251
|
Morikawa F, Kobayashi R, Murayama T, Fukuya S, Tabata K, Fujishiro H, Nakayama M, Naoe J. Evaluating Electroconvulsive Therapy for Dementia With Lewy Bodies, Including the Prodromal Stage: A Retrospective Study on Safety and Efficacy. Int J Geriatr Psychiatry 2024; 39:e70020. [PMID: 39608804 DOI: 10.1002/gps.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVES Managing symptoms, notably psychiatric symptoms, in dementia with Lewy bodies (DLB) is complex, affecting both patients and caregivers. People with DLB often react poorly to antipsychotics, limiting treatment options. Although electroconvulsive therapy (ECT)'s potential for DLB is acknowledged, evidence is scarce owing to limited studies. This study investigated ECT's effectiveness and safety for DLB and prodromal DLB with antecedent psychiatric symptoms. METHODS This retrospective study investigated people with DLB (N = 12) and mild cognitive impairment (MCI) with LB (N = 13), a prodromal form of DLB, who underwent ECT for psychiatric symptoms and had abnormal findings confirmed using dopamine transporter single-photon emission computed tomography and 123I-metaiodobenzylguanidine myocardial scintigraphy. We reviewed these patients' medical records and determined the severity of psychotic symptoms before and 1 week after the final ECT session with the Clinical Global Impressions Severity Scale (CGI-S). Improvement in psychotic symptoms was evaluated approximately 1 week after the final ECT session using the CGI Improvement Scale (CGI-I). Additionally, we assessed cognitive function and dementia severity before and after ECT, as well as any adverse events caused by ECT. RESULTS ECT significantly improved psychiatric symptoms, as assessed using the CGI-S, with CGI-I reports in the order of 60% "very much improved," 20% "much improved," 16% "minimally improved," and 4% "no change." Parkinsonism improved (Hoehn and Yahr: 1.76 ± 1.2 before vs. 1.04 ± 0.7 after, p < 0.001) as did dementia severity (Clinical Dementia Rating, p = 0.037). Adverse events included delirium in 24% of patients and amnesia in 4% of patients. ECT did not worsen cognitive function. CONCLUSIONS ECT for DLB and MCI with LB with antecedent psychiatric symptoms appears safe and effective in managing psychiatric symptoms and Parkinsonism. Further large-scale multicenter studies are warranted to conclusively establish its effectiveness and safety.
Collapse
Affiliation(s)
- Fumiyoshi Morikawa
- Department of Psychiatry, Asahikawa Keisenkai Hospital, Asahikawa, Japan
| | - Ryota Kobayashi
- Department of Psychiatry, Yamagata University School of Medicine, Yamagata, Japan
| | - Tomonori Murayama
- Department of Psychiatry, Asahikawa Keisenkai Hospital, Asahikawa, Japan
| | - Shota Fukuya
- Department of Psychiatry, Asahikawa Keisenkai Hospital, Asahikawa, Japan
| | - Kazuki Tabata
- Department of Psychiatry, Asahikawa Keisenkai Hospital, Asahikawa, Japan
| | - Hiroshige Fujishiro
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Juichiro Naoe
- Department of Psychiatry, Asahikawa Keisenkai Hospital, Asahikawa, Japan
| |
Collapse
|
252
|
Shiner T, Kavé G, Mirelman A, Regev K, Piura Y, Goldstein O, Gana Weisz M, Bar‐Shira A, Gurevich T, Orr‐Urtreger A, Alcalay RN, Giladi N, Bregman N. Effect of GBA1 Mutations and APOE Polymorphisms on Survival and Progression Among Ashkenazi Jews with Dementia with Lewy Bodies. Mov Disord 2024; 39:2280-2285. [PMID: 39212252 PMCID: PMC11657010 DOI: 10.1002/mds.30003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/28/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Glucocerebrosidase 1 (GBA1) mutations are associated with reduced survival in Parkinson's disease but their effect on survival in dementia with Lewy bodies (DLB) is unclear. OBJECTIVE To assess the impact of GBA1 mutations on survival among Ashkenazi Jews with DLB, while controlling for APOE status. METHODS One hundred and forty participants from Tel Aviv Medical Center, Israel were genotyped for GBA1 mutations and APOE polymorphisms. Survival rates and follow-up cognitive screening scores were analyzed. RESULTS GBA1 mutation carriers had a two-fold increased risk of death (HR = 1.999), while APOE status did not independently affect survival. In a subset of patients with available clinical data (N = 63), carriers of the APOE ε4 allele showed faster cognitive deterioration, while GBA1 mutation carriers also declined more rapidly albeit not significantly. CONCLUSION Understanding the genetic effects on survival and progression is crucial for patient counseling and inclusion in clinical trials.
Collapse
Affiliation(s)
- Tamara Shiner
- Cognitive Neurology Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medicine and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Gitit Kavé
- Department of Education and PsychologyThe Open UniversityRaananaIsrael
| | - Anat Mirelman
- Faculty of Medicine and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Laboratory for Early Markers of Neurodegeneration (LEMON)Neurological Institute, Tel Aviv Medical CenterTel AvivIsrael
| | - Keren Regev
- Neuroimmunology Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Yoav Piura
- Cognitive Neurology Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Orly Goldstein
- Laboratory of Biomarkers and Genomic of NeurodegenerationTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Mali Gana Weisz
- Laboratory of Biomarkers and Genomic of NeurodegenerationTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Anat Bar‐Shira
- Genetic Laboratory, Genetic InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Tanya Gurevich
- Faculty of Medicine and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Avi Orr‐Urtreger
- Faculty of Medicine and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Laboratory of Biomarkers and Genomic of NeurodegenerationTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Roy N. Alcalay
- Faculty of Medicine and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Laboratory of Biomarkers and Genomic of NeurodegenerationTel Aviv Sourasky Medical CenterTel AvivIsrael
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Nir Giladi
- Faculty of Medicine and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Movement Disorders Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Noa Bregman
- Cognitive Neurology Unit, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medicine and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
253
|
Galvin JE. Lewy Body Dementia. Continuum (Minneap Minn) 2024; 30:1673-1698. [PMID: 39620839 DOI: 10.1212/con.0000000000001496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE Lewy body dementia (LBD) is an umbrella term describing two closely related conditions: Parkinson disease dementia (PDD) and dementia with Lewy bodies (DLB). LBD is the second most common cause of neurodegenerative dementia but is often underrecognized in clinical practice. This review covers the key epidemiologic, clinical, cognitive, behavioral, and biomarker features of LBD and discusses current treatment options. LATEST DEVELOPMENTS Indicative biomarkers of LBD improve the ability to make a diagnosis and include single-photon emission computed tomography (SPECT) of the dopamine system (brain) and the noradrenergic system (cardiac), and polysomnography. α-Synuclein-specific biomarkers in spinal fluid, skin, plasma, and brain imaging are in active development with some available for clinical use. Prodromal stages of PDD and DLB have been contextualized, and diagnostic criteria have been published. An emerging theme is whether an integrated staging system focusing on protein aggregation, rather than clinical symptoms, may advance research efforts. ESSENTIAL POINTS LBD is a common cause of cognitive impairment in older adults but is often subject to significant delays in diagnosis and treatment, increasing the burden on patients and family care partners. Understanding key features of disease and the use of biomarkers will improve recognition. Earlier detection may also facilitate the development of new therapeutics and enrollment in clinical trials.
Collapse
|
254
|
Ma Y, Farris CM, Weber S, Schade S, Nguyen H, Pérez-Soriano A, Giraldo DM, Fernández M, Soto M, Cámara A, Painous C, Muñoz E, Valldeoriola F, Martí MJ, Clarimon J, Kallunki P, Ma TC, Alcalay RN, Gomes BF, Blennow K, Zetterberg H, Constantinescu J, Mengel D, Kadam V, Parchi P, Brockmann K, Tropea TF, Siderowf A, Synofzik M, Kang UJ, Compta Y, Svenningsson P, Mollenhauer B, Concha-Marambio L. Sensitivity and specificity of a seed amplification assay for diagnosis of multiple system atrophy: a multicentre cohort study. Lancet Neurol 2024; 23:1225-1237. [PMID: 39577923 DOI: 10.1016/s1474-4422(24)00395-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/21/2024] [Accepted: 09/17/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND The pathological hallmarks of multiple system atrophy and Parkinson's disease are, respectively, misfolded-α-synuclein-laden glial cytoplasmic inclusions and Lewy bodies. CSF-soluble misfolded α-synuclein aggregates (seeds) are readily detected in people with Parkinson's disease by α-synuclein seed amplification assay (synSAA), but identification of seeds associated with multiple system atrophy for diagnostic purposes has proven elusive. We aimed to assess whether a novel synSAA could reliably distinguish seeds from Lewy bodies and glial cytoplasmic inclusions. METHODS In this multicentre cohort study, a novel synSAA that multiplies and detects seeds by fluorescence was used to analyse masked CSF and brain samples from participants with either clinically diagnosed or pathology-confirmed multiple system atrophy, Parkinson's disease, dementia with Lewy bodies, isolated rapid eye movement sleep behaviour disorder (IRBD), disorders that were not synucleinopathies, or healthy controls. Participants were from eight available cohorts from seven medical centres in four countries: New York Brain Bank, New York, USA (NYBB); University of Pennsylvania, Philadelphia, PA, USA (UPENN); Paracelsus-Elena-Klinik, Kassel, Germany (DeNoPa and KAMSA); Hospital Clinic Barcelona, Spain (BARMSA); Universität Tübingen, Tübingen, Germany (EKUT); Göteborgs Universitet, Göteborgs, Sweden (UGOT); and Karolinska Institutet, Stockholm, Sweden (KIMSA). Clinical cohorts were classified for expected diagnostic accuracy as either research (longitudinal follow-up visits) or real-life (single visit). Sensitivity and specificity were estimated according to pathological (gold standard) and clinical (reference standard) diagnoses. FINDINGS In 23 brain samples (from the NYBB cohort), those containing Lewy bodies were synSAA-positive and produced high fluorescence amplification patterns (defined as type 1); those containing glial cytoplasmic inclusions were synSAA-positive and produced intermediate fluorescence (defined as type 2); and those without α-synuclein pathology produced below-threshold fluorescence and were synSAA-negative. In 21 pathology-confirmed CSF samples (from the UPENN cohort), those with Lewy bodies were synSAA-positive type 1; those with glial cytoplasmic inclusions were synSAA-positive type 2; and those with four-repeat tauopathy were synSAA-negative. In the DeNoPa research cohort (which had no samples from people with multiple system atrophy), the novel synSAA had sensitivities of 95% (95% CI 88-99) for 80 participants with Parkinson's disease and 95% (76-100) for 21 participants with IRBD, and a specificity of 95% (86-99) for 60 healthy controls. Overall (combining BARMSA, EKUT, KAMSA, UGOT, and KIMSA cohorts that were enriched for cases of multiple system atrophy), the novel synSAA had 87% sensitivity for multiple system atrophy (95% CI 80-93) and specificity for type 2 seeds was 77% (67-85). For participants with multiple system atrophy just in research cohorts (BARMSA and EKUT), the novel synSAA had a sensitivity of 84% (95% CI 71-92) and a specificity for type 2 seeds of 87% (74-95), whereas cases from real-life cohorts (KAMSA, KIMSA, and UGOT) had a sensitivity of 91% (95% CI 80-97) but a decreased specificity for type 2 seeds of 68% (53-81). INTERPRETATION The novel synSAA produced amplification patterns that enabled the identification of underlying α-synuclein pathology, showing two levels of fluorescence that corresponded with different pathological hallmarks of synucleinopathy. The synSAA might be useful for early diagnosis of synucleinopathies in clinical trials, and potentially for clinical use, but additional formal validation work is needed. FUNDING Michael J Fox Foundation for Parkinson's Research, Amprion.
Collapse
Affiliation(s)
| | | | - Sandrina Weber
- Department of Neurology, University Medical Center Göttingen, Germany; Paracelsus-Elena-Klinik, Kassel, Germany
| | | | | | - Alexandra Pérez-Soriano
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Darly M Giraldo
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Manel Fernández
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Marta Soto
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Ana Cámara
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Celia Painous
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Esteban Muñoz
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Francesc Valldeoriola
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Maria J Martí
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | | | | | - Thong Chi Ma
- Department of Neuroscience and Physiology, New York University, Grossman School of Medicine, New York, NY, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, NY, USA; Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Göteborgs Universitet, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Göteborgs Universitet, Mölndal, Sweden; Department of Neurology, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Institute of Neurology, and UK Dementia Research Institute, University College London, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | | | - David Mengel
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Vaibhavi Kadam
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany
| | - Piero Parchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Kathrin Brockmann
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Thomas F Tropea
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Siderowf
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Un Jung Kang
- Department of Neuroscience and Physiology, New York University, Grossman School of Medicine, New York, NY, USA
| | - Yaroslau Compta
- Parkinson's Disease & Movement Disorders Unit, Neurology Service, Hospital Cliínic i Universitari de Barcelona, Barcelona, Spain; IDIBAPS, CIBERNED, ERN-RND, Institut Cliínic de Neurociències UBNeuro (Maria de Maeztu Excellence Centre), Universitat de Barcelona, Barcelona, Spain
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Göttingen, Germany; Paracelsus-Elena-Klinik, Kassel, Germany
| | | |
Collapse
|
255
|
Shinagawa S, Hashimoto M, Yamakage H, Toya S, Ikeda M. Eating problems in people with dementia with Lewy bodies: Associations with various symptoms and the physician's understanding. Int Psychogeriatr 2024; 36:1194-1204. [PMID: 38404021 DOI: 10.1017/s1041610224000346] [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: 08/21/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVES Eating problems frequently occur in people with dementia with Lewy bodies (DLB), but few studies have investigated the clinical background of this phenomenon. This study examined the relationship between eating problems and various symptoms of DLB and the relation between the treatment needs for DLB people with eating problems and the understanding of their eating problems by caregivers and physicians. DESIGN, MEASUREMENTS, AND PARTICIPANTS This was a subanalysis of a cross-sectional, questionnaire-based survey study. Two hundred sixty-one subjects with DLB were divided into subjects with or without eating problems. Logistic or linear regression analysis was used to investigate the factors influencing eating problems. The treatment needs of DLB people for their eating problems and the understanding of these needs by caregivers and physicians were calculated as participant-caregiver and participant-physician kappa coefficient. RESULTS Of the 261 participants, 27% suffered from eating problems. The presence of eating problems in participants with DLB was related to depression (p = 0.01, OR : 2.19, 95% CI: 1.23-3.91) and apathy (p = 0.01, OR 2.15, 95% CI: 1.20-3.87), while the worsening of eating problems was related to dysphagia (β = 0.24, p = 0.03), apathy (β = 0.23, p = 0.05), and nighttime behavior (β = 0.24, p = 0.04). The participant-physician kappa coefficient for physician understanding of constipation, weight loss, dysphagia, weight gain, and increase in appetite was significantly lower than the corresponding participant-caregiver kappa coefficient (p-value of five symptoms < 0.01). CONCLUSIONS Physicians need to pay more attention to eating problems and their neuropsychiatric background in the long-term support and management of DLB subjects.
