultsTemporo-parietal perfusion and GM, as well as the inferior temporal to medial temporal ratio of perfusion correlated negatively with the 'cholinergic deficit'. A smaller Ch4p area of the BF was associated with a more significant 'cholinergic deficit', albeit to a lesser degree than cortical measures.ConclusionsIn clinically diagnosed AD, temporo-parietal GM and perfusion are more closely associated with the 'cholinergic deficit' than BF volumes, making them possible markers for cholinergic treatment response in amnestic neurodegeneration.
Collapse
Affiliation(s)
- Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Germany
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, Germany
| | - Laura Breidenbach
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, Germany
| | - Maximilian Ht Schmieschek
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, Germany
| | | | - Gereon R Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Germany
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, Germany
| | - Oezguer A Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Germany
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, Germany
| |
Collapse
|
4
|
Wisse LEM, Wuestefeld A, Murray ME, Jagust W, La Joie R. Role of tau versus TDP-43 pathology on medial temporal lobe atrophy in aging and Alzheimer's disease. Alzheimers Dement 2025; 21:e14582. [PMID: 39985478 PMCID: PMC11846482 DOI: 10.1002/alz.14582] [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: 10/01/2024] [Revised: 12/16/2024] [Accepted: 01/12/2025] [Indexed: 02/24/2025]
Abstract
Hippocampal atrophy on magnetic resonance imaging is an important biomarker in Alzheimer's disease (AD). While hippocampal atrophy was thought to result from tau tangles in AD, different neuropathologies can lead to hippocampal atrophy, especially TAR DNA-binding protein 43 (TDP-43) pathology. In this narrative review, we evaluate existing studies on the relative contribution of tau and TDP-43 pathology to medial temporal lobe (MTL) atrophy. We report a clear association of both tau and TDP-43 neuropathology with MTL atrophy, even after correcting for other neuropathologies. Next, we discuss a potential synergism between tau and TDP-43 and the relative timing of the effects of both neuropathologies. Finally, avenues for future research will be discussed. A better understanding of the interplay between tau and TDP-43 neuropathologies and their effect on atrophy will help with the development of more specific biomarkers for limbic-predominant age-related TDP-43 encephalopathy and pinpointing of the optimal timing for testing anti-tau and anti-TDP-43 treatments in trials. HIGHLIGHTS: Both tau and TAR DNA-binding protein 43 (TDP-43) pathology contribute to medial temporal lobe atrophy. There is a positive association between tau and TDP-43 and potentially a synergism. It is unclear if tau and TDP-43 have an additive or synergistic effect on atrophy. The relative timing of the tau and TDP-43 effects on atrophy remains unclear. Clarifying the interplay between tau and TDP-43 will help improve magnetic resonance imaging biomarkers.
Collapse
Affiliation(s)
| | - Anika Wuestefeld
- Clinical Memory Research Unit, Department of Clinical Sciences MalmöLund UniversityLundSweden
| | - Melissa E. Murray
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
- Department of Laboratory Medicine and PathologyMayo Clinic FloridaJacksonvilleFloridaUSA
| | - William Jagust
- Department of NeuroscienceUniversity of California BerkeleyBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
5
|
Schumacher J, Teipel S, Storch A. Association of Alzheimer's and Lewy body disease pathology with basal forebrain volume and cognitive impairment. Alzheimers Res Ther 2025; 17:28. [PMID: 39865328 PMCID: PMC11771035 DOI: 10.1186/s13195-025-01678-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Degeneration of the basal forebrain cholinergic system is a hallmark feature shared by Alzheimer's disease (AD) and Lewy body disease (LBD) whereas hippocampus atrophy is more specifically related to AD. We aimed to investigate the relationship between basal forebrain and hippocampus atrophy, cognitive decline, and neuropathology in a large autopsy sample. METHODS Data were obtained from the National Alzheimer's Coordinating Center (NACC). Basal forebrain and hippocampus volumes were extracted using an established automated MRI volumetry approach. Associations of regional volumes with pathological markers (Braak stage, CERAD score, and McKeith criteria for LB pathology) and cognitive performance were assessed using Bayesian statistical methods. RESULTS We included people with autopsy-confirmed pure AD (N = 248), pure LBD (N = 22), and mixed AD/LBD (N = 185). Posterior basal forebrain atrophy was most severe in mixed AD/LB pathology compared to pure AD (Bayes factor against the null hypothesis BF10 = 16.2) or pure LBD (BF10 = 4.5). In contrast, hippocampal atrophy was primarily associated with AD pathology, independent of LB pathology (pure AD vs. pure LBD: BF10 = 166, pure AD vs. mixed AD/LBD: BF10 = 0.11, pure LBD vs. mixed AD/LBD: BF10 = 350). Cognitive performance was more impaired in AD pathology groups, with Braak stage being the strongest predictor. Hippocampal volume partially mediated this relationship between tau pathology and cognitive impairment, while basal forebrain volume had a limited role in mediating the relationship between pathological burden and cognitive outcomes. CONCLUSION In a heterogeneous autopsy sample, AD and LB pathology both contribute to cholinergic basal forebrain degeneration whereas hippocampus atrophy is more specifically related to AD pathology. Cognitive deficits are primarily associated with tau pathology which is partly mediated by hippocampus, but not basal forebrain atrophy.
Collapse
Affiliation(s)
- Julia Schumacher
- Department of Neurology, University Medical Center Rostock, 18147, Rostock, Germany.
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany.
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany
- Department of Psychosomatic Medicine, University Medical Center Rostock, 18147, Rostock, Germany
| | - Alexander Storch
- Department of Neurology, University Medical Center Rostock, 18147, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany
| |
Collapse
|
6
|
Ortega-Cruz D, Rabano A, Strange BA. Neuropathological contributions to grey matter atrophy and white matter hyperintensities in amnestic dementia. Alzheimers Res Ther 2025; 17:16. [PMID: 39789603 PMCID: PMC11714914 DOI: 10.1186/s13195-024-01633-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/29/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Dementia patients commonly present multiple neuropathologies, worsening cognitive function, yet structural neuroimaging signatures of dementia have not been positioned in the context of combined pathology. In this study, we implemented an MRI voxel-based approach to explore combined and independent effects of dementia pathologies on grey and white matter structural changes. METHODS In 91 amnestic dementia patients with post-mortem brain donation, grey matter density and white matter hyperintensity (WMH) burdens were obtained from pre-mortem MRI and analyzed in relation to Alzheimer's, vascular, Lewy body, TDP-43, and hippocampal sclerosis (HS) pathologies. After exploring co-occurrence profiles of these pathologies, voxel-based morphometry was implemented to determine their joint and independent effects on grey matter loss. The impact of these pathologies on WMH burden was then evaluated both in spatial and quantitative combined analyses, using voxel-based and generalized linear models respectively. RESULTS 86.8% of patients in this cohort presented more than one pathology. The combined structural effect of these pathologies was a focal impact on hippocampal grey matter atrophy, primarily driven by HS and Alzheimer's pathology (family-wise error corrected, p < 0.05), which also exhibited the strongest individual effects (uncorrected, p < 0.001). WMHs, predominant in middle and anterior cerebral portions, were most strongly associated with vascular (T = 2.47, p = 0.017) and tau pathologies (T = 2.09, p = 0.041). CONCLUSIONS The mixed associations of these dementia neuroimaging hallmarks are relevant for the fine-tuning of diagnostic protocols and underscore the need for comprehensive pathology evaluations in the study of dementia phenotypes.
Collapse
Affiliation(s)
- Diana Ortega-Cruz
- Laboratory for Clinical Neuroscience, Center for Biomedical Technology, Universidad Politécnica de Madrid, IdISSC, Crta M40, km38, Madrid, 28223, Spain
- Alzheimer's Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, 28031, Spain
| | - Alberto Rabano
- Alzheimer's Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, 28031, Spain
| | - Bryan A Strange
- Laboratory for Clinical Neuroscience, Center for Biomedical Technology, Universidad Politécnica de Madrid, IdISSC, Crta M40, km38, Madrid, 28223, Spain.
- Alzheimer's Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, 28031, Spain.
| |
Collapse
|
7
|
Kang S, Jeon S, Kim Y, Jeon S, Choi M, Lee Y, Yun M, Ye BS. White matter hyperintensities and cholinergic degeneration as Lewy body disease. Ann Clin Transl Neurol 2025; 12:97-109. [PMID: 39654300 PMCID: PMC11752093 DOI: 10.1002/acn3.52257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/25/2024] [Accepted: 11/06/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Although basal forebrain (BF) cholinergic degeneration and white matter hyperintensities (WMHs) are important in neurodegeneration in Alzheimer's disease (AD) and dementia with Lewy bodies (DLB), their relationships with dopaminergic degeneration and clinical manifestations remain unclear. METHODS A total of 407 patients with cognitive impairment meeting the diagnostic criteria for AD, DLB, or both (AD+DLB) were assessed. All participants underwent 3T MRI, dopamine transporter (DAT) positron emission tomography, neuropsychological tests, and assessments for parkinsonism, cognitive fluctuation, visual hallucination, and rapid eye movement sleep behavior disorder (RBD). General linear and logistic regression models were used to investigate the relationships among BF volume, DAT uptake in the anterior caudate (DAT-AC), WMH volumes in anterior, posterior, periventricular, and deep regions, and clinical manifestations. RESULTS DAT-AC was positively associated with BF volume and negatively associated with anterior periventricular WMH volume, but not with deep WMHs. Both deep and periventricular WMHs volumes were associated with hypertension and the number of microbleeds and lacunae. Lower BF volume and DAT-AC were independently associated with increased risk of cognitive fluctuation and visual hallucination, whereas lower DAT-AC was additionally associated with increased risk of RBD and greater parkinsonian severity. Both lower BF volume and DAT-AC were independently associated with widespread cognitive impairment, whereas higher anterior periventricular WMH volume was associated with executive dysfunction. INTERPRETATION BF cholinergic degeneration and anterior periventricular WMHs are closely associated with dopaminergic degeneration. Anterior periventricular WMHs may represent axonal alterations caused by the interplay between Lewy body-related degeneration and vascular pathologies.