Collapse
Affiliation(s)
| | - Mamoru Hashimoto
- Department of Neuropsychiatry, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hajime Yamakage
- Insight Clinical Development Group, 3H Medi Solution Inc, Tokyo, Japan
| | - Shunji Toya
- Medical Science, Sumitomo Pharma Co., Ltd., Tokyo, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
256
|
Terkelsen MH, Iranzo A, Serradell M, Baun AM, Stokholm MG, Danielsen EH, Østergaard K, Otto M, Svendsen KB, Møller M, Johnsen EL, Garrido A, Vilas D, Santamaria J, Møller A, Gaig C, Brooks DJ, Borghammer P, Tolosa E, Pavese N. Cholinergic dysfunction in isolated rapid eye movement sleep behaviour disorder links to impending phenoconversion. Eur J Neurol 2024; 31:e16503. [PMID: 39360592 PMCID: PMC11554850 DOI: 10.1111/ene.16503] [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: 06/07/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND PURPOSE Most patients with isolated rapid eye movement sleep behaviour disorder (iRBD) progress to a parkinsonian alpha-synucleinopathy. However, time to phenoconversion shows great variation. The aim of this study was to investigate whether cholinergic and dopaminergic dysfunction in iRBD patients was associated with impending phenoconversion. METHODS Twenty-one polysomnography-confirmed iRBD patients underwent baseline 11C-donepezil and 6-Fluoro-(18F)-l-3,4-dihydroxyphenylalanine (18F-DOPA) positron emission tomography (PET). Potential phenoconversion was monitored for up to 8 years. PET images were analysed according to patients' diagnoses after 3 and 8 years using linear regression. Time-to-event analysis was made with Cox regression, dividing patients into low and high tracer uptake groups. RESULTS Follow-up was accomplished in 17 patients. Eight patients progressed to either Parkinson's disease (n = 4) or dementia with Lewy bodies (n = 4), while nine remained non-phenoconverters. Compared with non-phenoconverters, 8-year phenoconverters had lower mean 11C-donepezil uptake in the parietal (p = 0.032) and frontal cortex (p = 0.042), whereas mean 11C-donepezil uptake in 3-year phenoconverters was lower in the parietal cortex (p = 0.005), frontal cortex (p = 0.025), thalamus (p = 0.043) and putamen (p = 0.049). Phenoconverters within 3 years and 8 years had lower 18F-DOPA uptake in the putamen (p < 0.001). iRBD patients with low parietal 11C-donepezil uptake had a 13.46 (95% confidence interval 1.42;127.21) times higher rate of phenoconversion compared with those with higher uptake (p = 0.023). iRBD patients with low 18F-DOPA uptake in the most affected putamen were all phenoconverters with higher rate of phenoconversion (p = 0.0002). CONCLUSIONS These findings suggest that cortical cholinergic dysfunction, particularly within the parietal cortex, could be a biomarker candidate for predicting short-term phenoconversion in iRBD patients. This study aligns with previous reports suggesting dopaminergic dysfunction is associated with forthcoming phenoconversion.
Collapse
Affiliation(s)
- Miriam H. Terkelsen
- Department of Nuclear Medicine and PETInstitute of Clinical Medicine, Aarhus UniversityAarhusDenmark
- Department of NeurologyAarhus University HospitalAarhusDenmark
| | - Alex Iranzo
- Department of NeurologyHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Hospital ClínicSleep Disorders CenterBarcelonaSpain
| | - Mónica Serradell
- Department of NeurologyHospital Clínic de BarcelonaBarcelonaSpain
- Hospital ClínicSleep Disorders CenterBarcelonaSpain
| | - Andreas M. Baun
- Department of Nuclear Medicine and PETInstitute of Clinical Medicine, Aarhus UniversityAarhusDenmark
| | - Morten G. Stokholm
- Department of Nuclear Medicine and PETInstitute of Clinical Medicine, Aarhus UniversityAarhusDenmark
| | | | | | - Marit Otto
- Department of NeurologyAarhus University HospitalAarhusDenmark
- Department of Clinical NeurophysiologyAarhus University HospitalAarhusDenmark
| | | | - Mette Møller
- Department of NeurologyAarhus University HospitalAarhusDenmark
| | - Erik L. Johnsen
- Department of NeurologyAarhus University HospitalAarhusDenmark
| | - Alicia Garrido
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clinic/Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of BarcelonaBarcelonaCataloniaSpain
| | - Dolores Vilas
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clinic/Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of BarcelonaBarcelonaCataloniaSpain
| | - Joan Santamaria
- Department of NeurologyHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Hospital ClínicSleep Disorders CenterBarcelonaSpain
| | - Arne Møller
- Center of Functionally Integrative NeuroscienceAarhus UniversityAarhusDenmark
| | - Carles Gaig
- Department of NeurologyHospital Clínic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Hospital ClínicSleep Disorders CenterBarcelonaSpain
| | - David J. Brooks
- Department of Nuclear Medicine and PETInstitute of Clinical Medicine, Aarhus UniversityAarhusDenmark
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Per Borghammer
- Department of Nuclear Medicine and PETInstitute of Clinical Medicine, Aarhus UniversityAarhusDenmark
| | - Eduardo Tolosa
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Parkinson's Disease and Movement Disorders Unit, Neurology ServiceHospital Clinic/Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of BarcelonaBarcelonaCataloniaSpain
| | - Nicola Pavese
- Department of Nuclear Medicine and PETInstitute of Clinical Medicine, Aarhus UniversityAarhusDenmark
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
257
|
Scholz SW, Cobos I. Genetics and Neuropathology of Neurodegenerative Dementias. Continuum (Minneap Minn) 2024; 30:1801-1822. [PMID: 39620845 DOI: 10.1212/con.0000000000001505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE This article provides an overview of the current understanding of the genetic and pathologic features of neurodegenerative dementias, with an emphasis on Alzheimer disease and related dementias. LATEST DEVELOPMENTS In recent years, there has been substantial progress in genetic research, contributing significant knowledge to our understanding of the molecular risk factors involved in neurodegenerative dementia syndromes. Several genes have been linked to monogenic forms of dementia (eg, APP, PSEN1, PSEN2, SNCA, GRN, C9orf72, MAPT) and an even larger number of genetic variants are known to influence susceptibility for developing dementia. As anti-amyloid therapies for patients with early-stage Alzheimer disease have entered the clinical arena, screening for the apolipoprotein E ε4 high-risk allele has come into focus, emphasizing the importance of genetic counseling. Similarly, advances in the pathologic classifications of neurodegenerative dementia syndromes and molecular pathology highlight their heterogeneity and overlapping features and provide insights into the pathogenesis of these conditions. ESSENTIAL POINTS Recent progress in neurogenetics and molecular pathology has improved our understanding of the complex pathogenetic changes associated with neurodegenerative dementias, facilitating improved disease modeling, enhanced diagnostics, and individualized counseling. The hope is that this knowledge will ultimately pave the way for the development of novel therapeutics.
Collapse
|
258
|
Carrozzo G, Miglis MG, Contin M, Cani I, Cortelli P, Guaraldi P, Calandra-Buonaura G. Intrasubject reproducibility of supine norepinephrine plasma concentrations in patients with cardiovascular sympathetic failure. Auton Neurosci 2024; 256:103216. [PMID: 39260098 DOI: 10.1016/j.autneu.2024.103216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Plasma levels of the catecholamine norepinephrine (NE) has emerged as a useful tool to help differentiate pre- and post-ganglionic disorders in patients with cardiovascular autonomic failure (AF). However, data on intrasubject reliability in individuals with these conditions are limited. We evaluated the intrasubject reproducibility of supine plasma NE levels drawn across two consecutive time points under controlled conditions during head-up table testing in a large cohort of patients with alpha-synucleinopathies and both pre- and post-ganglionic cardiovascular AF. METHODS Antecubital venous blood drawn via an indwelling cannula with the subject supine was assayed for plasma level of catecholamines. We collected two consecutive samples, the first after 20 min of supine rest (NE1) and the second 5 min later (NE2), from a group of 279 participants including 57 with Parkinson's disease/Lewy body dementia (44 M; 65.5 ± 11.1 y), 131 with multiple system atrophy (81 M; 63.2 ± 8.5 y), 41 with pure autonomic failure (25 M, 65.1 ± 9.3 y), and 50 healthy controls (27 M; 46.7 ± 19.4 y). RESULTS We found no difference between NE1 and NE2 (p = 0.645), with a mean intrasubject reproducibility (NE maximum - NE minimum) × 100 / NE maximum) of 11.5 % ± 10.64. This finding was confirmed when controlling for diagnosis (p = 0.669), gender (p = 0.493), age (p = 0.865), disease duration (p = 0.596) or considering all factors together (p = 0.527). CONCLUSIONS We found excellent test-retest reliability of consecutive supine NE measurements in patients with alpha-synucleinopathies and cardiovascular AF, independent of age, gender and disease duration. This lends evidence to support the use of a single supine NE measurement in these conditions.
Collapse
Affiliation(s)
- Giannicola Carrozzo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Mitchell G Miglis
- Department of Neurology and Neurological Sciences, Stanford Medical Center, Palo Alto, CA, USA
| | - Manuela Contin
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Ilaria Cani
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Pietro Cortelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Pietro Guaraldi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.
| | - Giovanna Calandra-Buonaura
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
259
|
Day GS. Diagnosing Alzheimer Disease. Continuum (Minneap Minn) 2024; 30:1584-1613. [PMID: 39620836 DOI: 10.1212/con.0000000000001507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE This article reviews the current understanding of Alzheimer disease (AD), including the natural history, common risk factors, and expected progression of AD neuropathologic change so that neurologists can apply this knowledge to identify patients with symptoms, signs, and findings on common diagnostic tests consistent with AD. LATEST DEVELOPMENTS The advent of potential disease-modifying therapies emphasizes the need to develop and deploy a practical and efficient approach to diagnose patients with cognitive impairment due to AD. ESSENTIAL POINTS The accumulation and spread of cerebral amyloid plaques and tau tangles in patients with AD leads to synaptic dysfunction, neuronal loss, and the eventual emergence and progression of cognitive impairment. A pragmatic and organized approach is needed to recognize patients with symptomatic AD in clinical practice, stage the level of impairment, confirm the clinical diagnosis, and apply this information to advance therapeutic decision making.
Collapse
|
260
|
Revankar GS, Ozono T, Suzuki M, Kanemoto H, Furuya K, Shigenobu K, Yoshiyama K, Yamamoto Y, Ogasawara I, Yoshida N, Iwasaki S, Saeki C, Nishio Y, Nakatani D, Asai K, Kajiyama Y, Shimizu M, Hayashi T, Taniguchi S, Suzuki Y, Inada R, Taminato T, Nagai Y, Hashimoto M, Ikeda M, Mori E, Mochizuki H, Nakata K. Perceptual constancy of pareidolias across paper and digital testing formats in neurodegenerative diseases. Heliyon 2024; 10:e40254. [PMID: 39584082 PMCID: PMC11585718 DOI: 10.1016/j.heliyon.2024.e40254] [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: 02/10/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024] Open
Abstract
Pareidolias refer to visual perceptual deficits where ambiguous shapes take on meaningful appearances. In neurodegenerative diseases, pareidolias are examined via a paper-based neuropsychological tool called the noise pareidolia test. In this study, we present initial findings regarding the utilization of pareidolia test on a digital format to analyze variations between paper-based and digital testing approaches. We performed our experiments on healthy controls, patients diagnosed with Alzheimer's disease (AD), Dementia with Lewy body disease (DLB) and Parkinson's disease (PD). Baseline MMSE assessments were conducted, followed by pareidolia testing using both paper-based tools and smartphones. Bland-Altman analysis was performed to evaluate the agreement between the two methods. We found that the illusionary phenomenon of pareidolia is consistent across paper and digital modalities of testing; that perceptual constancy is maintained across patient groups despite variations in image sizes; and pareidolic misperceptions, to some extent, are stabilized on a digital format. Our findings demonstrate a practical way of testing pareidolias on smartphones without compromising on the functionality of the test.
Collapse
Affiliation(s)
- Gajanan S. Revankar
- Center for Global Health, Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tatsuhiko Ozono
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Maki Suzuki
- Department of Behavioral Neurology and Neuropsychiatry, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Hideki Kanemoto
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kota Furuya
- Center for Global Health, Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Kazue Shigenobu
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Psychiatry, Asakayama General Hospital, Osaka, Japan
| | - Kenji Yoshiyama
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuki Yamamoto
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Psychiatry, Nippon Life Hospital, Osaka, Japan
| | - Issei Ogasawara
- Department of Health and Sport Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Natsuki Yoshida
- Department of Health and Sport Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Susumu Iwasaki
- Department of Health and Sport Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Health and Human Performance, Fort Lewis College, Durango, CO, USA
| | - Chizu Saeki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshiyuki Nishio
- Department of Behavioral Neurology and Neuropsychiatry, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Daisaku Nakatani
- Center for Global Health, Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Kanako Asai
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuta Kajiyama
- Department of Neurology, Kawasaki Medical School, Okayama, Japan
| | - Mikito Shimizu
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tatsuya Hayashi
- Department of Health and Sport Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Seira Taniguchi
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yu Suzuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Rino Inada
- Department of Neurology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Tomoya Taminato
- Department of Neurology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yoshitaka Nagai
- Department of Neurology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Mamoru Hashimoto
- Department of Psychiatry, Kindai University Faculty of Medicine, Osaka, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Psychiatry, Nippon Life Hospital, Osaka, Japan
| | - Etsuro Mori
- Department of Behavioral Neurology and Neuropsychiatry, United Graduate School of Child Development, Osaka University, Osaka, Japan
- Department of Psychiatry, Nippon Life Hospital, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ken Nakata
- Center for Global Health, Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
- Department of Health and Sport Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
261
|
Aljuhani M, Ashraf A, Edison P. Use of Artificial Intelligence in Imaging Dementia. Cells 2024; 13:1965. [PMID: 39682713 PMCID: PMC11640381 DOI: 10.3390/cells13231965] [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: 08/28/2024] [Revised: 11/16/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease is the most common cause of dementia in the elderly population (aged 65 years and over), followed by vascular dementia, Lewy body dementia, and rare types of neurodegenerative diseases, including frontotemporal dementia. There is an unmet need to improve diagnosis and prognosis for patients with dementia, as cycles of misdiagnosis and diagnostic delays are challenging scenarios in neurodegenerative diseases. Neuroimaging is routinely used in clinical practice to support the diagnosis of neurodegenerative diseases. Clinical neuroimaging is amenable to errors owing to varying human judgement as the imaging data are complex and multidimensional. Artificial intelligence algorithms (machine learning and deep learning) enable automation of neuroimaging interpretation and may reduce potential bias and ameliorate clinical decision-making. Graph convolutional network-based frameworks implicitly provide multimodal sparse interpretability to support the detection of Alzheimer's disease and its prodromal stage, mild cognitive impairment. In patients with amyloid-related imaging abnormalities, radiologists had significantly better detection performances with both ARIA-E (sensitivity higher in the assisted/deep learning method [87%] compared to unassisted [71%]) and for ARIA-H signs (sensitivity was higher in assisted [79%] compared to unassisted [69%]). A convolutional neural network method was developed, and external validation predicted final clinical diagnoses of Alzheimer's disease, dementia with Lewy bodies, mild cognitive impairment due to Alzheimer's disease, or cognitively normal with FDG-PET. The translation of artificial intelligence to clinical practice is plagued with technical, disease-related, and institutional challenges. The implementation of artificial intelligence methods in clinical practice has the potential to transform the diagnostic and treatment landscape and improve patient health and outcomes.
Collapse
Affiliation(s)
- Manal Aljuhani
- Radiological Science and Medical Imaging Department, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Azhaar Ashraf
- Division of Neurology, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK (P.E.)
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK (P.E.)
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF24 4HQ, Wales, UK
| |
Collapse
|
262
|
Manabe Y. Clinical Utility and Safety of an Ultrasonic Head Stimulator in Dementia With Lewy Bodies. Alzheimer Dis Assoc Disord 2024; 39:00002093-990000000-00136. [PMID: 39588702 PMCID: PMC11841721 DOI: 10.1097/wad.0000000000000652] [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: 04/11/2024] [Accepted: 10/17/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The potential of Ultra-Ma, an ultrasonic head stimulator, for the supplementary treatment of dementia with Lewy bodies (DLB) was evaluated in patients with various symptoms under poor control by drug therapy. METHODS Patients with DLB treated with choline esterase inhibitor or L-DOPA, either alone or in combination, and who met inclusion criteria were enrolled. Four weeks of placebo stimulation was followed by 8 weeks of active ultrasonic stimulation and a 4-week follow-up. Primary endpoints were the effects of ultrasonic head stimulation on both cognitive dysfunction and behavioral and psychological symptoms of dementia (BPSD). Cognitive dysfunction was evaluated using the Japanese versions of the Mini-Mental State Examination and Montreal Cognitive Assessment, and BPSD was assessed using the Neuropsychiatric Inventory Brief Questionnaire Form. For cognitive fluctuations, the Cognitive Fluctuation Inventory served as an index. Improvements in parkinsonism, activities of daily living, and caregiver burden were examined as secondary endpoints. RESULTS Twelve patients were enrolled. The primary endpoint was significantly improved during the active stimulation period, as were secondary endpoint ratings for parkinsonism and caregiver burden. No notable adverse events occurred. CONCLUSIONS The findings suggest that ultrasonic head stimulation has supplementary potential when combined with drug treatment in DLB.
Collapse
|
263
|
Alexandres CA, McCarter SJ, Tabatabai GM, LeClair-Visonneau L, Feemster JC, Gossard TR, Strainis EP, Jagielski JT, Kelleher MR, Finstuen T, Ali F, Botha H, Graff-Radford J, Olson EJ, Sandness DJ, Morgenthaler TJ, Kantarci K, Savica R, Singer W, Covassin N, Somers VK, Kirkland JL, Junna M, Lipford M, Matarese CA, Moore JL, Tippmann-Peikert M, Carvalho DZ, Boeve BF, Silber MH, St Louis EK. Phenoconversion in Women and Men With Isolated REM Sleep Behavior Disorder: A Retrospective Cohort Study. Neurology 2024; 103:e209993. [PMID: 39454123 PMCID: PMC11515114 DOI: 10.1212/wnl.0000000000209993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 09/16/2024] [Indexed: 10/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Isolated REM sleep behavior disorder (iRBD) is strongly associated with synucleinopathies. Previous iRBD cohort studies have primarily focused on older (>50 years), male-predominant cohorts. Risk of phenoconversion in women and younger adults remains unclear. The study aimed to determine clinical features associated with conversion to a defined neurodegenerative disorder in women and men with iRBD. METHODS One hundred eighty-six women and 186 men with iRBD were matched by polysomnography month. Baseline clinical variables and subsequent neurodegenerative outcomes were abstracted by chart review. Kaplan-Meier curves assessed conversion rates. Cox proportional hazards modeling evaluated factors associated with phenoconversion risk. RESULTS Age at iRBD diagnosis was younger in women compared with men (54.9 vs 62.5 years, p < 0.01). Forty-eight patients (12.9%), including 18 women (9.7%) and 30 men (16.1%), phenoconverted during a median follow-up of 6.0 years. Conversion rates were lower in antidepressant users and patients with chronic pain or psychiatric comorbidity while rates were higher in those with vascular comorbidity. Only age at diagnosis (HR 1.09, 95% CI 1.06-1.13) was associated with phenoconversion after adjusting for RBD symptom duration; sex; antidepressant use; and psychiatric, chronic pain, and vascular comorbidities. DISCUSSION Age at diagnosis was independently associated with phenoconversion risk in women and men with iRBD.
Collapse
Affiliation(s)
- Christina A Alexandres
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Stuart J McCarter
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Grace M Tabatabai
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Laurene LeClair-Visonneau
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - John C Feemster
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Thomas R Gossard
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Emma P Strainis
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Jack T Jagielski
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Makayla R Kelleher
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Thomas Finstuen
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Farwa Ali
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Hugo Botha
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Jonathan Graff-Radford
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Eric J Olson
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - David J Sandness
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Timothy J Morgenthaler
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Kejal Kantarci
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Rodolfo Savica
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Wolfgang Singer
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Naima Covassin
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Virend K Somers
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - James L Kirkland
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Mithri Junna
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Melissa Lipford
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Christine A Matarese
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - James L Moore
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Maja Tippmann-Peikert
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Diego Z Carvalho
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Bradley F Boeve
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Michael H Silber
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| | - Erik K St Louis
- From the Mayo Center for Sleep Medicine (C.A.A., S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., E.J.O., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.), Department of Neurology; Mayo Center for Sleep Medicine (S.J.M., G.M.T., L.L.-V., J.C.F., T.R.G., E.P.S., J.T.J., M.R.K., T.F., D.J.S., T.J.M., M.J., M.L., J.L.M., M.T.-P., D.Z.C., M.H.S., E.K.S.L.),; Department of Neurology (F.A., H.B., J.G.-R., R.S., W.S., B.F.B.); Department of Radiology (K.K.); Department of Cardiovascular Medicine (N.C., V.K.S.); Department of Medicine (J.L.K.); and Mayo Center for Sleep Medicine (C.A.M.), Department of Pediatrics, Mayo Clinic
| |
Collapse
|
264
|
van Gils AM, Tolonen A, van Harten AC, Vigneswaran S, Barkhof F, Visser LNC, Koikkalainen J, Herukka SK, Hasselbalch SG, Mecocci P, Remes AM, Soininen H, Lemstra AW, Teunissen CE, Jönsson L, Lötjönen J, van der Flier WM, Rhodius-Meester HFM. Computerized decision support to optimally funnel patients through the diagnostic pathway for dementia. Alzheimers Res Ther 2024; 16:256. [PMID: 39587679 PMCID: PMC11590510 DOI: 10.1186/s13195-024-01614-5] [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: 03/15/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The increasing prevalence of dementia and the introduction of disease-modifying therapies (DMTs) highlight the need for efficient diagnostic pathways in memory clinics. We present a data-driven approach to efficiently guide stepwise diagnostic testing for three clinical scenarios: 1) syndrome diagnosis, 2) etiological diagnosis, and 3) eligibility for DMT. METHODS We used data from two memory clinic cohorts (ADC, PredictND), including 504 patients with dementia (302 Alzheimer's disease, 107 frontotemporal dementia, 35 vascular dementia, 60 dementia with Lewy bodies), 191 patients with mild cognitive impairment, and 188 cognitively normal controls (CN). Tests included digital cognitive screening (cCOG), neuropsychological and functional assessment (NP), MRI with automated quantification, and CSF biomarkers. Sequential testing followed a predetermined order, guided by diagnostic certainty. Diagnostic certainty was determined using a clinical decision support system (CDSS) that generates a disease state index (DSI, 0-1), indicating the probability of the syndrome diagnosis or underlying etiology. Diagnosis was confirmed if the DSI exceeded a predefined threshold based on sensitivity/specificity cutoffs relevant to each clinical scenario. Diagnostic accuracy and the need for additional testing were assessed at each step. RESULTS Using cCOG as a prescreener for 1) syndrome diagnosis has the potential to accurately reduce the need for extensive NP (42%), resulting in syndrome diagnosis in all patients, with a diagnostic accuracy of 0.71, which was comparable to using NP alone. For 2) etiological diagnosis, stepwise testing resulted in an etiological diagnosis in 80% of patients with a diagnostic accuracy of 0.77, with MRI needed in 77%, and CSF in 37%. When 3) determining DMT eligibility, stepwise testing (100% cCOG, 83% NP, 75% MRI) selected 60% of the patients for confirmatory CSF testing and eventually identified 90% of the potentially eligible patients with AD dementia. CONCLUSIONS Different diagnostic pathways are accurate and efficient depending on the setting. As such, a data-driven tool holds promise for assisting clinicians in selecting tests of added value across different clinical contexts. This becomes especially important with DMT availability, where the need for more efficient diagnostic pathways is crucial to maintain the accessibility and affordability of dementia diagnoses.
Collapse
Affiliation(s)
- Aniek M van Gils
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands.
| | | | - Argonde C van Harten
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
| | - Sinthujah Vigneswaran
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Leonie N C Visser
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
- Department of Medical Psychology, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
- Amsterdam Public Health, Quality of Care, Amsterdam, 1081HV, The Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Centre, University of Copenhagen, Blegdamsvej 9, 2100, RigshospitaletCopenhagen, Denmark
| | - Patrizia Mecocci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06129, Perugia, Italy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, SE, Sweden
| | - Anne M Remes
- Research Unit of Clinical Medicine, Neurology, University of Oulu, 90014, Oulu, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Afina W Lemstra
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
| | - Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | - Wiesje M van der Flier
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
- Department of Epidemiology and Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, the Netherlands
| | - Hanneke F M Rhodius-Meester
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
- Department of Internal Medicine, Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
- Department of Geriatric Medicine, The Memory Clinic, Oslo University Hospital, 0379, Oslo, Norway
| |
Collapse
|
265
|
Lee SS, Civitelli L, Parkkinen L. Brain-derived and in vitro-seeded alpha-synuclein fibrils exhibit distinct biophysical profiles. eLife 2024; 13:RP92775. [PMID: 39584804 PMCID: PMC11588339 DOI: 10.7554/elife.92775] [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: 11/26/2024] Open
Abstract
The alpha-synuclein (αSyn) seeding amplification assay (SAA) that allows the generation of disease-specific in vitro seeded fibrils (SAA fibrils) is used as a research tool to study the connection between the structure of αSyn fibrils, cellular seeding/spreading, and the clinicopathological manifestations of different synucleinopathies. However, structural differences between human brain-derived and SAA αSyn fibrils have been recently highlighted. Here, we characterize the biophysical properties of the human brain-derived αSyn fibrils from the brains of patients with Parkinson's disease with and without dementia (PD, PDD), dementia with Lewy bodies (DLB), multiple system atrophy (MSA), and compare them to the 'model' SAA fibrils. We report that the brain-derived αSyn fibrils show distinct biochemical profiles, which were not replicated in the corresponding SAA fibrils. Furthermore, the brain-derived αSyn fibrils from all synucleinopathies displayed a mixture of 'straight' and 'twisted' microscopic structures. However, the PD, PDD, and DLB SAA fibrils had a 'straight' structure, whereas MSA SAA fibrils showed a 'twisted' structure. Finally, the brain-derived αSyn fibrils from all four synucleinopathies were phosphorylated (S129). Interestingly, phosphorylated αSyn were carried over to the PDD and DLB SAA fibrils. Our findings demonstrate the limitation of the SAA fibrils modeling the brain-derived αSyn fibrils and pay attention to the necessity of deepening the understanding of the SAA fibrillation methodology.