Collapse
Affiliation(s)
- Sungwoo Kang
- Department of NeurologyYonsei University College of MedicineSeoul03722Republic of Korea
| | - Seun Jeon
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Yeoju Kim
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Su‐Hee Jeon
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Minsun Choi
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Young‐gun Lee
- Department of Neurology, Ilsan Paik HospitalInje University College of MedicineGoyang10380Republic of Korea
| | - Mijin Yun
- Department of Nuclear MedicineYonsei University College of MedicineSeoul03722Republic of Korea
| | - Byoung Seok Ye
- Department of NeurologyYonsei University College of MedicineSeoul03722Republic of Korea
| |
Collapse
|
8
|
Taheri M, Moradi MH, Koraee Y, Moghadam FH, Ershad Nedaei S, Veisi M, Ghafouri H. Neuroprotective properties of a thiazolidine-2,4-dione derivative as an inhibitory agent against memory impairment and phosphorylated tau: In vitro and in vivo investigations. Neuroscience 2024; 562:227-238. [PMID: 39489476 DOI: 10.1016/j.neuroscience.2024.10.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) is the most common form of neurodegeneration that results in memory disorders and cognitive impairment. The present study investigated the neuroprotective effects of the synthesized thiazolidine-2,4-dione derivative, (E)-5-(4-chlorobenzylidene)-3-(2-oxo-2-phenylethyl)thiazolidine-2,4-dione (TZ4C), an inhibitor of p-Tau and memory impairment, using a SH-SY5Y cell model of methamphetamine-induced tauopathy and a scopolamine-induced memory impairment model in Wistar rats. In the present study, the neuroprotective effect of TZ4C was studied in a SH-SY5Y cellular model of methamphetamine-induced (2 mM) tauopathy and a scopolamine-induced (1.5 mg/kg/day) memory impairment model in male Wistar rats (n = 48). The memory functions and learning abilities of the rats were evaluated using the Morris water maze (MWM) and passive avoidance tests. Additionally, AChE activity in the rat hippocampus was quantified, and the expression of p-Tau, HSP70, and caspase-3 in both in vitro and in vivo samples was evaluated through Western blot analysis. TZ4C (0.1-1000 µM) did not exhibit significantly toxic effects on SH-SY5Y cell viability. Western blot results indicated that TZ4C led to reduced expression of p-Tau, HSP70, and cleaved caspase-3 in SH-SY5Y cells (3 and 10 µM) and the rat hippocampus (2 and 4 mg/kg). Additionally, the findings suggested that TZ4C enhanced memory function in rats with scopolamine-induced impairment and decreased acetylcholinesterase (AChE) specific activity. The comprehensive analysis of in vitro and in vivo experiments underscores the neuroprotective potential (improved neuropathology and reduced memory impairment) of TZ4C. These findings highlight the promise of TZ4C as a candidate for drug discovery programs to identify effective therapies for AD.
Collapse
Affiliation(s)
- Maryam Taheri
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Mohammad Hadi Moradi
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Yasaman Koraee
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Farshad Homayouni Moghadam
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Seyed Ershad Nedaei
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojgan Veisi
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Ghafouri
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran.
| |
Collapse
|
9
|
Wisse LEM, Spotorno N, Rossi M, Grothe MJ, Mammana A, Tideman P, Baiardi S, Strandberg O, Ticca A, van Westen D, Mattsson-Carlgren N, Palmqvist S, Stomrud E, Parchi P, Hansson O. MRI Signature of α-Synuclein Pathology in Asymptomatic Stages and a Memory Clinic Population. JAMA Neurol 2024; 81:1051-1059. [PMID: 39068668 PMCID: PMC11284633 DOI: 10.1001/jamaneurol.2024.2713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/15/2024] [Indexed: 07/30/2024]
Abstract
Importance The lack of an in vivo measure for α-synuclein (α-syn) pathology until recently has limited thorough characterization of its brain atrophy pattern, especially during early disease stages. Objective To assess the association of state-of-the-art cerebrospinal fluid (CSF) seed amplification assays (SAA) α-syn positivity (SAA α-syn+) with magnetic resonance imaging (MRI) structural measures, across the continuum from clinically unimpaired (CU) to cognitively impaired (CI) individuals, in 3 independent cohorts, and separately in CU and CI individuals, the latter reflecting a memory clinic population. Design, Setting, and Participants Cross-sectional data were used from the Swedish BioFINDER-2 study (inclusion, 2017-2023) as the discovery cohort and the Swedish BioFINDER-1 study (inclusion, 2007-2015) and Alzheimer's Disease Neuroimaging Initiative (ADNI; inclusion 2005-2022) as replication cohorts. All cohorts are from multicenter studies, but the BioFINDER cohorts used 1 MRI scanner. CU and CI individuals fulfilling inclusion criteria and without missing data points in relevant metrics were included in the study. All analyses were performed from 2023 to 2024. Exposures Presence of α-syn pathology, estimated by baseline CSF SAA α-syn. Main Outcomes and Measures The primary outcomes were cross-sectional structural MRI measures either through voxel-based morphometry (VBM) or regions of interest (ROI) including an automated pipeline for cholinergic basal forebrain nuclei CH4/4p (nucleus basalis of Meynert [NBM]) and CH1/2/3. Secondary outcomes were domain-specific cross-sectional cognitive measures. Analyses were adjusted for CSF biomarkers of Alzheimer pathology. Results A total of 2961 participants were included in this study: 1388 (mean [SD] age, 71 [10] years; 702 female [51%]) from the BioFINDER-2 study, 752 (mean [SD] age, 72 [6] years; 406 female [54%]) from the BioFINDER-1 study, and 821 (mean [SD] age, 75 [8] years; 449 male [55%]) from ADNI. In the BioFINDER-2 study, VBM analyses in the whole cohort revealed a specific association between SAA α-syn+ and the cholinergic NBM, even when adjusting for Alzheimer copathology. ROI-based analyses in the BioFINDER-2 study focused on regions involved in the cholinergic system and confirmed that SAA α-syn+ was indeed independently associated with smaller NBM (β = -0.271; 95% CI, -0.399 to -0.142; P <.001) and CH1/2/3 volumes (β = -0.227; 95% CI, -0.377 to -0.076; P =.02). SAA α-syn+ was also independently associated with smaller NBM volumes in the separate CU (β = -0.360; 95% CI, -0.603 to -0.117; P =.03) and CI (β = -0.251; 95% CI, -0.408 to -0.095; P =.02) groups. Overall, the association between SAA α-syn+ and NBM volume was replicated in the BioFINDER-1 study and ADNI cohort. In CI individuals, NBM volumes partially mediated the association of SAA α-syn+ with attention/executive impairments in all cohorts (BioFINDER-2, β = -0.017; proportion-mediated effect, 7%; P =.04; BioFINDER-1, β = -0.096; proportion-mediated effect, 19%; P =.04; ADNI, β = -0.061; proportion-mediated effect, 20%; P =.007). Conclusions and Relevance In this cohort study, SAA α-syn+ was consistently associated with NBM atrophy already during asymptomatic stages. Further, in memory clinic CI populations, SAA α-syn+ was associated with NBM atrophy, which partially mediated α-syn-induced attention/executive impairment.
Collapse
Affiliation(s)
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Marcello Rossi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Michel J. Grothe
- Reina Sofia Alzheimer Center, CIEN Foundation, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigacion Biomédica en Red Sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Angela Mammana
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Simone Baiardi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Alice Ticca
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Danielle van Westen
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, Lund, Sweden
- Image and Function, Skåne University Hospital, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
10
|
Schumacher J, Ray N, Teipel S, Storch A. Associations of cholinergic system integrity with cognitive decline in GBA1 and LRRK2 mutation carriers. NPJ Parkinsons Dis 2024; 10:127. [PMID: 38951174 PMCID: PMC11217433 DOI: 10.1038/s41531-024-00743-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/14/2024] [Indexed: 07/03/2024] Open
Abstract
In Parkinson's disease (PD), GBA1- and LRRK2-mutations are associated with different clinical phenotypes which might be related to differential involvement of the cholinergic system. We investigated cholinergic integrity in 149 asymptomatic GBA1 and 169 asymptomatic LRRK2 mutation carriers, 112 LRRK2 and 60 GBA1 carriers with PD, 492 idiopathic PD, and 180 controls from the PPMI cohort. Basal forebrain volumes were extracted and white matter pathways from nucleus basalis of Meynert (NBM) to cortex and from pedunculopontine nucleus (PPN) to thalamus were assessed with a free water-corrected DTI model. Bayesian ANCOVAs were conducted for group comparisons and Bayesian linear mixed models to assess associations with cognitive decline. Basal forebrain volumes were increased in asymptomatic GBA1 (Bayes Factor against the null hypothesis (BF10) = 75.2) and asymptomatic LRRK2 (BF10 = 57.0) compared to controls. Basal forebrain volumes were increased in LRRK2- compared to GBA1-PD (BF10 = 14.5) and idiopathic PD (BF10 = 3.6*107), with no difference between idiopathic PD and PD-GBA1 (BF10 = 0.25). Mean diffusivity along the medial NBM pathway was decreased in asymptomatic GBA1 compared to controls (BF10 = 30.3). Over 5 years, idiopathic PD and PD-GBA1 declined across all cognitive domains whereas PD-LRRK2 patients only declined in processing speed. We found an interaction between basal forebrain volume and time in predicting multiple cognitive domains in idiopathic PD and PD-GBA1, but not in PD-LRRK2. While LRRK2 and GBA1 mutations are both associated with increased basal forebrain volume at asymptomatic stages, this increase persists at the symptomatic PD stage only in LRRK2 and might be related to slower cognitive decline in these patients.
Collapse
Affiliation(s)
- Julia Schumacher
- Department of Neurology, University Medical Center Rostock, 18147, Rostock, Germany.
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany.
| | - Nicola Ray
- Health, Psychology and Communities Research Centre, Department of Psychology, Manchester Metropolitan University, Manchester, UK
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany
- Department of Psychosomatic Medicine, University Medical Center Rostock, 18147, Rostock, Germany
| | - Alexander Storch
- Department of Neurology, University Medical Center Rostock, 18147, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany
| |
Collapse
|
11
|
Nabizadeh F. Disruption in functional networks mediated tau spreading in Alzheimer's disease. Brain Commun 2024; 6:fcae198. [PMID: 38978728 PMCID: PMC11227975 DOI: 10.1093/braincomms/fcae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/27/2024] [Accepted: 06/07/2024] [Indexed: 07/10/2024] Open
Abstract
Alzheimer's disease may be conceptualized as a 'disconnection syndrome', characterized by the breakdown of neural connectivity within the brain as a result of amyloid-beta plaques, tau neurofibrillary tangles and other factors leading to progressive degeneration and shrinkage of neurons, along with synaptic dysfunction. It has been suggested that misfolded tau proteins spread through functional connections (known as 'prion-like' properties of tau). However, the local effect of tau spreading on the synaptic function and communication between regions is not well understood. I aimed to investigate how the spreading of tau aggregates through connections can locally influence functional connectivity. In total, the imaging data of 211 participants including 117 amyloid-beta-negative non-demented and 94 amyloid-beta-positive non-demented participants were recruited from the Alzheimer's Disease Neuroimaging Initiative. Furthermore, normative resting-state functional MRI connectomes were used to model tau spreading through functional connections, and functional MRI of the included participants was used to determine the effect of tau spreading on functional connectivity. I found that lower functional connectivity to tau epicentres is associated with tau spreading through functional connections in both amyloid-beta-negative and amyloid-beta-positive participants. Also, amyloid-beta-PET in tau epicentres mediated the association of tau spreading and functional connectivity to epicentres suggesting a partial mediating effect of amyloid-beta deposition in tau epicentres on the local effect of tau spreading on functional connectivity. My findings provide strong support for the notion that tau spreading through connection is locally associated with disrupted functional connectivity between tau epicentre and non-epicentre regions independent of amyloid-beta pathology. Also, I defined several groups based on the relationship between tau spreading and functional disconnection, which provides quantitative assessment to investigate susceptibility or resilience to functional disconnection related to tau spreading. I showed that amyloid-beta, other copathologies and the apolipoprotein E epsilon 4 allele can be a leading factor towards vulnerability to tau relative functional disconnection.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran 441265421414, Iran
| |
Collapse
|
12
|
Teipel S, Grazia A, Dyrba M, Grothe MJ, Pomara N. Basal forebrain volume and metabolism in carriers of the Colombian mutation for autosomal dominant Alzheimer's disease. Sci Rep 2024; 14:11268. [PMID: 38760448 PMCID: PMC11101449 DOI: 10.1038/s41598-024-60799-9] [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: 01/03/2024] [Accepted: 04/26/2024] [Indexed: 05/19/2024] Open
Abstract
We aimed to study atrophy and glucose metabolism of the cholinergic basal forebrain in non-demented mutation carriers for autosomal dominant Alzheimer's disease (ADAD). We determined the level of evidence for or against atrophy and impaired metabolism of the basal forebrain in 167 non-demented carriers of the Colombian PSEN1 E280A mutation and 75 age- and sex-matched non-mutation carriers of the same kindred using a Bayesian analysis framework. We analyzed baseline MRI, amyloid PET, and FDG-PET scans of the Alzheimer's Prevention Initiative ADAD Colombia Trial. We found moderate evidence against an association of carrier status with basal forebrain volume (Bayes factor (BF10) = 0.182). We found moderate evidence against a difference of basal forebrain metabolism (BF10 = 0.167). There was only inconclusive evidence for an association between basal forebrain volume and delayed memory and attention (BF10 = 0.884 and 0.184, respectively), and between basal forebrain volume and global amyloid load (BF10 = 2.1). Our results distinguish PSEN1 E280A mutation carriers from sporadic AD cases in which cholinergic involvement of the basal forebrain is already detectable in the preclinical and prodromal stages. This indicates an important difference between ADAD and sporadic AD in terms of pathogenesis and potential treatment targets.
Collapse
Affiliation(s)
- Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Gehlsheimer Str. 20, 18147, Rostock, Germany.
- Department of Psychosomatic Medicine, University Medicine Rostock, Gehlsheimer Str. 20, 18147, Rostock, Germany.
| | - Alice Grazia
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Martin Dyrba
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Michel J Grothe
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Nunzio Pomara
- Geriatric Psychiatry Division, Nathan Kline Institute/Department of Psychiatry and Pathology, NYU Grossman School of Medicine, Orangeburg, NY, USA
| |
Collapse
|
13
|
German‐Castelan L, Shanks HRC, Gros R, Saito T, Saido TC, Saksida LM, Bussey TJ, Prado MAM, Schmitz TW, Prado VF. Sex-dependent cholinergic effects on amyloid pathology: A translational study. Alzheimers Dement 2024; 20:995-1012. [PMID: 37846816 PMCID: PMC10916951 DOI: 10.1002/alz.13481] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION About two-thirds of Alzheimer's Disease (AD) patients are women, who exhibit more severe pathology and cognitive decline than men. Whether biological sex causally modulates the relationship between cholinergic signaling and amyloid pathology remains unknown. METHODS We quantified amyloid beta (Aβ) in male and female App-mutant mice with either decreased or increased cholinergic tone and examined the impact of ovariectomy and estradiol replacement in this relationship. We also investigated longitudinal changes in basal forebrain (cholinergic function) and Aβ in elderly individuals. RESULTS We show a causal relationship between cholinergic tone and amyloid pathology in males and ovariectomized female mice, which is decoupled in ovary-intact and ovariectomized females receiving estradiol. In elderly humans, cholinergic loss exacerbates Aβ. DISCUSSION Our findings emphasize the importance of reflecting human menopause in mouse models. They also support a role for therapies targeting estradiol and cholinergic signaling to reduce Aβ. HIGHLIGHTS Cholinergic tone regulates amyloid beta (Aβ) pathology in males and ovariectomized female mice. Estradiol uncouples the relationship between cholinergic tone and Aβ. In elderly humans, cholinergic loss correlates with increased Aβ in both sexes.
Collapse
Affiliation(s)
- Liliana German‐Castelan
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Hayley R. C. Shanks
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Robert Gros
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of MedicineSchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Takashi Saito
- Department of Neurocognitive ScienceInstitute of Brain ScienceNagoya City University Graduate School of Medical SciencesNagoyaJapan
- Laboratory for Proteolytic NeuroscienceRIKEN Center for Brain ScienceWako, SaitamaJapan
| | - Takaomi C. Saido
- Laboratory for Proteolytic NeuroscienceRIKEN Center for Brain ScienceWako, SaitamaJapan
| | - Lisa M. Saksida
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
| | - Timothy J. Bussey
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
| | - Marco A. M. Prado
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | - Taylor W. Schmitz
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Lawson Health Research InstituteSt. Joseph's HospitalLondonOntarioCanada
| | - Vania F. Prado
- Robarts Research InstituteSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Neuroscience programSchulich School of Medicine and DentistryUniversity of Western OntarioLondonOntarioCanada
- Department of Physiology and PharmacologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
- Western Institute for NeuroscienceUniversity of Western OntarioLondonOntarioCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine & DentistryUniversity of Western OntarioLondonOntarioCanada
| | | |
Collapse
|
14
|
Yoo HS, Kim HK, Lee JH, Chun JH, Lee HS, Grothe MJ, Teipel S, Cavedo E, Vergallo A, Hampel H, Ryu YH, Cho H, Lyoo CH. Association of Basal Forebrain Volume with Amyloid, Tau, and Cognition in Alzheimer's Disease. J Alzheimers Dis 2024; 99:145-159. [PMID: 38640150 DOI: 10.3233/jad-230975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Background Degeneration of cholinergic basal forebrain (BF) neurons characterizes Alzheimer's disease (AD). However, what role the BF plays in the dynamics of AD pathophysiology has not been investigated precisely. Objective To investigate the baseline and longitudinal roles of BF along with core neuropathologies in AD. Methods In this retrospective cohort study, we enrolled 113 subjects (38 amyloid [Aβ]-negative cognitively unimpaired, 6 Aβ-positive cognitively unimpaired, 39 with prodromal AD, and 30 with AD dementia) who performed brain MRI for BF volume and cortical thickness, 18F-florbetaben PET for Aβ, 18F-flortaucipir PET for tau, and detailed cognitive testing longitudinally. We investigated the baseline and longitudinal association of BF volume with Aβ and tau standardized uptake value ratio and cognition. Results Cross-sectionally, lower BF volume was not independently associated with higher cortical Aβ, but it was associated with tau burden. Tau burden in the orbitofrontal, insular, lateral temporal, inferior temporo-occipital, and anterior cingulate cortices were associated with progressive BF atrophy. Lower BF volume was associated with faster Aβ accumulation, mainly in the prefrontal, anterior temporal, cingulate, and medial occipital cortices. BF volume was associated with progressive decline in language and memory functions regardless of baseline Aβ and tau burden. Conclusions Tau deposition affected progressive BF atrophy, which in turn accelerated amyloid deposition, leading to a vicious cycle. Also, lower baseline BF volume independently predicted deterioration in cognitive function.
Collapse
Affiliation(s)
- Han Soo Yoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Han-Kyeol Kim
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae-Hoon Lee
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joong-Hyun Chun
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Sun Lee
- Department of Biostatistics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Michel J Grothe
- Reina Sofia Alzheimer Center, CIEN Foundation-ISCIII, Madrid, Spain
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)-Rostock/Greifswald, Rostock, Germany
- Department of Psychosomatic Medicine, University Medicine Rostock, Germany
| | - Enrica Cavedo
- Sorbonne University Alzheimer Precision Medicine, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University Alzheimer Precision Medicine, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Harald Hampel
- Sorbonne University Alzheimer Precision Medicine, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
15
|
Rozalem Aranha M, Iulita MF, Montal V, Pegueroles J, Bejanin A, Vaqué-Alcázar L, Grothe MJ, Carmona-Iragui M, Videla L, Benejam B, Arranz J, Padilla C, Valldeneu S, Barroeta I, Altuna M, Fernández S, Ribas L, Valle-Tamayo N, Alcolea D, González-Ortiz S, Bargalló N, Zetterberg H, Blennow K, Blesa R, Wisniewski T, Busciglio J, Cuello AC, Lleó A, Fortea J. Basal forebrain atrophy along the Alzheimer's disease continuum in adults with Down syndrome. Alzheimers Dement 2023; 19:4817-4827. [PMID: 37021589 DOI: 10.1002/alz.12999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Basal forebrain (BF) degeneration occurs in Down syndrome (DS)-associated Alzheimer's disease (AD). However, the dynamics of BF atrophy with age and disease progression, its impact on cognition, and its relationship with AD biomarkers have not been studied in DS. METHODS We included 234 adults with DS (150 asymptomatic, 38 prodromal AD, and 46 AD dementia) and 147 euploid controls. BF volumes were extracted from T-weighted magnetic resonance images using a stereotactic atlas in SPM12. We assessed BF volume changes with age and along the clinical AD continuum and their relationship to cognitive performance, cerebrospinal fluid (CSF) and plasma amyloid/tau/neurodegeneration biomarkers, and hippocampal volume. RESULTS In DS, BF volumes decreased with age and along the clinical AD continuum and significantly correlated with amyloid, tau, and neurofilament light chain changes in CSF and plasma, hippocampal volume, and cognitive performance. DISCUSSION BF atrophy is a potentially valuable neuroimaging biomarker of AD-related cholinergic neurodegeneration in DS.
Collapse
Affiliation(s)
- Mateus Rozalem Aranha
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Lídia Vaqué-Alcázar
- Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michel J Grothe
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Bessy Benejam
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Javier Arranz
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Concepción Padilla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sílvia Valldeneu
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Susana Fernández
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Laia Ribas
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Natalia Valle-Tamayo
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sofía González-Ortiz
- Hospital del Mar - Parc de Salut Mar, Barcelona, Spain
- Neuroradiology Section, Radiology Department, Diagnostic Image Center, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Núria Bargalló
- Neuroradiology Section, Radiology Department, Diagnostic Image Center, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
- Magnetic Resonance Image Core Facility (IDIBAPS), Barcelona, Spain
| | - Henrik Zetterberg
- Queen Square Institute of Neurology, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute, University College London, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, China, Hong Kong
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry and Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, New York, USA
| | - Jorge Busciglio
- Department of Neurobiology & Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, California, USA
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology, Oxford University, Oxford, UK
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| |
Collapse
|
16
|
Alam JJ, Nixon RA. Drug development targeting degeneration of the basal forebrain cholinergic system: its time has come. Mol Neurodegener 2023; 18:74. [PMID: 37794391 PMCID: PMC10552418 DOI: 10.1186/s13024-023-00663-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 09/29/2023] [Indexed: 10/06/2023] Open
Affiliation(s)
- John J Alam
- CervoMed Inc., 20 Park Plaza, Suite 424, Boston, MA, 02116, USA.
| | - Ralph A Nixon
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Departments of Cell Biology and Psychiatry, NYU Langone Medical Center, NYU Neuroscience Institute, New York, NY, USA
| |
Collapse
|
17
|
Yang A, Li G. Nucleus basalis of Meynert predicts cognitive changes in isolated REM sleep behavior disorder. Sleep Med 2023; 109:11-17. [PMID: 37393717 DOI: 10.1016/j.sleep.2023.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Degeneration of the nucleus basalis of Meynert (NBM) has been implicated in cognitive impairments in Parkinson's disease. The role of the NBM volumes in the cognitive function in isolated rapid eye movement (REM) sleep behavior disorder (iRBD) has not been explored. METHOD We investigated changes in NBM volumes and their associations with cognitive deficits in iRBD. Baseline NBM volumes were compared between 29 iRBD patients and 29 healthy controls by using structural MRI data from the Parkinson Progression Marker Initiative database. Partial correlation analyses were used to evaluate cross-sectional relationships between baseline NBM volumes and cognitive performance in iRBD. Linear mixed models were applied to assess between-group differences in longitudinal cognitive changes, and whether baseline NBM volumes could predict longitudinal changes of cognition in iRBD. RESULTS Compared with controls, NBM volumes were significantly reduced in iRBD patients. In patients with iRBD, higher NBM volumes were significantly associated with greater performance in global cognition function. In the longitudinal analyses, iRBD patients showed more severe and rapid decline on tests of global cognition compared to healthy controls. Furthermore, greater baseline NBM volumes were significantly associated with greater follow-up Montreal Cognitive Assessment (MoCA) scores, thus predicting less longitudinal cognitive changes in iRBD. CONCLUSION This study provides important in vivo evidence for an association between the NBM degeneration and cognitive impairments in iRBD.