Collapse
Affiliation(s)
- Selene Seoyun Lee
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson’s Disease Center, University of OxfordOxfordUnited Kingdom
| | - Livia Civitelli
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson’s Disease Center, University of OxfordOxfordUnited Kingdom
| | - Laura Parkkinen
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson’s Disease Center, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
266
|
Fischer MHF, Zibrandtsen IC, Johannsen P, Siersma V, Rasmussen JB, Larsen JB, Høgh P. Therapeutic drug monitoring for dose optimization in Alzheimer's disease and in dementia with Lewy bodies: A randomized single-blinded clinical trial. J Alzheimers Dis Rep 2024; 8:1516-1528. [PMID: 40034344 PMCID: PMC11863742 DOI: 10.1177/25424823241289373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 08/31/2024] [Indexed: 03/05/2025] Open
Abstract
Background Previous evidence suggests serum concentrations of donepezil varies in clinical populations and that a dose higher than standard may have additional positive effect on cognition. Therapeutic drug monitoring (TDM) is a tool for dose optimization (DO) whereby treatment is adjusted based on previous quantification of the prescribed drug. Objective Investigate whether TDM-based DO of donepezil or memantine improves clinical outcomes and/or reduce the frequency of adverse reactions (ARs) in neurodegenerative conditions commonly treated with these two study drugs. Methods Single-blinded 1:1 randomized controlled study in an outpatient memory clinic. Eligible participants either newly diagnosed with Alzheimer's disease dementia (AD), dementia with Lewy bodies (DLB), or Parkinson's disease dementia (PDD) scheduled for treatment with donepezil or memantine. The intervention group received TDM based DO. The control group received DO solely based on clinical assessment. Clinical outcomes were change in Mini-Mental State Examination, Addenbrooke's Cognitive Examination, Neuropsychiatric Inventory, and Disability Assessment in Dementia from baseline to 12 months. Additionally, data on incidence and severity of ARs and proportion of participants with a serum concentration within the therapeutic reference range were collected. Results 132 participants recruited (125 AD, 7 DLB, none with PDD) of whom 107 completed the study (101 AD and 6 DLB), fewer in the control group than planned. Statistical analysis did not reveal significant differences between groups neither for clinical outcomes nor for frequency of ARs. Conclusions TDM based DO did not significantly improve clinical outcomes nor reduce the frequency of ARs albeit important caveats to the results apply. ClincialTrialsgov identifier NCT04117178 (first posted October 7, 2019).
Collapse
Affiliation(s)
| | | | | | - Volkert Siersma
- The Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Peter Høgh
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
- Department of Neurology, Herlev Hospital, Herlev, Denmark
| |
Collapse
|
267
|
Wu J, Jin X, Xie W, Liu L, Wang F, Zhu L, Shen Y, Qiu L. Global research trends and hotspots in Parkinson's disease psychosis: a 25-year bibliometric and visual analysis. Front Aging Neurosci 2024; 16:1480234. [PMID: 39649718 PMCID: PMC11621064 DOI: 10.3389/fnagi.2024.1480234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/07/2024] [Indexed: 12/11/2024] Open
Abstract
Background Parkinson's disease psychosis (PDP) is one of the most severe and disabling non-motor symptoms in the progression of Parkinson's disease (PD), significantly impacting the prognosis of PD patients. In recent years, there has been an increase in literature on PDP. However, bibliometrics has rarely been applied to PDP research. This study provides an overview of the current state of PDP research and predicts future trends in this field. Methods The literature search was conducted using the Web of Science Core Collection, with the search terms (Parkinson* AND (psychotic* OR hallucination* OR illusion* OR delusion* OR misperception* OR psychosis OR psychoses)). VOSviewer and CiteSpace software were employed to perform bibliometric analysis and visual representation of the search results. Results A total of 603 articles were effectively included. Since 2017, there has been a significant upward trend in publications related to PDP. The United States, the United Kingdom, and Canada were the top three contributing countries in terms of publication volume, with France also having a strong influence in this field. Movement Disorders and King's College London included and published the most articles on PDP. The paper titled "Hallucinations in Parkinson's Disease: Prevalence, Phenomenology, and Risk Factors" received the highest number of citations and average citations. Cluster analysis results identified brain, prevalence, connectivity, and atypical antipsychotics as key hotspots in this field. High-frequency keywords were grouped into three themes: neurobiology, therapeutic strategies, and symptom research. Among them, pimavanserin, risk, and functional connectivity have been the most studied areas in the past 7 years and are likely to remain key topics in future research. Conclusion Research on PDP has garnered increasing attention. This study visualizes PDP research over the past 25 years to analyze global hotspots and trends. It offers researchers a valuable perspective for identifying key topics and understanding research trajectories in this expanding field.
Collapse
Affiliation(s)
- Jianhong Wu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Xin Jin
- Northern Jiangsu People’s Hospital, Yangzhou, Jiangsu, China
| | - Weiming Xie
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Liang Liu
- Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fei Wang
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Ling Zhu
- Jiangyin People's Hospital, Wuxi, Jiangsu, China
| | - Yuan Shen
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Linghe Qiu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
268
|
Duong MT, Das SR, Khandelwal P, Lyu X, Xie L, McGrew E, Dehghani N, McMillan CT, Lee EB, Shaw LM, Yushkevich PA, Wolk DA, Nasrallah IM. Hypometabolic mismatch with atrophy and tau pathology in mixed Alzheimer and Lewy body disease. Brain 2024:awae352. [PMID: 39573823 DOI: 10.1093/brain/awae352] [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: 04/08/2024] [Revised: 08/27/2024] [Accepted: 11/13/2024] [Indexed: 01/19/2025] Open
Abstract
Polypathology is a major driver of heterogeneity in clinical presentation and extent of neurodegeneration (N) in patients with Alzheimer Disease (AD). Beyond amyloid (A) and tau (T) pathologies, over half of patients with AD have concomitant pathology such as α-synuclein (S) in mixed AD with Lewy Body Disease (LBD). Patients with Mixed Etiology Dementia (MED) such as AD+LBD have faster progression and potentially differential responses to targeted treatments, though the diagnosis of AD+LBD can be challenging given overlapping clinical and imaging features. Development and validation of improved in vivo biomarkers are required to study relationships between N and S and identify imaging patterns reflecting mixed AD+LBD pathologies. We hypothesize that individual proteinopathies, such as T and S, are associated with commensurate levels of N. Thus, we assessed biomarkers of A, T, N and S with positron emission tomography (PET), magnetic resonance imaging (MRI) and cerebrospinal fluid (CSF) seeding amplification assay (SAA) data to determine molecular presentations of mixed A+S+ vs. A+S- cognitively impaired patients from the Alzheimer's Disease Neuroimaging Initiative (ADNI). We found A+S+ patients had parieto-occipital 18F-Fluorodeoxyglucose hypometabolism (a measure of N) disproportionate to the degree of regional atrophy or T burden, highlighting worse hypometabolism associated with S+ SAA. Following up on this hypometabolic mismatch with CSF metabolite and proteome analyses, we found that A+S+ patients exhibited lower CSF levels of dopamine metabolites and synaptic markers like neuronal pentraxin-2 (NPTX2), suggesting that altered neurotransmission and neuron integrity contribute to this dissociation between metabolic PET and MRI. Potential confounders exist when studying relations between N, AD and LBD pathologies, including neuroinflammation and other non-Alzheimer pathologies in MED, though our findings imply posterior hypometabolic mismatch is related more to S than vascular or TDP-43 pathology. A+S+ patients had posterior mismatch with excessive 18F-Fluorodeoxyglucose hypometabolism relative to atrophy or T load, possibly reflecting impaired neuron integrity. Further research must disentangle the impact of multiple proteinopathies and clinicopathologic factors on hypometabolism and atrophy. Cumulatively, patients with mixed AD+LBD etiologies harbor a unique metabolic PET mismatch signature.
Collapse
Affiliation(s)
- Michael Tran Duong
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandhitsu R Das
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pulkit Khandelwal
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xueying Lyu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Long Xie
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily McGrew
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nadia Dehghani
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Corey T McMillan
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward B Lee
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leslie M Shaw
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A Yushkevich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David A Wolk
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ilya M Nasrallah
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
269
|
Kimura T, Fujita K, Sakurai T, Niida S, Ozaki K, Shigemizu D. Whole-genome sequencing to identify rare variants in East Asian patients with dementia with Lewy bodies. NPJ AGING 2024; 10:52. [PMID: 39572598 PMCID: PMC11582613 DOI: 10.1038/s41514-024-00180-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Dementia with Lewy bodies (DLB) is the second most common form of age-related dementia, following Alzheimer's disease (AD). DLB is associated with a worse prognosis than AD and is characterized by a more rapid progression of cognitive impairment and a poorer quality of life. In addition, the pathogenesis of DLB is less understood than that of AD, and only three genes-SNCA (α-synuclein), APOE (apolipoprotein E), and GBA1 (glucosylceramidase beta 1)-have been convincingly demonstrated to be associated with DLB. In this study, we utilized whole-genome sequencing data from 1744 Japanese individuals, comprising 45 DLB patients and 1699 cognitively normal older adults, aiming to identify new genes associated with DLB. Our genome-wide association studies of genes with potentially deleterious mutations identified the CDH23 gene as being associated with DLB, reaching a Bonferroni-corrected significance (P = 7.43 × 10-4). The gene contained three ethnicity-specific heterozygous missense variants (rs181275139, rs563688802, and rs137937502). CDH23 has been linked to deafness syndromes, and DLB patients carrying these mutations displayed symptoms of subjective hearing loss, suggesting a potential association between DLB onset and auditory impairment. Additionally, we explored human leukocyte antigen (HLA) genotypes associated with DLB but found no significant associations. This result suggests that the pathology of DLB differs from that of Parkinson's disease, which has been reported to have an association with HLA. Although a limitation of this study is the lack of replication of our findings, which require further validation in independent cohorts, our study enhances the understanding of the etiology of DLB in the Japanese population and provides new insights into the underlying mechanisms of its pathogenesis.
Collapse
Affiliation(s)
- Tetsuaki Kimura
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kosuke Fujita
- Department of Prevention and Care Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Takashi Sakurai
- Department of Prevention and Care Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Shumpei Niida
- Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.
| |
Collapse
|
270
|
Garnier-Crussard A, Grangé C, Dorey JM, Chapelet G. [Diagnosis and management of delirium in older adults]. Rev Med Interne 2024:S0248-8663(24)01278-5. [PMID: 39578195 DOI: 10.1016/j.revmed.2024.11.005] [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: 08/23/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 11/24/2024]
Abstract
Delirium, also known as acute confusional state, is an acute brain disorder characterized by cognitive disturbances, particularly attentional deficits, potential behavioral changes, and altered vigilance, with a sudden onset and fluctuating course. It is a common condition among hospitalized older patients and has serious consequences for the patient, their family, and the healthcare system. It is considered an "acute brain failure" that often occurs in the context of underlying cognitive and cerebral vulnerability, similar to how acute kidney injury complicates chronic kidney disease or how acute heart failure exacerbates chronic heart failure - usually in the presence of a precipitating medical factor, often infectious, metabolic, perioperative, or neurological. This narrative review aims to describe the symptoms that allow the diagnosis of delirium in older adults, the available diagnostic or screening tools, as well as the complex and bidirectional relationships between delirium and dementia. The management of delirium, including non-pharmacological measures, will be discussed, along with symptomatic pharmacological treatments, which should be reserved for severe cases despite their low level of evidence.
Collapse
Affiliation(s)
- Antoine Garnier-Crussard
- Clinical and Research Memory Center of Lyon, Charpennes Hospital, Department of Geriatric Medicine, Lyon Institute For Aging, Université Claude-Bernard Lyon 1, Hospices Civils de Lyon, Villeurbanne, France; UNICAEN, Inserm, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", NeuroPresage Team, Institut Blood and Brain @ Caen-Normandie, Cyceron, Normandie University, 14000 Caen, France.
| | - Clémence Grangé
- Clinical and Research Memory Center of Lyon, Charpennes Hospital, Department of Geriatric Medicine, Lyon Institute For Aging, Université Claude-Bernard Lyon 1, Hospices Civils de Lyon, Villeurbanne, France
| | - Jean-Michel Dorey
- Clinical and Research Memory Center of Lyon, Charpennes Hospital, Department of Geriatric Medicine, Lyon Institute For Aging, Université Claude-Bernard Lyon 1, Hospices Civils de Lyon, Villeurbanne, France; Department of Aging Psychiatry, Hospital Le Vinatier, Bron, France; Inserm U1028 - CNRS UMR5292 - PsyR2, Lyon Neuroscience Research Center, Neurocampus, Lyon 1 University, Centre Hospitalier Le Vinatier, Bron cedex, France
| | | |
Collapse
|
271
|
Kuang Y, Mao H, Huang X, Chen M, Dai W, Gan T, Wang J, Sun H, Lin H, Liu Q, Yang X, Xu PY. α-Synuclein seeding amplification assays for diagnosing synucleinopathies: an innovative tool in clinical implementation. Transl Neurodegener 2024; 13:56. [PMID: 39574205 PMCID: PMC11580393 DOI: 10.1186/s40035-024-00449-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/25/2024] [Indexed: 11/25/2024] Open
Abstract
The spectrum of synucleinopathies, including Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB), is characterized by α-synuclein (αSyn) pathology, which serves as the definitive diagnostic marker. However, current diagnostic methods primarily rely on motor symptoms that manifest years after the initial neuropathological changes, thereby delaying potential treatment. The symptomatic overlap between PD and MSA further complicates the diagnosis, highlighting the need for precise and differential diagnostic methods for these overlapping neurodegenerative diseases. αSyn misfolding and aggregation occur before clinical symptoms appear, suggesting that detection of pathological αSyn could enable early molecular diagnosis of synucleinopathies. Recent advances in seed amplification assay (SAA) offer a tool for detecting neurodegenerative diseases by identifying αSyn misfolding in fluid and tissue samples, even at preclinical stages. Extensive research has validated the effectiveness and reproducibility of SAAs for diagnosing synucleinopathies, with ongoing efforts focusing on optimizing conditions for detecting pathological αSyn in more accessible samples and identifying specific αSyn species to differentiate between various synucleinopathies. This review offers a thorough overview of SAA technology, exploring its applications for diagnosing synucleinopathies, addressing the current challenges, and outlining future directions for its clinical use.