Collapse
Affiliation(s)
- Amei Yang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guanglu Li
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
18
|
Teipel S, Grothe MJ. MRI-based basal forebrain atrophy and volumetric signatures associated with limbic TDP-43 compared to Alzheimer's disease pathology. Neurobiol Dis 2023; 180:106070. [PMID: 36898615 DOI: 10.1016/j.nbd.2023.106070] [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: 01/04/2023] [Revised: 02/23/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND It is not clear to which degree limbic TDP-43 pathology associates with a cholinergic deficit in the absence of Alzheimer's disease (AD) pathology. OBJECTIVE Replicate and extend recent evidence on cholinergic basal forebrain atrophy in limbic TDP-43 and evaluate MRI based patterns of atrophy as a surrogate marker for TDP-43. METHODS We studied ante-mortem MRI data of 11 autopsy cases with limbic TDP-43 pathology, 47 cases with AD pathology, and 26 mixed AD/TDP-43 cases from the ADNI autopsy sample, and 17 TDP-43, 170 AD, and 58 mixed AD/TDP-43 cases from the NACC autopsy sample. Group differences in basal forebrain and other brain volumes of interest were assessed using Bayesian ANCOVA. We assessed the diagnostic utility of MRI based patterns of brain atrophy using voxel-based receiver operating characteristics and random forest analyses. RESULTS In the NACC sample, we found moderate evidence for the absence of a difference in basal forebrain volumes between AD, TDP-43, and mixed pathologies (Bayes factor(BF)10 = 0.324), and very strong evidence for lower hippocampus volume in TDP-43 and mixed cases compared with AD cases (BF10 = 156.1). The ratio of temporal to hippocampus volume reached an AUC of 75% for separating pure TDP-43 from pure AD cases. Random-forest analysis between TDP-43, AD, and mixed pathology reached only a multiclass AUC of 0.63 based on hippocampus, middle-inferior temporal gyrus, and amygdala volumes. Findings in the ADNI sample were consistent with these results. CONCLUSION A comparable degree of basal forebrain atrophy in pure TDP-43 cases compared to AD cases encourages studies on the effect of cholinergic treatment in amnestic dementia due to TDP-43. A distinct pattern of temporo-limbic brain atrophy may serve as a surrogate marker to enrich samples in clinical trials for the presence of TDP-43 pathology.
Collapse
Affiliation(s)
- Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany.
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| |
Collapse
|
19
|
Liu X, Zeng Q, Luo X, Li K, Xu X, Hong L, Li J, Guan X, Xu X, Huang P, Zhang M. Effects of APOE ε2 allele on basal forebrain functional connectivity in mild cognitive impairment. CNS Neurosci Ther 2022; 29:597-608. [PMID: 36468416 PMCID: PMC9873529 DOI: 10.1111/cns.14038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 12/10/2022] Open
Abstract
BACKGROUND Basal forebrain cholinergic system (BFCS) dysfunction is associated with cognitive decline in Alzheimer's disease (AD) and mild cognitive impairment (MCI). Apolipoprotein E (APOE) ε2 is a protective genetic factor in AD and MCI, and cholinergic sprouting depends on APOE. OBJECTIVE We investigated the effect of the APOE ε2 allele on BFCS functional connectivity (FC) in cognitively normal (CN) subjects and MCI patients. METHOD We included 60 MCI patients with APOE ε3/ε3, 18 MCI patients with APOE ε2/ε3, 73 CN subjects with APOE ε3/ε3, and 36 CN subjects with APOE ε2/ε3 genotypes who had resting-state functional magnetic resonance imaging data from the Alzheimer's disease Neuroimaging Initiative. We used BFCS subregions (Ch1-3 and Ch4) as seeds and calculated the FC with other brain areas. Using a mixed-effect analysis, we explored the interaction effects of APOE ε2 allele × cognitive status on BFCS-FC. Furthermore, we examined the relationships between imaging metrics, cognitive abilities, and AD pathology markers, controlling for sex, age, and education as covariates. RESULTS An interaction effect on functional connectivity was found between the right Ch4 (RCh4) and left insula (p < 0.05, corrected), and between the RCh4 and left Rolandic operculum (p < 0.05, corrected). Among all subjects and APOE ε2 carriers, RCh4-left Insula FC was associated with early tau deposition. Furthermore, no correlation was found between imaging metrics and amyloid burden. Among all subjects and APOE ε2 carriers, FC metrics were associated with cognitive performance. CONCLUSION The APOE ε2 genotype may play a protective role during BFCS degeneration in MCI.
Collapse
Affiliation(s)
- Xiaocao Liu
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Qingze Zeng
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiao Luo
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Kaicheng Li
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaopei Xu
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Luwei Hong
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Jixuan Li
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaojun Guan
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Xiaojun Xu
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Peiyu Huang
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Min‐Ming Zhang
- Department of RadiologyThe 2nd Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | | |
Collapse
|
20
|
Mohanty R, Ferreira D, Frerich S, Muehlboeck JS, Grothe MJ, Westman E. Neuropathologic Features of Antemortem Atrophy-Based Subtypes of Alzheimer Disease. Neurology 2022; 99:e323-e333. [PMID: 35609990 PMCID: PMC9421777 DOI: 10.1212/wnl.0000000000200573] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/04/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To investigate whether antemortem MRI-based atrophy subtypes of Alzheimer disease (AD) differ in neuropathologic features and comorbid non-AD pathologies at postmortem. METHODS From the Alzheimer's Disease Neuroimaging Initiative cohort, we included individuals with antemortem MRI evaluating brain atrophy within 2 years before death, antemortem diagnosis of AD dementia/mild cognitive impairment, and postmortem-confirmed AD neuropathologic change. Antemortem atrophy subtypes were modeled as continuous phenomena based on a recent conceptual framework: typicality (spanning limbic-predominant AD to hippocampal-sparing AD) and severity (spanning typical AD to minimal atrophy AD). Postmortem neuropathologic evaluation included AD hallmarks, β-amyloid, and tau as well as non-AD pathologies, alpha-synuclein and TAR DNA-binding protein 43 (TDP-43). We also investigated the overall concomitance across these pathologies. Partial correlations assessed the associations between antemortem atrophy subtypes and postmortem neuropathologic outcomes. RESULTS In 31 individuals (26 AD dementia/5 mild cognitive impairment, mean age = 80 years, 26% females), antemortem typicality was significantly negatively associated with neuropathologic features, including β-amyloid (rho = -0.39 overall), tau (rho = -0.38 regionally), alpha-synuclein (rho = -0.39 regionally), TDP-43 (rho = -0.49 overall), and concomitance of pathologies (rho = -0.59 regionally). Limbic-predominant AD was associated with higher Thal phase, neuritic plaque density, and presence of TDP-43 compared with hippocampal-sparing AD. Regionally, limbic-predominant AD showed a higher presence of tau and alpha-synuclein pathologies in medial temporal structures, a higher presence of TDP-43, and concomitance of pathologies subcortically/cortically compared with hippocampal-sparing AD. Antemortem severity was significantly negatively associated with concomitance of pathologies (rho = -0.43 regionally), such that typical AD showed higher concomitance of pathologies than minimal atrophy AD. DISCUSSION We provide a direct antemortem-to-postmortem validation, highlighting the importance of understanding atrophy-based heterogeneity in AD relative to AD and non-AD pathologies. We suggest that (1) typicality and severity in atrophy reflect differential aspects of susceptibility of the brain to AD and non-AD pathologies; and (2) limbic-predominant AD and typical AD subtypes share similar biological pathways, making them more vulnerable to AD and non-AD pathologies compared with hippocampal-sparing AD, which may follow a different biological pathway. Our findings provide a deeper understanding of associations of atrophy subtypes in AD with different pathologies, enhancing the prevailing knowledge of biological heterogeneity in AD and could contribute toward tracking disease progression and designing clinical trials in the future.
Collapse
Affiliation(s)
- Rosaleena Mohanty
- From the Division of Clinical Geriatrics (R.M., D.F., S.F., J.S.M., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Radiology (D.F.), Mayo Clinic, Rochester, MN; Institute for Stroke and Dementia Research (E.W.), University Hospital, Ludwig-Maximilian-University (LMU) Munich, Germany; Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Clinical Dementia Research Section (M.J.G.), German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.
| | - Daniel Ferreira
- From the Division of Clinical Geriatrics (R.M., D.F., S.F., J.S.M., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Radiology (D.F.), Mayo Clinic, Rochester, MN; Institute for Stroke and Dementia Research (E.W.), University Hospital, Ludwig-Maximilian-University (LMU) Munich, Germany; Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Clinical Dementia Research Section (M.J.G.), German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Simon Frerich
- From the Division of Clinical Geriatrics (R.M., D.F., S.F., J.S.M., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Radiology (D.F.), Mayo Clinic, Rochester, MN; Institute for Stroke and Dementia Research (E.W.), University Hospital, Ludwig-Maximilian-University (LMU) Munich, Germany; Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Clinical Dementia Research Section (M.J.G.), German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - J-Sebastian Muehlboeck
- From the Division of Clinical Geriatrics (R.M., D.F., S.F., J.S.M., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Radiology (D.F.), Mayo Clinic, Rochester, MN; Institute for Stroke and Dementia Research (E.W.), University Hospital, Ludwig-Maximilian-University (LMU) Munich, Germany; Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Clinical Dementia Research Section (M.J.G.), German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Michel J Grothe
- From the Division of Clinical Geriatrics (R.M., D.F., S.F., J.S.M., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Radiology (D.F.), Mayo Clinic, Rochester, MN; Institute for Stroke and Dementia Research (E.W.), University Hospital, Ludwig-Maximilian-University (LMU) Munich, Germany; Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Clinical Dementia Research Section (M.J.G.), German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Eric Westman
- From the Division of Clinical Geriatrics (R.M., D.F., S.F., J.S.M., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Radiology (D.F.), Mayo Clinic, Rochester, MN; Institute for Stroke and Dementia Research (E.W.), University Hospital, Ludwig-Maximilian-University (LMU) Munich, Germany; Unidad de Trastornos del Movimiento (M.J.G.), Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Clinical Dementia Research Section (M.J.G.), German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
21
|
Schumacher J, Ray NJ, Hamilton CA, Donaghy PC, Firbank M, Roberts G, Allan L, Durcan R, Barnett N, O’Brien JT, Taylor JP, Thomas AJ. Cholinergic white matter pathways in dementia with Lewy bodies and Alzheimer's disease. Brain 2022; 145:1773-1784. [PMID: 34605858 PMCID: PMC9166545 DOI: 10.1093/brain/awab372] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/23/2021] [Accepted: 09/06/2021] [Indexed: 11/17/2022] Open
Abstract
Patients who have dementia with Lewy bodies and Alzheimer's disease show early degeneration of the cholinergic nucleus basalis of Meynert. However, how white matter projections between the nucleus basalis of Meynert and the cortex are altered in neurodegenerative disease is unknown. Tractography of white matter pathways originating from the nucleus basalis of Meynert was performed using diffusion-weighted imaging in 46 patients with Alzheimer's disease dementia, 48 with dementia with Lewy bodies, 35 with mild cognitive impairment with Alzheimer's disease, 38 with mild cognitive impairment with Lewy bodies and 71 control participants. Mean diffusivity of the resulting pathways was compared between groups and related to cognition, attention, functional EEG changes and dementia conversion in the mild cognitive impairment groups. We successfully tracked a medial and a lateral pathway from the nucleus basalis of Meynert. Mean diffusivity of the lateral pathway was higher in both dementia and mild cognitive impairment groups than controls (all P < 0.03). In the patient groups, increased mean diffusivity of this pathway was related to more impaired global cognition (β = -0.22, P = 0.06) and worse performance on an attention task (β = 0.30, P = 0.03). In patients with mild cognitive impairment, loss of integrity of both nucleus basalis of Meynert pathways was associated with increased risk of dementia progression [hazard ratio (95% confidence interval), medial pathway: 2.51 (1.24-5.09); lateral pathway: 2.54 (1.24-5.19)]. Nucleus basalis of Meynert volume was reduced in all clinical groups compared to controls (all P < 0.001), but contributed less strongly to cognitive impairment and was not associated with attention or dementia conversion. EEG slowing in the patient groups as assessed by a decrease in dominant frequency was associated with smaller nucleus basalis of Meynert volumes (β = 0.22, P = 0.02) and increased mean diffusivity of the lateral pathway (β = -0.47, P = 0.003). We show that degeneration of the cholinergic nucleus basalis of Meynert in Alzheimer's disease and dementia with Lewy bodies is accompanied by an early reduction in integrity of white matter projections that originate from this structure. This is more strongly associated with cognition and attention than the volume of the nucleus basalis of Meynert itself and might be an early indicator of increased risk of dementia conversion in people with mild cognitive impairment.
Collapse
Affiliation(s)
- Julia Schumacher
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Nicola J Ray
- Health, Psychology and Communities Research Centre, Department of Psychology, Manchester Metropolitan University, Manchester, UK
| | - Calum A Hamilton
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Paul C Donaghy
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Michael Firbank
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Gemma Roberts
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Louise Allan
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
- Institute of Health Research, University of Exeter, Exeter, UK
| | - Rory Durcan
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Nicola Barnett
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge School of Medicine, Cambridge CB2 0SP, UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Alan J Thomas
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
22
|
Bashir S, Uzair M, Abualait T, Arshad M, Khallaf RA, Niaz A, Thani Z, Yoo WK, Túnez I, Demirtas-Tatlidede A, Meo SA. Effects of transcranial magnetic stimulation on neurobiological changes in Alzheimer's disease (Review). Mol Med Rep 2022; 25:109. [PMID: 35119081 PMCID: PMC8845030 DOI: 10.3892/mmr.2022.12625] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/15/2021] [Indexed: 11/05/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and brain neuronal loss. A pioneering field of research in AD is brain stimulation via electromagnetic fields (EMFs), which may produce clinical benefits. Noninvasive brain stimulation techniques, such as transcranial magnetic stimulation (TMS), have been developed to treat neurological and psychiatric disorders. The purpose of the present review is to identify neurobiological changes, including inflammatory, neurodegenerative, apoptotic, neuroprotective and genetic changes, which are associated with repetitive TMS (rTMS) treatment in patients with AD. Furthermore, it aims to evaluate the effect of TMS treatment in patients with AD and to identify the associated mechanisms. The present review highlights the changes in inflammatory and apoptotic mechanisms, mitochondrial enzymatic activities, and modulation of gene expression (microRNA expression profiles) associated with rTMS or sham procedures. At the molecular level, it has been suggested that EMFs generated by TMS may affect the cell redox status and amyloidogenic processes. TMS may also modulate gene expression by acting on both transcriptional and post‑transcriptional regulatory mechanisms. TMS may increase brain cortical excitability, induce specific potentiation phenomena, and promote synaptic plasticity and recovery of impaired functions; thus, it may re‑establish cognitive performance in patients with AD.
Collapse
Affiliation(s)
- Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic and Applied Sciences, International Islamic University Islamabad, Islamabad 44000, Pakistan
| | - Turki Abualait
- College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province 34212, Saudi Arabia
| | - Muhammad Arshad
- Department of Biological Sciences, Faculty of Basic and Applied Sciences, International Islamic University Islamabad, Islamabad 44000, Pakistan
| | - Roaa A. Khallaf
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Asim Niaz
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Ziyad Thani
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Eastern Province 32253, Saudi Arabia
| | - Woo-Kyoung Yoo
- Department of Physical Medicine and Rehabilitation, Hallym University College of Medicine, Anyang, Gyeonggi-do 24252, Republic of Korea
| | - Isaac Túnez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Nursing/ Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Cordoba, Cordoba 14071, Spain
- Cooperative Research Thematic Excellent Network on Brain Stimulation (REDESTIM), Ministry for Economy, Industry and Competitiveness, 28046 Madrid, Spain
| | | | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
23
|
Duong MT, Das SR, Lyu X, Xie L, Richardson H, Xie SX, Yushkevich PA, Wolk DA, Nasrallah IM. Dissociation of tau pathology and neuronal hypometabolism within the ATN framework of Alzheimer's disease. Nat Commun 2022; 13:1495. [PMID: 35314672 PMCID: PMC8938426 DOI: 10.1038/s41467-022-28941-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/11/2022] [Indexed: 11/08/2022] Open
Abstract
Alzheimer's disease (AD) is defined by amyloid (A) and tau (T) pathologies, with T better correlated to neurodegeneration (N). However, T and N have complex regional relationships in part related to non-AD factors that influence N. With machine learning, we assessed heterogeneity in 18F-flortaucipir vs. 18F-fluorodeoxyglucose positron emission tomography as markers of T and neuronal hypometabolism (NM) in 289 symptomatic patients from the Alzheimer's Disease Neuroimaging Initiative. We identified six T/NM clusters with differing limbic and cortical patterns. The canonical group was defined as the T/NM pattern with lowest regression residuals. Groups resilient to T had less hypometabolism than expected relative to T and displayed better cognition than the canonical group. Groups susceptible to T had more hypometabolism than expected given T and exhibited worse cognitive decline, with imaging and clinical measures concordant with non-AD copathologies. Together, T/NM mismatch reveals distinct imaging signatures with pathobiological and prognostic implications for AD.
Collapse
Affiliation(s)
- Michael Tran Duong
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Sandhitsu R Das
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xueying Lyu
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Long Xie
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hayley Richardson
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon X Xie
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Yushkevich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ilya M Nasrallah
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Teipel SJ, Grothe MJ. Ante-mortem basal forebrain atrophy in pure limbic TAR DNA-binding protein 43 pathology compared with pure Alzheimer pathology. Eur J Neurol 2022; 29:1394-1401. [PMID: 35122358 DOI: 10.1111/ene.15270] [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: 10/28/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Currently, the extent of cholinergic basal forebrain atrophy in relatively pure limbic TAR DNA-binding protein 43 (TDP-43) pathology compared with relatively pure Alzheimer's disease (AD) is unclear. METHODS We compared ante-mortem MRI based atrophy of the basal forebrain and medial and lateral temporal lobe volumes between 10 autopsy cases with limbic TDP-43 pathology and 33 cases with AD pathology on post-mortem neuropathologic examination from the ADNI cohort. For reference, we studied MRI volumes from cognitively healthy, amyloid PET negative people (n = 145). Group differences were assessed using Bayesian ANCOVA. In addition, we assessed brain-wide regional volume changes using partial least-squares regression (PLSR). RESULTS We found extreme evidence (Bayes factor (BF)01 > 600) for a smaller basal forebrain volume in both TDP-43 and AD cases compared with amyloid-negative controls, and moderate evidence (BF01 = 4.9) that basal forebrain volume was not larger in TDP-43 than in AD cases. The ratio of hippocampus to lateral temporal lobe volumes discriminated between TDP-43 and AD cases with an accuracy of 0.78. PLSR showed higher grey matter in lateral temporal lobes and cingulate and precuneus, and reduced grey matter in precentral and postcentral gyri and hippocampus in TDP-43 compared with AD cases. CONLCUSIONS Atrophy of the cholinergic basal forebrain appears to be similarly pronounced in cases with limbic TDP-43 pathology as in AD. This suggests that a clinical trial of the efficacy of cholinesterase inhibitors in amyloid-negative cases with amnestic dementia and an imaging signature of TDP-43 pathology may be warranted.
Collapse
Affiliation(s)
- Stefan J Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, Rostock, Germany.,Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany
| | - Michel J Grothe
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, Rostock, Germany.,Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Seville, Spain
| | | |
Collapse
|
25
|
Richter N, David LS, Grothe MJ, Teipel S, Dietlein M, Tittgemeyer M, Neumaier B, Fink GR, Onur OA, Kukolja J. Age and Anterior Basal Forebrain Volume Predict the Cholinergic Deficit in Patients with Mild Cognitive Impairment due to Alzheimer’s Disease. J Alzheimers Dis 2022; 86:425-440. [DOI: 10.3233/jad-210261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background: Early and severe neuronal loss in the cholinergic basal forebrain is observed in Alzheimer’s disease (AD). To date, cholinomimetics play a central role in the symptomatic treatment of AD dementia. Although basic research indicates that a cholinergic deficit is present in AD before dementia, the efficacy of cholinomimetics in mild cognitive impairment (MCI) remains controversial. Predictors of cholinergic impairment could guide individualized therapy. Objective: To investigate if the extent of the cholinergic deficit, measured using positron emission tomography (PET) and the tracer 11C-N-methyl-4-piperidyl acetate (MP4A), could be predicted from the volume of cholinergic basal forebrain nuclei in non-demented AD patients. Methods: Seventeen patients with a high likelihood of MCI due to AD and 18 age-matched cognitively healthy adults underwent MRI-scanning. Basal forebrain volume was assessed using voxel-based morphometry and a cytoarchitectonic atlas of cholinergic nuclei. Cortical acetylcholinesterase (AChE) activity was measured using MP4A-PET. Results: Cortical AChE activity and nucleus basalis of Meynert (Ch4 area) volume were significantly decreased in MCI. The extent of the cholinergic deficit varied considerably across patients. Greater volumes of anterior basal forebrain nuclei (Ch1/2 area) and younger age (Spearman’s rho (17) = –0.596, 95% -CI [–0.905, –0.119] and 0.593, 95% -CI [0.092, 0.863])) were associated with a greater cholinergic deficit. Conclusion: Data suggest that less atrophy of the Ch1/2 area and younger age are associated with a more significant cholinergic deficit in MCI due to AD. Further investigations are warranted to determine if the individual response to cholinomimetics can be inferred from these measures.