Collapse
Affiliation(s)
- Yaoyun Kuang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hengxu Mao
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiaoyun Huang
- Houjie Hospital of Dongguan, Dongguan, 523000, China
| | - Minshan Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wei Dai
- Department of Neurology, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, 830054, Xinjiang, China
| | - Tingting Gan
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Jiaqi Wang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hui Sun
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hao Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Qin Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xinling Yang
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| | - Ping-Yi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
272
|
Taomoto D, Nishio Y, Hidaka Y, Kanemoto H, Takahashi S, Ikeda M. Delirium-onset prodromal Lewy body disease: A series of 5 cases. Clin Park Relat Disord 2024; 11:100289. [PMID: 39650052 PMCID: PMC11625343 DOI: 10.1016/j.prdoa.2024.100289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/11/2024] Open
Abstract
Background Delirium-onset prodromal Lewy body disease (LBD) has been proposed as one of the primary phenotypes of prodromal stages of LBD. The detailed clinical features and biomarker profiles of delirium-onset prodromal LBD have not been well characterized. Methods Five consecutive cases of delirium-onset prodromal LBD were documented. The diagnosis of prodromal LBD was made based on neuroimaging biomarkers, including dopamine transporter single-photon emission computed tomography (SPECT), cardiac 123I-metaiodobenzylguanidine scintigraphy, and/or brain perfusion SPECT, as well as clinical findings in the post-delirium follow-up periods. Results In all cases, one or more of the core or supportive clinical features of dementia with Lewy bodies, including rapid eye movement sleep behavior disorder, minor hallucinations, hyposmia, or autonomic dysfunction, were present prior to the onset of delirium. The precipitating factors for delirium were diverse, including surgery, radiation therapy, chemotherapy, and infection. The duration of delirium was prolonged for several months in two cases, whereas it was resolved within a few weeks in the other cases. In most cases, persistent mild cognitive or behavioral symptoms were observed, which were improved with donepezil. Conclusions Our observations suggest that delirium-onset prodromal LBD may represent the later stages of the prodromal LBD rather than its initial stages. It is possible that delirium in the prodromal stages of LBD may represent subthreshold cognitive fluctuations that are transformed into clinically detectable states by a variety of precipitating factors.
Collapse
Affiliation(s)
- Daiki Taomoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshiyuki Nishio
- Department of Behavioral Neurology and Neuropsychiatry, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
| | - Yousuke Hidaka
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hideki Kanemoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | - Shun Takahashi
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Clinical Research and Education Center, Asakayama General Hospital, Sakai 5900018, Osaka, Japan
- Graduate School of Rehabilitation Science, Osaka Metropolitan University, Habikino 5838555, Osaka, Japan
- Department of Neuropsychiatry, Wakayama Medical University, Wakayama 6410012, Wakayama, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
273
|
Ebina J, Mizumura S, Shibukawa M, Morioka H, Nagasawa J, Yanagihashi M, Hirayama T, Ishii N, Kobayashi Y, Inaba A, Orimo S, Kano O. Comparison of MIBG uptake in the major salivary glands between Lewy body disease and progressive supranuclear palsy. Clin Park Relat Disord 2024; 11:100287. [PMID: 39659394 PMCID: PMC11629250 DOI: 10.1016/j.prdoa.2024.100287] [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: 10/03/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Cardiac sympathetic denervation is specific to Lewy body disease (LBD). In Parkinson's disease (PD), sympathetic denervation in the major salivary glands (parotid glands [PG] and submandibular glands [SMG]) has been demonstrated by 123I-metaiodobenzylguanidine (MIBG) scintigraphy. We compared sympathetic denervation in the MSG between PD, dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). Methods We recruited 81 patients with PD, 12 with DLB, 13 with PSP and 25 with control subjects. We evaluated MIBG uptake in the major salivary glands and heart using a quantitative semi-automatic method. We compared MIBG uptake between PD, DLB, and PSP patients and controls, and we evaluated disease sensitivity and specificity. We compared olfactory function with MIBG uptake between PD and PSP patients. Results MIBG uptake in the PG and SMG in the delayed phase was significantly lower in PD and DLB patients than in PSP patients and controls. Conversely, MIBG uptake in the major salivary glands and heart was comparable between PD and DLB. Between LBD and non-LBD, MIBG uptake showed 56-100 % specificity in the PG, while it had 55.6-87.5 % sensitivity in the SMG. Between PD and PSP, MIBG uptake in the PG and SMG had higher disease specificity than olfactory function, while the sensitivity of SMG MIBG uptake was comparable to olfactory function. Conclusion PD and DLB patients showed lower MIBG uptake in the major salivary glands than PSP patients, especially in the delayed phase. MIBG uptake in the major salivary glands may differentiate PD from hyposmic PSP.
Collapse
Affiliation(s)
- Junya Ebina
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Sunao Mizumura
- Department of Radiology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Mari Shibukawa
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Harumi Morioka
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Junpei Nagasawa
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Masaru Yanagihashi
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Takehisa Hirayama
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Nobutomo Ishii
- Central Radiology Division, Department of Radiology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yukio Kobayashi
- Department of Radiological Technology, Kanto Central Hospital, Tokyo, Japan
| | - Akira Inaba
- Department of Neurology, Kanto Central Hospital, Tokyo, Japan
| | | | - Osamu Kano
- Department of Neurology, Toho University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
274
|
Zhang G, Liu S, Xu Y, Ma LY, Zhang W, Ji Y. Elevated plasma total homocysteine levels are associated with behavioral and psychological symptoms in dementia with Lewy bodies. Front Neurosci 2024; 18:1406694. [PMID: 39628654 PMCID: PMC11611840 DOI: 10.3389/fnins.2024.1406694] [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: 03/25/2024] [Accepted: 10/14/2024] [Indexed: 12/06/2024] Open
Abstract
Objective To investigate the association between plasma total homocysteine (tHcy) levels and behavioral and psychological symptoms of dementia (BPSD) in dementia with Lewy bodies (DLB) patients. Methods A total of 82 DLB patients and 134 age-matched healthy controls were included in this study. DLB patients were assessed using the Mini-Mental Status Examination (MMSE), the Clinical Dementia Rating Scale (CDR), and the Neuropsychiatric Inventory (NPI). Plasma tHcy, serum vitamin B12, and folate levels were measured in all study participants. We used Spearman's rank correlation test to analyze the association between tHcy concentrations and NPI scores, MMSE, CDR, and the duration of dementia in DLB patients. Results Clinically significant BPSD was present in 92.7% of DLB patients. The most frequent BPSD were hallucinations (30.4%), apathy (30.4%), and delusions (26.8%). Elevated plasma tHcy levels were significantly associated with total NPI scores in DLB patients, particularly in 10 NPI sub-domains, except for agitation/aggression and disinhibition. No statistically significant association was found between plasma tHcy levels and MMSE, CDR, or dementia duration. Limitations Longitudinal studies with larger sample sizes are required to further explore the relationship between tHcy levels and BPSD in DLB patients as the disease progresses. Conclusion Our study highlighted the high incidence of BPSD and was the first to show that BPSD is associated with elevated plasma tHcy levels in DLB patients in China. These results support the hypothesis that controlling homocysteine levels could offer a new direction for managing BPSD.
Collapse
Affiliation(s)
- Guili Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shuai Liu
- Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
- Department of Neurology, Tianjin Dementia Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Ying Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ling-Yun Ma
- Department of Neurology, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yong Ji
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
- Department of Neurology, Tianjin Dementia Institute, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
275
|
Suzuki Y, Adachi T, Yoshida K, Sakuwa M, Hanajima R. Psychiatric symptoms and TDP-43 pathology in amyotrophic lateral sclerosis. J Neurol Sci 2024; 466:123249. [PMID: 39326369 DOI: 10.1016/j.jns.2024.123249] [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: 06/25/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND ALS is not a pure motor neuron disease but co-occurs with cognitive impairment and psychiatric symptoms. The neuropathological origin of the psychiatric symptoms is unclear. This study examined the association between the psychiatric symptoms and neuropathology of ALS. METHODS We investigated the clinicopathological characteristics of 15 autopsy cases of ALS, including neuronal loss, gliosis, and the burden of TDP-43 pathology. We divided TDP-43-positive structures by morphology into four categories (neuronal cytoplasmic inclusion, dystrophic neurite, dot, and glial cytoplasmic inclusion) and gave each a semiquantitative score in nine brain regions. Braak neurofibrillary tangle stage, Thal amyloid phase, Lewy-related pathology, and argyrophilic grains were also assessed. RESULTS Of the 15 ALS patients, seven had presented with psychiatric symptoms and eight had not. Significantly higher TDP-43 pathology scores were found in the group with psychiatric symptoms in the temporal tip, transentorhinal cortex, entorhinal cortex, subiculum, and the hippocampal CA1 region and dentate gyrus. Cognitive impairment was not significantly associated with the degree of TDP-43 pathology. There were no significant differences in the degree of neuronal loss/gliosis or in other concurrent pathologies between patients with and without psychiatric symptoms. Morphological evaluation showed that neuronal cytoplasmic inclusions, dystrophic neurites, and dots tended to be more common in the group with psychiatric symptoms. CONCLUSION Psychiatric symptoms in ALS may be related to TDP-43 pathology in the perforant pathway. (224 words).
Collapse
Affiliation(s)
- Yuki Suzuki
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tadashi Adachi
- Division of Neuropathology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan.
| | - Kentaro Yoshida
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Mayuko Sakuwa
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
276
|
Marsili L, Colosimo C. The Gunslinger's sign in atypical parkinsonism. J Neurol Sci 2024; 466:123280. [PMID: 39471636 DOI: 10.1016/j.jns.2024.123280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/01/2024]
Affiliation(s)
- Luca Marsili
- Department of Neurology and Rehabilitation Medicine, Gardner Family Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, USA.
| | - Carlo Colosimo
- Department of Neurology, Santa Maria University Hospital, Terni, Italy
| |
Collapse
|
277
|
Giannakis A, Vartholomatos E, Astrakas L, Anyfantis E, Tatsioni A, Argyropoulou M, Konitsiotis S. An SBM and TBSS Analysis in Early-stage Patients With Alzheimer's Disease, Lewy Body Dementias, and Corticobasal Syndrome. J Geriatr Psychiatry Neurol 2024:8919887241302110. [PMID: 39541987 DOI: 10.1177/08919887241302110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
OBJECTIVE To compare gray matter (GM) and white matter (WM) changes in patients with Alzheimer's disease (AD), Lewy body dementias (LBD), corticobasal syndrome (CBS), and healthy controls (HC). METHODS Surface-based morphometry (SBM) was assessed on 3D T1-weighted images using FreeSurfer image analysis and WM microstructure was studied using Tract-Based Spatial Statistics (TBSS) in 12 AD, 15 LBD, 10 CBS patients, and 10 HC. RESULTS Patients with AD, compared with HC, exhibited reduced cortical surface area and volume in the superior frontal, middle frontal, and medial orbitofrontal cortex. In TBSS, AD patients, compared with HC and LBD, displayed decreased fractional anisotropy, axial diffusivity, and increased radial diffusivity in all major WM tracts. Other comparisons between the groups yielded no differences, either in the SBM or the TBSS analysis. CONCLUSIONS The results indicate significant early structural changes in the GM of the frontal lobe, along with WM alterations early in AD patients.
Collapse
Affiliation(s)
- Alexandros Giannakis
- Department of Neurology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Evrysthenis Vartholomatos
- Department of Radiology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Loukas Astrakas
- Department of Medical Physics, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Emmanouil Anyfantis
- Department of Speech and Language Therapy, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Athina Tatsioni
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Maria Argyropoulou
- Department of Radiology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Spiridon Konitsiotis
- Department of Neurology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
278
|
Watanabe H, Duffy JR, Clark H, Machulda MM, Graff-Radford J, Thu Pham NT, Dickson DW, Lowe VJ, Whitwell JL, Josephs KA. Primary Progressive Aphasia Lacking Core Features of Nonfluent and Semantic Variants: Clinical, Neuroimaging, and Neuropathologic Features. Neurology 2024; 103:e209924. [PMID: 39656512 PMCID: PMC11474733 DOI: 10.1212/wnl.0000000000209924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/09/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Evidence has accumulated that the 2011 consensus criteria for primary progressive aphasia (PPA) do not fully capture features of logopenic variant PPA (lvPPA/LPA). We aimed to examine clinical, neuroimaging, and neuropathologic features of PPA lacking features of nonfluent/semantic variants and to provide practical additions to the 2011 consensus criteria. METHODS This was a retrospective examination of data from 2 observational cohort studies where patients with PPA were prospectively recruited at Mayo Clinic. Based on performance on 2 cardinal features (repetition and comprehension), patients were classified as: pure-LPA (poor repetition, acceptable comprehension), Wernicke-like (poor in both), anomic-like (acceptable in both), and transcortical sensory aphasia-like (TCSA-like) (acceptable repetition, poor comprehension). RESULTS The entire cohort consisted of 102 patients with PPA lacking features of nonfluent/semantic variants (median age at onset 63.5 years, 56% female). Thirty-one patients were followed up at 1 year. Twenty-three patients were included in a neuropathologic cohort. The proportion of repetition-preserved PPA (anomic-like and TCSA-like) was more than double that of repetition-impaired PPA (pure-LPA and Wernicke-like) (73% vs 27%). Regarding clinical course, the anomic-like subgroup was a prodromal state of the pure-LPA or TCSA-like subgroup, whereas the pure-LPA and TCSA-like subgroups were a prodromal state of the Wernicke-like subgroup. There was left temporoparietal atrophy on MRI and/or hypometabolism on 18F-fluorodeoxyglucose-PET in all groups. Furthermore, repetition-impaired PPA showed severe hypometabolism in the left superior temporal lobe associated with repetition ability. Regarding pathologic diagnoses, 70% had Alzheimer disease (AD). The pure-LPA, Wernicke-like, and TCSA-like subgroups all showed AD pathology. Only 53% of the anomic-like subgroup had AD. The remaining 47% showed Pick disease (7%), frontotemporal lobar degeneration with TDP-43-immunoreactive pathology (20%), and Lewy body disease (20%). DISCUSSION This observed clinical heterogeneity reflects different time points/severities of the same disease process and hence can be reconceptualized as an AD-related aphasia spectrum, incorporating lvPPA and the 4 subgroups (pure-LPA, Wernicke-like, anomic-like, and TCSA-like). Specifying moderate/severe repetition deficits as a core feature of lvPPA in the 2011 consensus criteria can enhance its pathologic correlations. Recognizing progressive anomic aphasia (anomic-like) as an additional PPA variant could lessen pathologic heterogeneity of lvPPA.
Collapse
Affiliation(s)
- Hiroyuki Watanabe
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Joseph R Duffy
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Heather Clark
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Mary M Machulda
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Jonathan Graff-Radford
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Nha Trang Thu Pham
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Dennis W Dickson
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Val J Lowe
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Jennifer L Whitwell
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Keith A Josephs
- From the Departments of Neurology (H.W., J.R.D., H.C., J.G.-R., K.A.J.), Psychology (M.M.M.), and Radiology (N.T.T.P., V.J.L., J.L.W.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (Neuropathology) (D.W.D.), Mayo Clinic, Jacksonville, FL
| |
Collapse
|
279
|
Huang Y, Cromarty R, Jia L, Han Y, O’Brien J, Taylor JP, Su L. Attention Network Dysfunctions in Lewy Body Dementia and Alzheimer's Disease. J Clin Med 2024; 13:6691. [PMID: 39597835 PMCID: PMC11594996 DOI: 10.3390/jcm13226691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Attention deficits are notable in Lewy body dementia (LBD) and in Alzheimer's disease (AD). In this study, we combined functional magnetic resonance imaging (fMRI) and electroencephalograph (EEG) to detect neural correlates of attention dysfunctions in LBD and AD. Methods: We recruited 33 patients with LBD, 15 patients with AD and 19 elderly healthy controls. The participants performed the modified Attention Network Task (ANT) to investigate the attention dysfunctions. Results: We found that LBD had alerting attention deficits and AD showed apparent orienting attention dysfunctions, while LBD and AD maintained relatively normal executive/conflict attention. Based on source-level EEG analyses, LBD had frontal-central deficits for alerting attention while AD showed inferior frontal and precentral impairments for orienting attention. In addition, the insular and inferior frontal areas were hyper-activated in LBD and AD for executive/conflict attention. Apart from these areas, LBD showed activity in the complementary temporal-central-occipital network for the modified ANT task. Furthermore, the oscillational sources for the ANT effects indicated that the alpha and theta bands were partly impaired in dementia patients. Conclusions: In summary, using source-localised EEG, we found that attention dysfunctions in LBD and AD engaged different neural networks.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Psychiatry, University of Cambridge, Cambridge CB22QQ, UK; (Y.H.)
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Xihu District, Hangzhou 310024, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Xihu District, Hangzhou 310024, China
| | - Ruth Cromarty
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE17RU, UK (J.-P.T.)
| | - Lina Jia
- Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Ying Han
- Beijing Xuanwu Hospital, Capital Medical University, Beijing 100088, China
| | - John O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge CB22QQ, UK; (Y.H.)
| | - John-Paul Taylor
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE17RU, UK (J.-P.T.)
| | - Li Su
- Department of Psychiatry, University of Cambridge, Cambridge CB22QQ, UK; (Y.H.)
- Department of Neuroscience, Neuroscience Institute, Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield S102TN, UK
| |
Collapse
|
280
|
Benussi A, Cantoni V, Rivolta J, Zoppi N, Cotelli MS, Bianchi M, Cotelli M, Borroni B. Alpha tACS Improves Cognition and Modulates Neurotransmission in Dementia with Lewy Bodies. Mov Disord 2024; 39:1993-2003. [PMID: 39136447 DOI: 10.1002/mds.29969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) is characterized by a marked shift of electroencephalographic (EEG) power and dominant rhythm, from the α toward the θ frequency range. Transcranial alternate current stimulation (tACS) is a non-invasive brain stimulation technique that allows entrainment of cerebral oscillations at desired frequencies. OBJECTIVES Our goal is to evaluate the effects of occipital α-tACS on cognitive functions and neurophysiological measures in patients with DLB. METHODS We conducted a double-blind, randomized, sham-controlled, cross-over clinical trial in 14 participants with DLB. Participants were randomized to receive either α-tACS (60 minutes of 3 mA peak-to-peak stimulation at 12 Hz) or sham stimulation applied over the occipital cortex. Clinical evaluations were performed to assess visuospatial and executive functions, as well as verbal episodic memory. Neurophysiological assessments and EEG recordings were conducted at baseline and following both α-tACS and sham stimulations. RESULTS Occipital α-tACS was safe and well-tolerated. We observed a significant enhancement in visuospatial abilities and executive functions, but no improvement in verbal episodic memory. We observed an increase in short latency afferent inhibition, a neurophysiological marker indirectly and partially dependent on cholinergic transmission, coinciding with an increase in α power and a decrease in Δ power following α-tACS stimulation, effects not seen with sham stimulation. CONCLUSIONS This study demonstrates that occipital α-tACS is safe and enhances visuospatial and executive functions in patients with DLB. Improvements in indirect markers of cholinergic transmission and EEG changes indicate significant neurophysiological engagement. These findings justify further exploration of α-tACS as a therapeutic option for DLB patients. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Valentina Cantoni
- Cognitive and Behavioural Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Jasmine Rivolta
- Cognitive and Behavioural Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Nicola Zoppi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Neurology, San Jacopo Hospital, Pistoia, Italy
| | - Maria Sofia Cotelli
- Cognitive and Behavioural Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili di Brescia, Brescia, Italy
- Neurology Unit, Valle Camonica Hospital, Brescia, Italy
| | - Marta Bianchi
- Neurology Unit, Valle Camonica Hospital, Brescia, Italy
| | - Maria Cotelli
- Neuropsychology Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Cognitive and Behavioural Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili di Brescia, Brescia, Italy
| |
Collapse
|
281
|
Kaya D, Yesil Gurel BH, Akpinar Soylemez B, Dost FS, Dokuzlar O, Mutlay F, Ates Bulut E, Petek K, Golimstok AB, Isik AT. Validity and reliability of the Turkish version of the ALBA screening instrument for Lewy body dementia in older adults. APPLIED NEUROPSYCHOLOGY. ADULT 2024; 31:1457-1462. [PMID: 36332080 DOI: 10.1080/23279095.2022.2142793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
ALBA screening instrument (ASI) has been demonstrated to be an effective, cheap, and noninvasive clinical instrument to screen for Lewy body dementia (LBD). We aimed to determine the validity and reliability of the Turkish version of ASI (ASI-T) in patients with LBD and to investigate the discriminative power of the test in patients with Alzheimer's Disease (AD), LBD, and cognitively healthy older adults (controls). 172 older adults over 60 years of age (43 with LBD, 41 AD, and 88 controls) were included. The sensitivity and specificity of the instrument were determined. A significant difference was found in ASI-T total score between people with LBD versus the controls (t=-9.259; p < 0.001), and versus patients with AD (t = 3.490; p = 0.001). Internal consistency of the ASI-T was good(Cronbach's alpha = 0.81). The cutoff score of 7 showed sensitivity (86%) and specificity (81%) (AUC= 0.888,CI0.95, p < 0.001) compared to controls. Also, compared to AD, it showed sensitivity (86%) and specificity(70%) (AUC = 0.590,CI .95, p < 0.001). Moreover, ASI-T demonstrated a significant concurrent validity with MMSE (r = -0.62; p < 0.001) and MoCA (r = -0.54; p = 0.003). In factor analysis, the five subscales accounted for 60% of the total variance. Our findings suggested that the ASI-T is a reliable, valid, and effective instrument for screening LBD. With acceptable psychometric properties, it has the power to distinguish patients with LBD from controls or those with AD.
Collapse
Affiliation(s)
- Derya Kaya
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
- The Geriatric Science Association, Izmir, Turkey
| | | | - Burcu Akpinar Soylemez
- Department of Internal Medicine Nursing, Faculty of Nursing, Dokuz Eylul University, Izmir, Turkey
| | - Fatma Sena Dost
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
- The Geriatric Science Association, Izmir, Turkey
| | | | - Feyza Mutlay
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
- The Geriatric Science Association, Izmir, Turkey
| | - Esra Ates Bulut
- The Geriatric Science Association, Izmir, Turkey
- Department of Geriatric Medicine, Adana City Research and Training Hospital, Adana, Turkey
| | - Kadriye Petek
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Angel Bernardo Golimstok
- Neurology Department of Italian Hospital of Buenos Aires, Buenos Aires, Argentina
- Lewy Body Association Argentina (ALBA), Buenos Aires, Argentina
| | - Ahmet Turan Isik
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
- The Geriatric Science Association, Izmir, Turkey
| |
Collapse
|
282
|
Goodheart AE, Gomperts SN. The association between cigarette smoking and dementia with Lewy bodies. Parkinsonism Relat Disord 2024; 128:107133. [PMID: 39276722 DOI: 10.1016/j.parkreldis.2024.107133] [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: 04/22/2024] [Revised: 08/03/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION Cigarette smoking is associated with a reduced risk of Parkinson's disease (PD). As dementia with Lewy bodies (DLB) and PD share core neuropathologic features, we set out to examine the relationship between smoking and DLB. METHODS Diagnosis at baseline visit and smoking history of participants ≥50 years old in the National Alzheimer's Coordinating Center (NACC) cohort were evaluated in this cross-sectional study. Odds of diagnosis of cognitive impairment due to DLB, PD, vascular dementia (VD), or Alzheimer's disease (AD) compared to normal cognition based on smoking status and duration were determined using logistic regression. RESULTS 37,478 participants were included (mean age 72 years (SD 9), 57 % female). The odds of DLB were reduced in all smoking status groups compared to never smokers (OR (95 % CI)): ever smokers 0.850 (0.745-0.971), former smokers 0.871 (0.761-0.997), current smokers 0.640 (0.419-0.947)) and in all smoking duration groups. As expected, the odds of PD were reduced in all smoking groups and fell with longer smoking duration. The odds of VD were increased in the current smoking group and rose with greater smoking duration. The odds of AD were unchanged in current smokers, decreased in ever and former smokers, and increased with longer cumulative smoking duration. CONCLUSIONS Cigarette smoking is associated with lower odds of diagnosis of DLB at baseline visit in the NACC cohort. In the context of the well-established reduced risk of PD among smokers, this preliminary evidence of a potential protective effect of smoking on DLB warrants further study.
Collapse
Affiliation(s)
- Anna E Goodheart
- Massachusetts General Hospital, Department of Neurology, Charlestown, MA, 02129, USA; Harvard Medical School, USA.
| | - Stephen N Gomperts
- Massachusetts General Hospital, Department of Neurology, Charlestown, MA, 02129, USA; Harvard Medical School, USA.
| |
Collapse
|
283
|
Liampas I, Siokas V, Zoupa E, Kyriakoulopoulou P, Stamati P, Provatas A, Tsouris Z, Tsimourtou V, Lyketsos CG, Dardiotis E. Neuropsychiatric symptoms and white matter hyperintensities in older adults without dementia. Int Psychogeriatr 2024; 36:1051-1063. [PMID: 38639110 PMCID: PMC11489321 DOI: 10.1017/s1041610224000607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
OBJECTIVE We aimed to examine associations between neuropsychiatric symptoms (NPS) and white matter hyperintensities (WMH) status in older adults without dementia under the hypothesis that WMH increased the odds of having NPS. DESIGN Longitudinal analysis of data acquired from the National Alzheimer's Coordinating Center Uniform Data Set. SETTINGS Data were derived from 46 National Institute on Aging - funded Alzheimer's Disease Research Centers. PARTICIPANTS NACC participants aged ≥50 years with available data on WMH severity with a diagnosis of mild cognitive impairment (MCI) or who were cognitively unimpaired (CU) were studied. Among 4617 CU participants, 376 had moderate and 54 extensive WMH. Among 3170 participants with MCI, 471 had moderate and 88 had extensive WMH. MEASUREMENTS Using Cardiovascular Health Study (CHS) scores, WMH were coded as no to mild (CHS score: 0-4), moderate (score: 5-6) or extensive (score: 7-8). NPS were quantified on the Neuropsychiatric Inventory Questionnaire. Binary logistic regression models estimated the odds of reporting each of 12 NPS by WMH status separately for individuals with MCI or who were CU. RESULTS Compared to CU individuals with no to mild WMH, the odds of having elation [9.87, (2.63-37.10)], disinhibition [4.42, (1.28-15.32)], agitation [3.51, (1.29-9.54)] or anxiety [2.74, (1.28-5.88)] were higher for the extensive WMH group, whereas the odds of having disinhibition were higher for the moderate WMH group [1.94, (1.05-3.61)]. In the MCI group, he odds of NPS did not vary by WMH status. CONCLUSIONS Extensive WMH were associated with higher odds of NPS in CU older adults but not in those with MCI.
Collapse
Affiliation(s)
- Ioannis Liampas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa 41100, Greece
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa 41100, Greece
| | - Elli Zoupa
- Larisa Day Care Center of People with Alzheimer’s Disease, Association for Regional Development and Mental Health (EPAPSY), 15124 Marousi, Greece
| | | | - Polyxeni Stamati
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa 41100, Greece
| | - Antonios Provatas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa 41100, Greece
| | - Zisis Tsouris
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa 41100, Greece
| | - Vana Tsimourtou
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa 41100, Greece
| | - Constantine G. Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa 41100, Greece
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
284
|
Stewart PV, Tapscott BE, Davis B, Boscarino JJ, Sanders K, Rodgers SE, Lichtenstein ML. Validation and extension of the quick dementia rating system (QDRS). APPLIED NEUROPSYCHOLOGY. ADULT 2024; 31:1375-1382. [PMID: 36240388 DOI: 10.1080/23279095.2022.2129056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Informant report dementia severity staging measures, such as the Quick Dementia Rating System (QDRS) offer clinicians useful diagnostic and staging information. These measures also potentially avoid many of the pitfalls inherent in mental status examinations (e.g., cultural bias, educational bias, floor and ceiling effects). We derive cut points for the QDRS and comprehensively examine their classification accuracy in a large, diagnostically heterogeneous, rural, memory disorder clinic sample. Our findings suggest the QDRS may be helpful when used in the context of a comprehensive diagnostic and staging evaluation. When used in isolation, the QDRS is insufficiently accurate for diagnosis and staging of dementia.