Collapse
Affiliation(s)
- Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
| | - Lara-Sophia David
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Michel J. Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Movement Disorders Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Marc Tittgemeyer
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
| | - Bernd Neumaier
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Research Center Jülich, Jülich, Germany
- Institute for Radiochemistry and Experimental Molecular Imaging, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Gereon R. Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Oezguer A. Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Juraj Kukolja
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
- Department of Neurology and Clinical Neurophysiology, Helios University Hospital Wuppertal, Wuppertal, Germany
- Faculty of Health, Witten/Herdecke University, Witten, Germany
| |
Collapse
|
26
|
Kantarci K, Nedelska Z, Chen Q, Senjem ML, Schwarz CG, Gunter JL, Przybelski SA, Lesnick TG, Kremers WK, Fields JA, Graff-Radford J, Savica R, Jones D, Botha H, Knopman DS, Lowe V, Graff-Radford NR, Murray MM, Dickson DW, Reichard RR, Jack CR, Petersen RC, Ferman TJ, Boeve BF. OUP accepted manuscript. Brain Commun 2022; 4:fcac013. [PMID: 35415608 PMCID: PMC8994111 DOI: 10.1093/braincomms/fcac013] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 11/10/2021] [Accepted: 02/02/2022] [Indexed: 12/02/2022] Open
Abstract
Mild cognitive impairment with the core clinical features of dementia with Lewy bodies is recognized as a prodromal stage of dementia with Lewy bodies. Although grey matter atrophy has been demonstrated in prodromal dementia with Lewy bodies, longitudinal rates of atrophy during progression to probable dementia with Lewy bodies are unknown. We investigated the regional patterns of cross-sectional and longitudinal rates of grey matter atrophy in prodromal dementia with Lewy bodies, including those who progressed to probable dementia with Lewy bodies. Patients with mild cognitive impairment with at least one core clinical feature of dementia with Lewy bodies (mean age = 70.5; 95% male), who were enrolled in the Mayo Clinic Alzheimer’s Disease Research Center and followed for at least two clinical evaluations and MRI examinations, were included (n = 56). A cognitively unimpaired control group (n = 112) was matched 2:1 to the patients with mild cognitive impairment by age and sex. Patients either remained stable (n = 28) or progressed to probable dementia with Lewy bodies (n = 28) during a similar follow-up period and pathologic confirmation was available in a subset of cases (n = 18). Cross-sectional and longitudinal rates of grey matter atrophy were assessed using voxel-based and atlas-based region of interest analyses. At baseline, prodromal dementia with Lewy bodies was characterized by atrophy in the nucleus basalis of Meynert both in those who remained stable and those who progressed to probable dementia with Lewy bodies (P < 0.05 false discovery rate corrected). Increase in longitudinal grey matter atrophy rates were widespread, with greatest rates of atrophy observed in the enthorhinal and parahippocampal cortices, temporoparietal association cortices, thalamus and the basal ganglia, in mild cognitive impairment patients who progressed to probable dementia with Lewy bodies at follow-up (P < 0.05 false discovery rate corrected). Rates of inferior temporal atrophy were associated with greater rates of worsening on the clinical dementia rating–sum of boxes. Seventeen of the 18 (94%) autopsied cases had Lewy body disease. Results show that atrophy in the nucleus basalis of Meynert is a feature of prodromal dementia with Lewy bodies regardless of proximity to progression to probable dementia with Lewy bodies. Longitudinally, grey matter atrophy progresses in regions with significant cholinergic innervation, in alignment with clinical disease progression, with widespread and accelerated rates of atrophy in patients who progress to probable dementia with Lewy bodies. Given the prominent neurodegeneration in the cholinergic system, patients with prodromal dementia with Lewy bodies may be candidates for cholinesterase inhibitor treatment.
Collapse
Affiliation(s)
- Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Correspondence to: Kejal Kantarci, MD, MS Department of Radiology Mayo Clinic 200 First Street SW Rochester, MN 55905, USA E-mail:
| | - Zuzana Nedelska
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Charles University, Prague, Czech Republic
| | - Qin Chen
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | - Walter K. Kremers
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Julie A. Fields
- Department of Psychology and Psychiatry, Mayo Clinic, Rochester, MN, USA
| | | | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - David Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Melissa M. Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Dennis W. Dickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Tanis J. Ferman
- Department of Psychology and Psychiatry, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
27
|
Lin CP, Frigerio I, Boon BDC, Zhou Z, Rozemuller AJM, Bouwman FH, Schoonheim MM, van de Berg WDJ, Jonkman LE. OUP accepted manuscript. Brain 2022; 145:2869-2881. [PMID: 35259207 PMCID: PMC9420016 DOI: 10.1093/brain/awac093] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/23/2022] [Accepted: 02/13/2022] [Indexed: 11/24/2022] Open
Abstract
Cognitive deficits in Alzheimer’s disease, specifically amnestic (memory dominant) deficits, are associated with cholinergic degeneration in the basal forebrain. The cholinergic nucleus within the basal forebrain, the nucleus basalis of Meynert, exhibits local atrophy and reduced cortical tract integrity on MRI, and reveals amyloid-β and phosphorylated-tau pathology at autopsy. To understand the pathophysiology of nucleus basalis of Meynert atrophy and its neocortical projections in Alzheimer’s disease, we used a combined post-mortem in situ MRI and histopathology approach. A total of 19 Alzheimer’s disease (10 amnestic and nine non-amnestic) and nine non-neurological control donors underwent 3 T T1-weighted MRI for anatomical delineation and volume assessment of the nucleus basalis of Meynert, and diffusion-weighted imaging for microstructural assessment of the nucleus and its projections. At subsequent brain autopsy, tissue dissection and immunohistochemistry were performed for amyloid-β, phosphorylated-tau and choline acetyltransferase. Compared to controls, we observed an MRI-derived volume reduction and altered microstructural integrity of the nucleus basalis of Meynert in Alzheimer’s disease donors. Furthermore, decreased cholinergic cell density was associated with reduced integrity of the nucleus and its tracts to the temporal lobe, specifically to the temporal pole of the superior temporal gyrus, and the parahippocampal gyrus. Exploratory post hoc subgroup analyses indicated that cholinergic cell density could be associated with cortical tract alterations in amnestic Alzheimer’s disease donors only. Our study illustrates that in Alzheimer’s disease, cholinergic degeneration in the nucleus basalis of Meynert may contribute to damaged cortical projections, specifically to the temporal lobe, leading to cognitive deterioration.
Collapse
Affiliation(s)
- Chen Pei Lin
- Correspondence to: Chen-Pei Lin De Boelelaan 1117 1081 HV, Amsterdam, The Netherlands E-mail:
| | - Irene Frigerio
- Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Baayla D C Boon
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, Department of Neurology, Alzheimer centrum Amsterdam, Amsterdam, The Netherlands
| | - Zihan Zhou
- Zhejiang University, College of Biomedical Engineering and Instrument Science, Zhejiang, China
| | - Annemieke J M Rozemuller
- Amsterdam UMC, location VUmc, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Femke H Bouwman
- Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, Department of Neurology, Alzheimer centrum Amsterdam, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Laura E Jonkman
- Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Kilimann I, Wucherer D, Ittermann T, Völzke H, Bülow R, Hoffmann W, Grabe HJ, Wittfeld K, Teipel SJ. Inverse association between the anticholinergic burden and hippocampus volume in a population-based cohort across the entire adult age range. GeroScience 2021; 44:1715-1726. [PMID: 34940948 PMCID: PMC9213601 DOI: 10.1007/s11357-021-00497-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/12/2021] [Indexed: 11/28/2022] Open
Abstract
Many medications of different indications have a relevant anticholinergic activity. The anticholinergic burden of medication has been shown to have significant effects on the cognition and the risk for cognitive impairment and dementia particularly in older patients. So far, most of the studies used data from geriatric patients and the effect of the anticholinergic burden on brain structures is still unexplored. Our study aimed to analyze possible associations of hippocampus and cholinergic basal forebrain volumes as vulnerable brain structures for the development of dementia and the anticholinergic burden in a population-based cohort of non-demented participants spanning the adult age range from 21 to 80 years. We analyzed associations between medication-related anticholinergic burden and structural MRI volumes from participants (n = 3087, 52.2% female) of the population-based “Study of Health in Pomerania” (SHIP). Anticholinergic burden was obtained from the current medication plan using the Anticholinergic Burden Scale (ACB). All analyses were adjusted for age, sex, education, and total intracranial volume. We found statistically significant associations between the ACB and the left and right hippocampus volume but not for the basal forebrain cholinergic system. Complementary voxel-based analysis across all participants revealed FWE-corrected (p = < 0.05) clusters in the temporo-parietal regions reaching into frontal areas, showing reduced volumes with higher ACB scores. We identified an association between anticholinergic burden of medication on hippocampal volume suggesting a potential inverse effect of such medication. This association highlights the importance of a careful prescription of medication with anticholinergic activity at any adult age.
Collapse
Affiliation(s)
- Ingo Kilimann
- German Center for Neurodegenerative Disease (DZNE), Rostock/Greifswald, Gehlsheimer Straße 20, 18147, Rostock, Germany. .,Department Psychosomatic Medicine and Psychotherapy, Rostock University Medical Center, Gehlsheimer Straße 20, 18147, Rostock, Germany.
| | - Diana Wucherer
- German Center for Neurodegenerative Disease (DZNE), Rostock/Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Till Ittermann
- SHIP Study Unit, Institute for Community Medicine, University Medicine Greifswald, Walther-Rathenau-Str.48, 17475, Greifswald, Germany
| | - Henry Völzke
- SHIP Study Unit, Institute for Community Medicine, University Medicine Greifswald, Walther-Rathenau-Str.48, 17475, Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Wolfgang Hoffmann
- German Center for Neurodegenerative Disease (DZNE), Rostock/Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany.,Institute for Community Medicine, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Hans Jörgen Grabe
- German Center for Neurodegenerative Disease (DZNE), Rostock/Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Katharina Wittfeld
- German Center for Neurodegenerative Disease (DZNE), Rostock/Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Stefan Johannes Teipel
- German Center for Neurodegenerative Disease (DZNE), Rostock/Greifswald, Gehlsheimer Straße 20, 18147, Rostock, Germany.,Department Psychosomatic Medicine and Psychotherapy, Rostock University Medical Center, Gehlsheimer Straße 20, 18147, Rostock, Germany
| |
Collapse
|
29
|
Nazmuddin M, van Dalen JW, Borra RJH, Stormezand GN, van der Horn HJ, van der Zee S, Boertien J, van Laar T. Postural and gait symptoms in de novo Parkinson's disease patients correlate with cholinergic white matter pathology. Parkinsonism Relat Disord 2021; 93:43-49. [PMID: 34784526 DOI: 10.1016/j.parkreldis.2021.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/26/2021] [Accepted: 11/09/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The postural instability gait difficulty motor subtype of patients with Parkinson's disease (PIGD-PD) has been associated with more severe cognitive pathology and a higher risk on dementia compared to the tremor-dominant subtype (TD-PD). Here, we investigated whether the microstructural integrity of the cholinergic projections from the nucleus basalis of Meynert (NBM) was different between these clinical subtypes. METHODS Diffusion-weighted imaging data of 98 newly-diagnosed unmedicated PD patients (44 TD-PD and 54 PIGD-PD subjects) and 10 healthy controls, were analysed using diffusion tensor imaging, focusing on the white matter tracts associated with cholinergic projections from the NBM (NBM-WM) as the tract-of-interest. Quantitative tract-based and voxel-based analyses were performed using FA and MD as the estimates of white matter integrity. RESULTS Voxel-based analyses indicated significantly lower FA in the frontal part of the medial and lateral NBM-WM tract of both hemispheres of PIGD-PD compared to TD-PD. Relative to healthy control, several clusters with significantly lower FA were observed in the frontolateral NBM-WM tract of both disease groups. Furthermore, significant correlations between the severity of the axial and gait impairment and NBM-WM FA and MD were found, which were partially mediated by NBM-WM state on subjects' attentional performance. CONCLUSIONS The PIGD-PD subtype shows a loss of microstructural integrity of the NBM-WM tract, which suggests that a loss of cholinergic projections in this PD subtype already presents in de novo PD patients.
Collapse
Affiliation(s)
- Muhammad Nazmuddin
- Department of Neurology, Parkinson Expertise Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Jan-Willem van Dalen
- Department of Neurology, Donders Institute for Brain, Behaviour, and Cognition, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Neurology, Amsterdam UMC, Location AMC, Amsterdam, the Netherlands
| | - Ronald J H Borra
- Department of Radiology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Gilles N Stormezand
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Harm Jan van der Horn
- Department of Neurology, Parkinson Expertise Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sygrid van der Zee
- Department of Neurology, Parkinson Expertise Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jeffrey Boertien
- Department of Neurology, Parkinson Expertise Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Teus van Laar
- Department of Neurology, Parkinson Expertise Center, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
30
|
Teipel S, Bruno D, Plaska CR, Heslegrave A, Ramos-Cejudo J, Osorio RS, Zetterberg H, Blennow K, Pomara N. Association of CSF sTREM2, a marker of microglia activation, with cholinergic basal forebrain volume in major depressive disorder. J Affect Disord 2021; 293:429-434. [PMID: 34246952 PMCID: PMC8803308 DOI: 10.1016/j.jad.2021.06.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Inflammatory mechanisms are believed to contribute to the manifestation of major depressive disorder (MDD). Central cholinergic activity may moderate this effect. Here, we tested if volume of the cholinergic basal forebrain is associated with cerebrospinal fluid (CSF) levels of sTREM2 as a marker of microglial activation in people with late life MDD. METHODS Basal forebrain volume was determined from structural MRI scans and levels of CSF sTREM2 with immunoassay in 29 people with late-life MDD and 20 healthy older controls at baseline and 3 years follow-up. Associations were determined using Bayesian analysis of covariance. RESULTS We found moderate level of evidence for an association of lower CSF levels of sTREM2 at 3 years follow up with MDD (Bayes factor in favor of an effect = 7.9). This level of evidence prevailed when controlling for overall antidepressant treatment and CSF levels of markers of AD pathology, i.e., Aβ42/Aβ40, ptau181 and total tau. Evidence was in favor of absence of an effect for baseline levels of CSF sTREM2 in MDD cases and for baseline and follow up data in controls. LIMITATIONS The sample size of repeated CSF examinations was relatively small. Therefore, we used Bayesian sequential analysis to assess if effects were affected by sample size. Still, the number of cases was too small to stratify effects for different antidepressive treatments. CONCLUSIONS Our data agree with the assumption that central cholinergic system integrity may contribute to regulation of microglia activity in late-life MDD.
Collapse
Affiliation(s)
- Stefan Teipel
- Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany; DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany.
| | - Davide Bruno
- School of Psychology, Liverpool John Moores University, Liverpool, United Kingdom
| | | | - Amanda Heslegrave
- UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Jaime Ramos-Cejudo
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Ricardo S Osorio
- Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Nunzio Pomara
- Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
31
|
Alldred MJ, Lee SH, Stutzmann GE, Ginsberg SD. Oxidative Phosphorylation Is Dysregulated Within the Basocortical Circuit in a 6-month old Mouse Model of Down Syndrome and Alzheimer's Disease. Front Aging Neurosci 2021; 13:707950. [PMID: 34489678 PMCID: PMC8417045 DOI: 10.3389/fnagi.2021.707950] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/13/2021] [Indexed: 01/14/2023] Open
Abstract
Down syndrome (DS) is the primary genetic cause of intellectual disability (ID), which is due to the triplication of human chromosome 21 (HSA21). In addition to ID, HSA21 trisomy results in a number of neurological and physiological pathologies in individuals with DS, including progressive cognitive dysfunction and learning and memory deficits which worsen with age. Further exacerbating neurological dysfunction associated with DS is the concomitant basal forebrain cholinergic neuron (BFCN) degeneration and onset of Alzheimer's disease (AD) pathology in early mid-life. Recent single population RNA sequencing (RNA-seq) analysis in the Ts65Dn mouse model of DS, specifically the medial septal cholinergic neurons of the basal forebrain (BF), revealed the mitochondrial oxidative phosphorylation pathway was significantly impacted, with a large subset of genes within this pathway being downregulated. We further queried oxidative phosphorylation pathway dysregulation in Ts65Dn mice by examining genes and encoded proteins within brain regions comprising the basocortical system at the start of BFCN degeneration (6 months of age). In select Ts65Dn mice we demonstrate significant deficits in gene and/or encoded protein levels of Complex I-V of the mitochondrial oxidative phosphorylation pathway in the BF. In the frontal cortex (Fr Ctx) these complexes had concomitant alterations in select gene expression but not of the proteins queried from Complex I-V, suggesting that defects at this time point in the BF are more severe and occur prior to cortical dysfunction within the basocortical circuit. We propose dysregulation within mitochondrial oxidative phosphorylation complexes is an early marker of cognitive decline onset and specifically linked to BFCN degeneration that may propagate pathology throughout cortical memory and executive function circuits in DS and AD.
Collapse
Affiliation(s)
- Melissa J. Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Departments of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| | - Sang Han Lee
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, United States
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| | - Grace E. Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Discipline of Neuroscience, Rosalind Franklin University/The Chicago Medical School, North Chicago, IL, United States
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
- Departments of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
- Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, United States
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
32
|
Wisse LEM, Ravikumar S, Ittyerah R, Lim S, Lane J, Bedard ML, Xie L, Das SR, Schuck T, Grossman M, Lee EB, Tisdall MD, Prabhakaran K, Detre JA, Mizsei G, Trojanowski JQ, Artacho-Pérula E, de Iñiguez de Onzono Martin MM, M Arroyo-Jiménez M, Muñoz Lopez M, Molina Romero FJ, P Marcos Rabal M, Cebada Sánchez S, Delgado González JC, de la Rosa Prieto C, Córcoles Parada M, Wolk DA, Irwin DJ, Insausti R, Yushkevich PA. Downstream effects of polypathology on neurodegeneration of medial temporal lobe subregions. Acta Neuropathol Commun 2021; 9:128. [PMID: 34289895 PMCID: PMC8293481 DOI: 10.1186/s40478-021-01225-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022] Open
Abstract
The medial temporal lobe (MTL) is a nidus for neurodegenerative pathologies and therefore an important region in which to study polypathology. We investigated associations between neurodegenerative pathologies and the thickness of different MTL subregions measured using high-resolution post-mortem MRI. Tau, TAR DNA-binding protein 43 (TDP-43), amyloid-β and α-synuclein pathology were rated on a scale of 0 (absent)-3 (severe) in the hippocampus and entorhinal cortex (ERC) of 58 individuals with and without neurodegenerative diseases (median age 75.0 years, 60.3% male). Thickness measurements in ERC, Brodmann Area (BA) 35 and 36, parahippocampal cortex, subiculum, cornu ammonis (CA)1 and the stratum radiatum lacunosum moleculare (SRLM) were derived from 0.2 × 0.2 × 0.2 mm3 post-mortem MRI scans of excised MTL specimens from the contralateral hemisphere using a semi-automated approach. Spearman's rank correlations were performed between neurodegenerative pathologies and thickness, correcting for age, sex and hemisphere, including all four proteinopathies in the model. We found significant associations of (1) TDP-43 with thickness in all subregions (r = - 0.27 to r = - 0.46), and (2) tau with BA35 (r = - 0.31) and SRLM thickness (r = - 0.33). In amyloid-β and TDP-43 negative cases, we found strong significant associations of tau with ERC (r = - 0.40), BA35 (r = - 0.55), subiculum (r = - 0.42) and CA1 thickness (r = - 0.47). This unique dataset shows widespread MTL atrophy in relation to TDP-43 pathology and atrophy in regions affected early in Braak stageing and tau pathology. Moreover, the strong association of tau with thickness in early Braak regions in the absence of amyloid-β suggests a role of Primary Age-Related Tauopathy in neurodegeneration.
Collapse
Affiliation(s)
- L E M Wisse
- Department of Diagnostic Radiology, Lund University, Klinikgatan 13b, Lund, Sweden.
- Department of Radiology, University of Pennsylvania, Philadelphia, USA.
| | - S Ravikumar
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - R Ittyerah
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - S Lim
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - J Lane
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - M L Bedard
- Department of Pharmacology, University of North Carolina At Chapel Hill, Chapel Hill, USA
| | - L Xie
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - S R Das
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - T Schuck
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - M Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - E B Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - M D Tisdall
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - K Prabhakaran
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - J A Detre
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - G Mizsei
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - J Q Trojanowski
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - E Artacho-Pérula
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | | | - M M Arroyo-Jiménez
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - M Muñoz Lopez
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - F J Molina Romero
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - M P Marcos Rabal
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - S Cebada Sánchez
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - J C Delgado González
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - C de la Rosa Prieto
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - M Córcoles Parada
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - D A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - D J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - R Insausti
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - P A Yushkevich
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
33
|
Rastegari A, Safavi M, Vafadarnejad F, Najafi Z, Hariri R, Bukhari SNA, Iraji A, Edraki N, Firuzi O, Saeedi M, Mahdavi M, Akbarzadeh T. Synthesis and evaluation of novel arylisoxazoles linked to tacrine moiety: in vitro and in vivo biological activities against Alzheimer's disease. Mol Divers 2021; 26:409-428. [PMID: 34273065 DOI: 10.1007/s11030-021-10248-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/05/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is now ranked as the third leading cause of death after heart disease and cancer. There is no definite cure for AD due to the multi-factorial nature of the disease, hence, multi-target-directed ligands (MTDLs) have attracted lots of attention. In this work, focusing on the efficient cholinesterase inhibitory activity of tacrine, design and synthesis of novel arylisoxazole-tacrine analogues was developed. In vitro acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibition assay confirmed high potency of the title compounds. Among them, compounds 7l and 7b demonstrated high activity toward AChE and BChE with IC50 values of 0.050 and 0.039 μM, respectively. Both compounds showed very good self-induced Aβ aggregation and AChE-induced inhibitory activity (79.4 and 71.4% for compound 7l and 61.8 and 58.6% for compound 7b, respectively). Also, 7l showed good anti-BACE1 activity with IC50 value of 1.65 µM. The metal chelation test indicated the ability of compounds 7l and 7b to chelate biometals (Zn2+, Cu2+, and Fe2+). However, they showed no significant neuroprotectivity against Aβ-induced damage in PC12 cells. Evaluation of in vitro hepatotoxicity revealed comparable toxicity of compounds 7l and 7b with tacrine. In vivo studies by Morris water maze (MWM) task demonstrated that compound 7l significantly reversed scopolamine-induced memory deficit in rats. Finally, molecular docking studies of compounds 7l and 7b confirmed establishment of desired interactions with the AChE, BChE, and BACE1 active sites.