Collapse
Affiliation(s)
| | - Brian E Tapscott
- Department of Psychiatry and Behavioral Sciences, Cleveland Clinic Akron General, Akron, OH, USA
| | - Beate Davis
- Department of Psychiatry, Geisinger, Danville, PA, USA
| | - Joseph J Boscarino
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | | | | | - Maya L Lichtenstein
- Department of Neurology, Memory and Cognition Program, Geisinger, Wilkes-Barre, PA, USA
| |
Collapse
|
285
|
Finsterer J. Workup of Parkinson's Disease, Parkinson Plus, Lewy Body, and Alzheimer's Dementia Should Include SPECT, PET and Genetics. Health Sci Rep 2024; 7:e70197. [PMID: 39512254 PMCID: PMC11541238 DOI: 10.1002/hsr2.70197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/18/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Affiliation(s)
- Josef Finsterer
- Neurology DepartmentNeurology & Neurophysiology CenterViennaAustria
| |
Collapse
|
286
|
Quaresima V, Pilotto A, Trasciatti C, Tolassi C, Parigi M, Bertoli D, Mordenti C, Galli A, Rizzardi A, Caratozzolo S, Benussi A, Ashton NJ, Blennow K, Zetterberg H, Giliani S, Brugnoni D, Padovani A. Plasma p-tau181 and amyloid markers in Alzheimer's disease: A comparison between Lumipulse and SIMOA. Neurobiol Aging 2024; 143:30-40. [PMID: 39208716 DOI: 10.1016/j.neurobiolaging.2024.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Aim of the project was to evaluate the technical and clinical validity of plasma Lumipulse p-tau, Aβ42 and Aβ40 species and their correlation with CSF core Alzheimer's Disease (AD) markers; a method comparison with SIMOA was also performed. One-hundred-thirthy-three participants, namely 55 A+T+N+ AD, 28 Neurodegenerative disorders (NDD) and 50 controls were enrolled for the study. Lumipulse technical validity showed high stability for p-tau181, Aβ42, and Aβ40, with higher stability of p-tau to repeated freezing thaw cycles. p-tau181 levels detected by both techniques were higher in AD compared to both NDD/controls and exhibited a similar correlation with CSF p-tau levels, whereas Aβ42 levels were slightly lower in AD with both methods. In the comparison between SIMOA and Lumipulse plasma markers, both techniques exhibited similar diagnostic accuracy for AD for p-tau181 (0.87; 95 %CI 0.81-0.94, vs 0.85; 95 %CI 0.78-0.93), whereas the best performance was reached by p-tau181/ Aβ42 Lumipulse ratio (ROC AUC 0.915, 95 %CI 0.86-0.97). The study thus confirmed the construct validity of both Lumipulse and SIMOA techniques for the identification of CSF AD pattern in clinical settings.
Collapse
Affiliation(s)
- Virginia Quaresima
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy; Residency Program in Clinical Pathology and Clinical Biochemistry, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy; A. Nocivelli Institute for Molecular Medicine Spedali Civili Hospital and Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy; Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy.
| | - Chiara Trasciatti
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy; Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
| | - Chiara Tolassi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy; Residency Program in Clinical Pathology and Clinical Biochemistry, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy; A. Nocivelli Institute for Molecular Medicine Spedali Civili Hospital and Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marta Parigi
- A. Nocivelli Institute for Molecular Medicine Spedali Civili Hospital and Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Diego Bertoli
- Central Clinical Laboratory, ASST Spedali Civili Hospital, Brescia, Italy
| | - Cristina Mordenti
- Central Clinical Laboratory, ASST Spedali Civili Hospital, Brescia, Italy
| | - Alice Galli
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy; Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy
| | - Andrea Rizzardi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy
| | - Salvatore Caratozzolo
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy; Neurology Clinic, Trieste University Hospital, Trieste, Italy
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway; King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK; Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK; Department of Old Age Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Silvia Giliani
- A. Nocivelli Institute for Molecular Medicine Spedali Civili Hospital and Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Duilio Brugnoni
- Central Clinical Laboratory, ASST Spedali Civili Hospital, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili Hospital, Brescia, Italy; Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy; Brain Health Center, University of Brescia, Brescia, Italy
| |
Collapse
|
287
|
Pilotto A, Galli A, Zatti C, Placidi F, Izzi F, Premi E, Caminiti SP, Presotto L, Rizzardi A, Catania M, Lupini A, Purin L, Pasolini MP, Mercuri NB, Chiaravalotti A, Fernandes M, Calvello C, Lucchini S, Bertagna F, Paghera B, Perani D, Berg D, Padovani A, Liguori C. Insular monoaminergic deficits in prodromal α-synucleinopathies. Ann Clin Transl Neurol 2024; 11:2836-2845. [PMID: 39444171 PMCID: PMC11572750 DOI: 10.1002/acn3.52151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 10/25/2024] Open
Abstract
METHODS This study assessed data from two cohorts of patients with alpha-synucleinopathies (University of Brescia and University of Rome Tor-Vergata cohorts). Consecutive participants with video-polysomnography-confirmed iRBD, Parkinson's disease (PD), dementia with Lewy bodies (DLB) and controls underwent neurological, clinical and 123I-FP-CIT SPECT imaging assessments. Individuals with iRBD were longitudinally monitored to collect clinical phenoconversion to PD or DLB. The main outcome was to identify whole brain 123 I-FP-CIT SPECT measures reflecting monoaminergic deficits in each clinical group as compared to controls. RESULTS The cohort (n = 184) included 45 patients with iRBD, 47 PD, 42 DLB and 50 age-matched controls. Individuals with iRBD were categorized as RBD-DAT- (n = 32) and RBD-DAT+ (n = 13), according to nigrostriatal assessment used in clinical practice. Compared to controls, RBD-DAT- showed an early involvement of the left insula, which increased in RBD-DAT+, and was present in patients with Parkinson's disease and dementia with Lewy bodies. Longitudinal cox regression analyses revealed a higher risk of phenoconversion in individuals with iRBD and insular monoaminergic deficits [HR = 3.387; CI 95%: 1.18-10.27]. INTERPRETATION In this study, altered insular monoaminergic binding in iRBD was associated with phenoconversion to DLB or PD. These findings may provide a helpful stratification approach for future pharmacological or non-pharmacological interventions.
Collapse
Affiliation(s)
- Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
- Department of Continuity of Care and Frailty, Neurology UnitASST Spedali Civili of BresciaBrescia25123Italy
- Laboratory of Digital Neurology and BiosensorsUniversity of BresciaBrescia25123Italy
- Neurobiorepository and Laboratory of Advanced Biological MarkersUniversity of Brescia and ASST Spedali Civili Hospital25123BresciaItaly
| | - Alice Galli
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
- Laboratory of Digital Neurology and BiosensorsUniversity of BresciaBrescia25123Italy
| | - Cinzia Zatti
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
- Department of Continuity of Care and Frailty, Neurology UnitASST Spedali Civili of BresciaBrescia25123Italy
- Laboratory of Digital Neurology and BiosensorsUniversity of BresciaBrescia25123Italy
| | - Fabio Placidi
- Sleep Medicine Centre, Neurology UnitUniversity Hospital of Rome Tor Vergata00133Italy
- Department of Systems MedicineUniversity of Rome Tor VergataRome00133Italy
| | - Francesca Izzi
- Sleep Medicine Centre, Neurology UnitUniversity Hospital of Rome Tor Vergata00133Italy
| | - Enrico Premi
- Vascular NeurologyASST Spedali Civili of BresciaBrescia25123Italy
| | - Silvia P. Caminiti
- Department of Brain and Behavioral SciencesUniveristy of PaviaPavia27100Italy
| | - Luca Presotto
- Department of Physics “G. Occhialini”University of Milano‐BicoccaMilan20126Italy
| | - Andrea Rizzardi
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
- Department of Continuity of Care and Frailty, Neurology UnitASST Spedali Civili of BresciaBrescia25123Italy
- Laboratory of Digital Neurology and BiosensorsUniversity of BresciaBrescia25123Italy
| | - Marcello Catania
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
| | - Alessandro Lupini
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
| | - Leandro Purin
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
| | - Maria P. Pasolini
- Sleep Disorder CenterASST Spedali Civili of BresciaBrescia25123Italy
| | - Nicola B. Mercuri
- Sleep Medicine Centre, Neurology UnitUniversity Hospital of Rome Tor Vergata00133Italy
- Department of Systems MedicineUniversity of Rome Tor VergataRome00133Italy
| | | | - Mariana Fernandes
- Department of Systems MedicineUniversity of Rome Tor VergataRome00133Italy
| | - Carmen Calvello
- Department of Systems MedicineUniversity of Rome Tor VergataRome00133Italy
| | | | | | | | | | - Daniela Berg
- Department of NeurologyChristian‐Albrechts‐University of KielKiel24098Germany
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental SciencesUniversity of BresciaBrescia25123Italy
- Department of Continuity of Care and Frailty, Neurology UnitASST Spedali Civili of BresciaBrescia25123Italy
- Laboratory of Digital Neurology and BiosensorsUniversity of BresciaBrescia25123Italy
- Neurobiorepository and Laboratory of Advanced Biological MarkersUniversity of Brescia and ASST Spedali Civili Hospital25123BresciaItaly
- Brain Health CenterUniversity of Brescia25123BresciaItaly
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology UnitUniversity Hospital of Rome Tor Vergata00133Italy
- Department of Systems MedicineUniversity of Rome Tor VergataRome00133Italy
| |
Collapse
|
288
|
Nagarajan A, Laird J, Ugochukwu O, Reppe S, Gautvik K, Ross RD, Bennett DA, Rosen C, Kiel DP, Higginbotham LA, Seyfried NT, Lary CW. Network Analysis of Brain and Bone Tissue Transcripts Reveals Shared Molecular Mechanisms Underlying Alzheimer's Disease and Related Dementias and Osteoporosis. J Gerontol A Biol Sci Med Sci 2024; 79:glae211. [PMID: 39194133 PMCID: PMC11503475 DOI: 10.1093/gerona/glae211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Alzheimer's disease and related dementias (ADRD) and osteoporosis (OP) are 2 prevalent diseases of aging with demonstrated epidemiological association, but the underlying molecular mechanisms contributing to this association are unknown. METHODS We used network analysis of bone and brain transcriptomes to discover common molecular mechanisms underlying these 2 diseases. Our study included RNA-sequencing data from the dorsolateral prefrontal cortex tissue of autopsied brains in 629 participants from ROSMAP (Religious Orders Study and the Rush Memory and Aging Project), with a subgroup of 298 meeting criteria for inclusion in 5 ADRD categories, and RNA array data from transiliac bone biopsies in 84 participants from the Oslo study of postmenopausal women. After developing each network within each tissue, we analyzed associations between modules (groups of coexpressed genes) with multiple bone and neurological traits, examined overlap in modules between networks, and performed pathway enrichment analysis to discover conserved mechanisms. RESULTS We discovered 3 modules in ROSMAP that showed significant associations with ADRD and bone-related traits and 4 modules in Oslo that showed significant associations with multiple bone outcomes. We found significant module overlap between the 2 networks in modules linked to signaling, tissue homeostasis, and development, and Wingless-related integration site (Wnt) signaling was found to be highly enriched in OP and ADRD modules of interest. CONCLUSIONS These results provide translational opportunities in the development of treatments and biomarkers for ADRD and OP.
Collapse
Affiliation(s)
- Archana Nagarajan
- Roux Institute, Northeastern University, Portland, Maine, USA
- Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Jason Laird
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Obiadada Ugochukwu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sjur Reppe
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Kaare Gautvik
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Ryan D Ross
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Clifford Rosen
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | - Douglas P Kiel
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Lenora A Higginbotham
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christine W Lary
- Roux Institute, Northeastern University, Portland, Maine, USA
- MaineHealth Institute for Research, Scarborough, Maine, USA
| |
Collapse
|
289
|
Murayama N, Ota K, Iseki E. The Bender Gestalt Test is useful for clinically diagnosing dementia with Lewy bodies: Analysis of its sensitivity, specificity, and clinical characteristics of the figure copy. APPLIED NEUROPSYCHOLOGY. ADULT 2024; 31:1296-1301. [PMID: 36121098 DOI: 10.1080/23279095.2022.2122059] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A previous study that evaluated the ability of the Bender Gestalt Test (BGT) to discriminate between dementia with Lewy bodies (DLB) and Alzheimer's disease (AD) suggested that a total score of 98 is the optimal cutoff value for discriminating between these two diseases and that DLB tends to exhibit unique errors; i.e., "element deformation" and "gestalt destruction." The objectives of the present study were: (1) to examine the sensitivity and specificity of a total BGT score of 98 as a cutoff value in greater numbers of DLB patients than in the previous study, (2) to set a new cutoff value if a cutoff value of 98 is not optimal, and (3) to clarify the frequency of element deformation and gestalt destruction in DLB patients. The participants were 133 DLB patients, 65 AD patients, and 30 cognitively normal elderly people. All of the participants underwent the Mini-Mental State Examination, BGT, and brain magnetic resonance imaging. As a result, the total BGT score cutoff value of 98 showed low sensitivity (0.58), and a cutoff value of 84 was indicated to be the optimal cutoff value for discriminating between DLB and AD. In addition, 32 out of 133 DLB patients and one out of 65 AD patients exhibited element deformation or gestalt destruction. This study suggested that the BGT is a useful neuropsychological test for differentiating DLB from AD. In addition, the need to evaluate the spatial and perceptual difficulties of DLB patients with various types of visual stimulation is also discussed.