Collapse
Affiliation(s)
- Arezoo Rastegari
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Fahimeh Vafadarnejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Najafi
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roshanak Hariri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Aljouf, 2014, Sakaka, Saudi Arabia
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Central Research laboratory, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Saeedi
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Akbarzadeh
- Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Grothe MJ, Labrador-Espinosa MA, Jesús S, Macías-García D, Adarmes-Gómez A, Carrillo F, Camacho EI, Franco-Rosado P, Lora FR, Martín-Rodríguez JF, Barberá MA, Pastor P, Arroyo SE, Vila BS, Foraster AC, Martínez JR, Padilla FC, Morlans MP, Aramburu IG, Ceberio JI, Vara JH, de Fábregues-Boixar O, de Deus Fonticoba T, Pascual-Sedano B, Kulisevsky J, Martínez-Martín P, Santos-García D, Mir P. In vivo cholinergic basal forebrain degeneration and cognition in Parkinson's disease: Imaging results from the COPPADIS study. Parkinsonism Relat Disord 2021; 88:68-75. [PMID: 34144230 DOI: 10.1016/j.parkreldis.2021.05.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION We aimed to assess associations between multimodal neuroimaging measures of cholinergic basal forebrain (CBF) integrity and cognition in Parkinson's disease (PD) without dementia. METHODS The study included a total of 180 non-demented PD patients and 45 healthy controls, who underwent structural MRI acquisitions and standardized neurocognitive assessment through the PD-Cognitive Rating Scale (PD-CRS) within the multicentric COPPADIS-2015 study. A subset of 73 patients also had Diffusion Tensor Imaging (DTI) acquisitions. Volumetric and microstructural (mean diffusivity, MD) indices of CBF degeneration were automatically extracted using a stereotactic CBF atlas. For comparison, we also assessed multimodal indices of hippocampal degeneration. Associations between imaging measures and cognitive performance were assessed using linear models. RESULTS Compared to controls, CBF volume was not significantly reduced in PD patients as a group. However, across PD patients lower CBF volume was significantly associated with lower global cognition (PD-CRStotal: r = 0.37, p < 0.001), and this association remained significant after controlling for several potential confounding variables (p = 0.004). Analysis of individual item scores showed that this association spanned executive and memory domains. No analogue cognition associations were observed for CBF MD. In covariate-controlled models, hippocampal volume was not associated with cognition in PD, but there was a significant association for hippocampal MD (p = 0.02). CONCLUSIONS Early cognitive deficits in PD without dementia are more closely related to structural MRI measures of CBF degeneration than hippocampal degeneration. In our multicentric imaging acquisitions, DTI-based diffusion measures in the CBF were inferior to standard volumetric assessments for capturing cognition-relevant changes in non-demented PD.
Collapse
Affiliation(s)
- Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel A Labrador-Espinosa
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Macías-García
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Astrid Adarmes-Gómez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Fátima Carrillo
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Elena Iglesias Camacho
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Pablo Franco-Rosado
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Florinda Roldán Lora
- Unidad de Radiodiagnóstico, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Juan Francisco Martín-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Pau Pastor
- Hospital Universitari Mutua de Terrassa, Terrassa, Barcelona, Spain
| | | | - Berta Solano Vila
- Institut Catalá de la Salud (Girona) - Institut d'Assisténcia Sanitaria (IAS), Spain
| | - Anna Cots Foraster
- Institut Catalá de la Salud (Girona) - Institut d'Assisténcia Sanitaria (IAS), Spain
| | - Javier Ruiz Martínez
- Instituto de Investigación Biodonostia, Hospital Universitario Donostia, San Sebastián, Spain
| | | | | | - Isabel González Aramburu
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Jon Infante Ceberio
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Jorge Hernández Vara
- Neurology Department and Neurodegenerative Diseases Research Group. Vall D'Hebron Universitary Campus, Barcelona, Spain
| | - Oriol de Fábregues-Boixar
- Neurology Department and Neurodegenerative Diseases Research Group. Vall D'Hebron Universitary Campus, Barcelona, Spain
| | - Teresa de Deus Fonticoba
- Complejo Hospitalario Universitario de Ferrol (CHUF), Hospital Arquitecto Marcide, Ferrol, Spain
| | - Berta Pascual-Sedano
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital de Sant Pau, Barcelona, Spain; Faculty of Health Sciences, Universitat Oberta de Catalunya (UOC), Barcelona, Spain
| | | | - Jaime Kulisevsky
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital de Sant Pau, Barcelona, Spain
| | - Pablo Martínez-Martín
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
35
|
Khurana K, Kumar M, Bansal N. Lacidipine Prevents Scopolamine-Induced Memory Impairment by Reducing Brain Oxido-nitrosative Stress in Mice. Neurotox Res 2021; 39:1087-1102. [PMID: 33721210 DOI: 10.1007/s12640-021-00346-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/08/2021] [Accepted: 02/28/2021] [Indexed: 02/06/2023]
Abstract
Cholinergic deficits and oxido-nitrosative stress are consistently associated with Alzheimer's disease (AD). Previous findings indicate that acetylcholine subdues Ca2+ current in the brain. Cholinergic antagonists (e.g., scopolamine) can instigate Ca2+-induced redox imbalance, inflammation, and cell-death pathways leading to AD-type memory impairment. Earlier, several Ca2+-channel blockers (CCB, e.g., dihydropyridine type) or cholinergic enhancers showed promising results in animal models of AD. In the present research, pretreatment effects of lacidipine (L-type CCB) on learning and memory functions were investigated using the scopolamine mouse model of AD. Swiss albino mice (20-25 g) were administered lacidipine (1 and 3 mg/kg) for 14 days. Scopolamine, an anti-muscarinic drug, was given (1 mg/kg) from days 8 to 14. The mice were subjected to elevated plus maze (EPM) and passive-avoidance (PA) paradigms. Bay-K8644 (a Ca2+-channel agonist) was administered before behavioral studies on days 13 and 14. Biochemical parameters of oxidative stress and acetylcholinesterase (AChE) activity were quantified using the whole brain. Behavioral studies showed an increase in transfer latency (TL) in the EPM test and a decrease in step-through latency (STL) in the PA test in scopolamine-administered mice. Scopolamine enhanced the AChE activity and oxidative stress in the brain of mice which resulted in memory impairment. Lacidipine prevented the amnesia against scopolamine and reduced the oxidative stress and AChE activity in the brain of mice. Bay-K8644 attenuated the lacidipine-induced improvement in memory and redox balance in scopolamine-administered mice. Lacidipine can prevent the oxidative stress and improve the cholinergic function in the brain. These properties of lacidipine can mitigate the pathogenesis of AD-type dementia.
Collapse
Affiliation(s)
- Kunal Khurana
- I.K. Gujral Punjab Technical University, Kapurthala, Punjab, 144603, India.,Department of Pharmacology, Amar Shaheed Baba Ajeet Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, 140111, India
| | - Manish Kumar
- Department of Pharmacology, Amar Shaheed Baba Ajeet Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, 140111, India.,Chitkara College of Pharmacy, Chitkara University, Punjab, 140111, India
| | - Nitin Bansal
- Department of Pharmacology, Amar Shaheed Baba Ajeet Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, 140111, India. .,Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University (CBLU), Bhiwani, Haryana, 127021, India.
| |
Collapse
|
36
|
Herdick M, Dyrba M, Fritz HCJ, Altenstein S, Ballarini T, Brosseron F, Buerger K, Can Cetindag A, Dechent P, Dobisch L, Duezel E, Ertl-Wagner B, Fliessbach K, Dawn Freiesleben S, Frommann I, Glanz W, Dylan Haynes J, Heneka MT, Janowitz D, Kilimann I, Laske C, Metzger CD, Munk MH, Peters O, Priller J, Roy N, Scheffler K, Schneider A, Spottke A, Jakob Spruth E, Tscheuschler M, Vukovich R, Wiltfang J, Jessen F, Teipel S, Grothe MJ. Multimodal MRI analysis of basal forebrain structure and function across the Alzheimer's disease spectrum. Neuroimage Clin 2020; 28:102495. [PMID: 33395986 PMCID: PMC7689403 DOI: 10.1016/j.nicl.2020.102495] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/19/2020] [Accepted: 11/02/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Dysfunction of the cholinergic basal forebrain (cBF) is associated with cognitive decline in Alzheimer's disease (AD). Multimodal MRI allows for the investigation of cBF changes in-vivo. In this study we assessed alterations in cBF functional connectivity (FC), mean diffusivity (MD), and volume across the spectrum of AD. We further assessed effects of amyloid pathology on these changes. METHODS Participants included healthy controls, and subjects with subjective cognitive decline (SCD), mild cognitive impairment (MCI), or AD dementia (ADD) from the multicenter DELCODE study. Resting-state functional MRI (rs-fMRI) and structural MRI data was available for 477 subjects, and a subset of 243 subjects also had DTI data available. Differences between diagnostic groups were investigated using seed-based FC, volumetric, and MD analyses of functionally defined anterior (a-cBF) and posterior (p-cBF) subdivisions of a cytoarchitectonic cBF region-of-interest. In complementary analyses groups were stratified according to amyloid status based on CSF Aβ42/40 biomarker data, which was available in a subset of participants. RESULTS a-cBF and p-cBF subdivisions showed regional FC profiles that were highly consistent with previously reported patterns, but there were only minimal differences between diagnostic groups. Compared to controls, cBF volumes and MD were significantly different in MCI and ADD but not in SCD. The Aβ42/40 stratified analyses largely matched these results. CONCLUSIONS We reproduced subregion-specific FC profiles of the cBF in a clinical sample spanning the AD spectrum. At least in this multicentric cohort study, cBF-FC did not show marked changes along the AD spectrum, and multimodal MRI did not provide more sensitive measures of AD-related cBF changes compared to volumetry.
Collapse
Affiliation(s)
- Meret Herdick
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Martin Dyrba
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Hans-Christian J Fritz
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Tommaso Ballarini
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE, Munich), Feodor-Lynen-Strasse 17, 81377 Munich, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Arda Can Cetindag
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Psychiatry and Psychotherapy, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Peter Dechent
- MR-Research in Neurology and Psychiatry, Georg-August-University Göttingen, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Emrah Duezel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Birgit Ertl-Wagner
- Institute for Clinical Radiology, Ludwig-Maximilians-University, Marchioninistr. 15, 81377 Munich, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Silka Dawn Freiesleben
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Psychiatry and Psychotherapy, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Ingo Frommann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - John Dylan Haynes
- Bernstein Center for Computational Neuroscience, Charité - Universitätsmedizin, Berlin, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Coraline D Metzger
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany; Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Psychiatry and Psychotherapy, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Department of Neurology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Maike Tscheuschler
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Ruth Vukovich
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, Von-Siebold-Str. 5, 37075 Goettingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany; Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931Köln, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany.
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.
| |
Collapse
|