Collapse
Affiliation(s)
- Norio Murayama
- Faculty of Humanities and Social Sciences, Showa Women's University, Tokyo, Japan
| | - Kazumi Ota
- Senior Mental Clinic Nihonbashi-Ningyocho, Tokyo, Japan
| | - Eizo Iseki
- Senior Mental Clinic Nihonbashi-Ningyocho, Tokyo, Japan
| |
Collapse
|
290
|
Jaramillo-Jimenez A, Tovar-Rios DA, Mantilla-Ramos YJ, Ochoa-Gomez JF, Bonanni L, Brønnick K. ComBat models for harmonization of resting-state EEG features in multisite studies. Clin Neurophysiol 2024; 167:241-253. [PMID: 39369552 DOI: 10.1016/j.clinph.2024.09.019] [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: 08/29/2023] [Revised: 07/23/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE Pooling multisite resting-state electroencephalography (rsEEG) datasets may introduce bias due to batch effects (i.e., cross-site differences in the rsEEG related to scanner/sample characteristics). The Combining Batches (ComBat) models, introduced for microarray expression and adapted for neuroimaging, can control for batch effects while preserving the variability of biological covariates. We aim to evaluate four ComBat harmonization methods in a pooled sample from five independent rsEEG datasets of young and old adults. METHODS RsEEG signals (n = 374) were automatically preprocessed. Oscillatory and aperiodic rsEEG features were extracted in sensor space. Features were harmonized using neuroCombat (standard ComBat used in neuroimaging), neuroHarmonize (variant with nonlinear adjustment of covariates), OPNested-GMM (variant based on Gaussian Mixture Models to fit bimodal feature distributions), and HarmonizR (variant based on resampling to handle missing feature values). Relationships between rsEEG features and age were explored before and after harmonizing batch effects. RESULTS Batch effects were identified in rsEEG features. All ComBat methods reduced batch effects and features' dispersion; HarmonizR and OPNested-GMM ComBat achieved the greatest performance. Harmonized Beta power, individual Alpha peak frequency, Aperiodic exponent, and offset in posterior electrodes showed significant relations with age. All ComBat models maintained the direction of observed relationships while increasing the effect size. CONCLUSIONS ComBat models, particularly HarmonizeR and OPNested-GMM ComBat, effectively control for batch effects in rsEEG spectral features. SIGNIFICANCE This workflow can be used in multisite studies to harmonize batch effects in sensor-space rsEEG spectral features while preserving biological associations.
Collapse
Affiliation(s)
- Alberto Jaramillo-Jimenez
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norway; Faculty of Health Sciences, University of Stavanger, Stavanger, Norway; Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia; Grupo Neuropsicología y Conducta, Universidad de Antioquia. Medellín, Colombia; Semillero de Investigación NeuroCo, Universidad de Antioquia, Medellín, Colombia.
| | - Diego A Tovar-Rios
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norway; Doctoral School Biomedical Sciences, KU Leuven, Leuven, Belgium; Grupo de Investigación en Estadística Aplicada - INFERIR, Universidad del Valle, Cali, Colombia; Prevención y Control de la Enfermedad Crónica - PRECEC, Universidad del Valle, Colombia.
| | - Yorguin-Jose Mantilla-Ramos
- Grupo Neuropsicología y Conducta, Universidad de Antioquia. Medellín, Colombia; Semillero de Investigación NeuroCo, Universidad de Antioquia, Medellín, Colombia; Cognitive and Computational Neuroscience Laboratory (CoCo Lab), University of Montreal, Montreal, Canada.
| | - John-Fredy Ochoa-Gomez
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia; Grupo Neuropsicología y Conducta, Universidad de Antioquia. Medellín, Colombia.
| | - Laura Bonanni
- Department of Medicine and Aging Sciences, G. d'Annunzio University, Chieti, Italy.
| | - Kolbjørn Brønnick
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norway; Faculty of Social Sciences, University of Stavanger, Stavanger, Norway.
| |
Collapse
|
291
|
Yoshino Y, Iga J, Ueno S. A Novel Mutation of FOXC1 (P136L) in an Axenfeld-Rieger Syndrome Patient With a Systematized Delusion of Jealousy: A Case Report and Literature Review. Mol Genet Genomic Med 2024; 12:e70008. [PMID: 39520097 PMCID: PMC11549375 DOI: 10.1002/mgg3.70008] [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: 03/11/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The main features of Axenfeld-Rieger Syndrome (ARS) are ocular, auditory, neurological, and morphological brain abnormalities. Mutations in forkhead box protein C1 (FOXC1) are among the responsible genes causing ARS, but neuropsychiatric features have rarely been reported. The case of an ARS patient (a 77-year-old man) with delusions of jealousy and impairment of working memory, in addition to the main clinical features, glaucoma and leukoencephalopathy, is presented. METHODS The mutation in the patient's genome was found with whole exome sequencing and in silico analysis using PolyPhen-2 and SIFT. Furthermore, AlphaFold2 and PyMOL were used to predict the protein structure based on the mutation. RESULTS A novel mutation at the forkhead domain of FOXC1 gene (c.408C>A, p.Phe136Leu) was found and confirmed in the patient's family, and it was predicted to cause protein damage; the SIFT score was 0, meaning deleterious, and the PolyPhen2 result also indicated damaging (score: 0.997). The predicted protein structure based on the novel mutation was different from that of the native structure. In the literature review, 6 of 95 (6.3%) cases showed neuropsychiatric features. Of them, 5 of 6 (83.3%) mutations were located in the forkhead domain. CONCLUSION A novel mutation was found in the FOXC1 gene (c.408C>A, p.Phe136Leu), which possibly induces delusions of jealousy and impairment of working memory, as well as features of ARS, by changing the protein structure. Mutations in that domain of the FOXC1 gene may be important not only for ocular abnormalities but also for brain function.
Collapse
Affiliation(s)
- Yuta Yoshino
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of MedicineToonEhimeJapan
| | - Jun‐ichi Iga
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of MedicineToonEhimeJapan
| | - Shu‐ichi Ueno
- Department of Neuropsychiatry, Molecules and FunctionEhime University Graduate School of MedicineToonEhimeJapan
| |
Collapse
|
292
|
Imokawa T, Yokoyama K, Takahashi K, Oyama J, Tsuchiya J, Sanjo N, Tateishi U. Brain perfusion SPECT in dementia: what radiologists should know. Jpn J Radiol 2024; 42:1215-1230. [PMID: 38888851 PMCID: PMC11522095 DOI: 10.1007/s11604-024-01612-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
The findings of brain perfusion single-photon emission computed tomography (SPECT), which detects abnormalities often before changes manifest in morphological imaging, mainly reflect neurodegeneration and contribute to dementia evaluation. A major shift is about to occur in dementia practice to the approach of diagnosing based on biomarkers and treating with disease-modifying drugs. Accordingly, brain perfusion SPECT will be required to serve as a biomarker of neurodegeneration. Hypoperfusion in Alzheimer's disease (AD) is typically seen in the posterior cingulate cortex and precuneus early in the disease, followed by the temporoparietal cortices. On the other hand, atypical presentations of AD such as the posterior variant, logopenic variant, frontal variant, and corticobasal syndrome exhibit hypoperfusion in areas related to symptoms. Additionally, hypoperfusion especially in the precuneus and parietal association cortex can serve as a predictor of progression from mild cognitive impairment to AD. In dementia with Lewy bodies (DLB), the differentiating feature is the presence of hypoperfusion in the occipital lobes in addition to that observed in AD. Hypoperfusion of the occipital lobe is not a remarkable finding, as it is assumed to reflect functional loss due to impairment of the cholinergic and dopaminergic systems rather than degeneration per se. Moreover, the cingulate island sign reflects the degree of AD pathology comorbid in DLB. Frontotemporal dementia is characterized by regional hypoperfusion according to the three clinical types, and the background pathology is diverse. Idiopathic normal pressure hydrocephalus shows apparent hypoperfusion around the Sylvian fissure and corpus callosum and apparent hyperperfusion in high-convexity areas. The cortex or striatum with diffusion restriction on magnetic resonance imaging in prion diseases reflects spongiform degeneration and brain perfusion SPECT reveals hypoperfusion in the same areas. Brain perfusion SPECT findings in dementia should be carefully interpreted considering background pathology.
Collapse
Affiliation(s)
- Tomoki Imokawa
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo, Japan
- Department of Radiology, Japanese Red Cross Omori Hospital, Ota-Ku, Tokyo, Japan
| | - Kota Yokoyama
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo, Japan.
| | - Kanae Takahashi
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo, Japan
| | - Jun Oyama
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo, Japan
| | - Junichi Tsuchiya
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo, Japan
| | - Nobuo Sanjo
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo, Japan
| | - Ukihide Tateishi
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo, Japan
| |
Collapse
|
293
|
Shimasaki R, Kurihara M, Hatano K, Goto R, Taira K, Ihara R, Higashihara M, Nishina Y, Kameyama M, Iwata A. Associations of cerebrospinal fluid monoamine metabolites with striatal dopamine transporter binding and 123I-meta-iodobenzylguanidine cardiac scintigraphy in Parkinson's disease: Multivariate analyses. Parkinsonism Relat Disord 2024; 128:107129. [PMID: 39241507 DOI: 10.1016/j.parkreldis.2024.107129] [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: 05/15/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Cerebrospinal fluid (CSF) homovanillic acid (HVA) and 5-hydroxyindole acetic acid (5-HIAA), dopamine and serotonin metabolites, are decreased in Parkinson's disease (PD). Although some reported associations between HVA and striatal dopamine transporter (DAT) or 5-HIAA and cardiac 123I-meta-iodobenzylguanidine (MIBG) findings, respectively, whether these are direct associations remained unknown. We retrospectively reviewed 57 drug-naïve patients with PD who underwent CSF analyses and DAT and cardiac MIBG imaging. Z-score of striatal DAT specific binding ratio (Z-SBR) was measured, and the positivity of MIBG abnormalities were judged by an expert. The mean age was 75.5 ± 8.7 years. Thirty-three were MIBG-positive and 24 were MIBG-negative. 5-HIAA levels were significantly lower in the MIBG-positive group. Logistic regression analysis showed that MIBG positivity was associated with 5-HIAA level (odds ratio = 0.751, p = 0.006) but not with age, sex, and HVA. DAT Z-SBR correlated with both HVA and 5-HIAA. Multiple regression analysis showed that HVA was the only significant variable associated with Z-SBR (t = 3.510, p < 0.001). We confirmed direct associations between 5-HIAA and cardiac MIBG, and between HVA and striatal DAT binding.
Collapse
Affiliation(s)
- Ryosuke Shimasaki
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Masanori Kurihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan; Integrated Research Initiative for Living Well with Dementia, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan.
| | - Keiko Hatano
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Ryoji Goto
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Kenichiro Taira
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Ryoko Ihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Yasushi Nishina
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Masashi Kameyama
- Department of Diagnostic Radiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan; Research Team for Neuroimaging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| | - Atsushi Iwata
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan; Integrated Research Initiative for Living Well with Dementia, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Japan
| |
Collapse
|
294
|
Couture V, Carmel J, Rousseau K, Létourneau G, Clerc D, Huot P, Forget M, Nguyen QD, Camicioli R, Desmarais P. Sex Differences in Neuropsychiatric Symptoms in Alpha-Synucleinopathies: A Systematic Review and Meta-Analysis. Mov Disord Clin Pract 2024; 11:1335-1344. [PMID: 39385552 PMCID: PMC11542278 DOI: 10.1002/mdc3.14227] [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: 06/16/2024] [Revised: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Alpha-synucleinopathies, such as Parkinson's disease (PD), Parkinson's disease dementia (PDD), and dementia with Lewy bodies (DLB), demonstrate sex differences with regard to prevalence, age of onset, and motor manifestations. Neuropsychiatric symptoms (NPS) are common early and late manifestations of these disorders. OBJECTIVES We aimed to describe sex differences in NPS across alpha-synucleinopathies. METHODS We searched Web of Science Core collection databases to identify observational studies published between January 1, 2000, and June 1, 2022, reporting the prevalence or severity of NPS among individuals with a diagnosis of PD, PDD, or DLB. Prevalence and severity were pooled for each NPS according to sex using random-effects models. RESULTS Two-hundred-and-forty studies, representing 796,026 participants (45% females), were included in the meta-analysis. Female sex was associated with a higher prevalence of anxiety (OR = 1.60 [95% CI: 1.40, 1.82]), depression (OR = 1.56 [1.45, 1.67]), fatigue (OR = 1.21 [1.02, 1.43]), and psychotic symptoms (OR = 1.26 [1.14, 1.40]) and more severe anxiety (g = 1.35 [95% CI: 0.58, 2.13]), depression (g = 1.57 [1.05, 2.08]), and fatigue (g = 0.86 [0.41, 1.32]), while male sex was associated with a higher prevalence of apathy (OR = 0.77 [0.63, 0.93]), impulse control disorders (OR = 0.67 [0.55, 0.82]), REM sleep behavior disorder (OR = 0.54 [0.42, 0.70]), hypersomnolence (OR = 0.67 [0.56, 0.80]), and suicide (OR = 0.30 [0.20, 0.44]). CONCLUSIONS NPS have different prevalences and severities in alpha-synucleinopathies according to sex. These findings support consideration of sex in the elaboration of clinical tools.
Collapse
Affiliation(s)
- Vincent Couture
- Department of Psychiatry, Division of Geriatric PsychiatryInstitut Universitaire en Santé Mentale de MontréalMontréalQCCanada
| | - Jean‐François Carmel
- Division of Geriatric PsychiatryInstitut Universitaire de Gériatrie de MontréalMontréalQCCanada
| | - Katerine Rousseau
- Department of Psychiatry, Division of Geriatric PsychiatryInstitut Universitaire en Santé Mentale de MontréalMontréalQCCanada
| | - Geneviève Létourneau
- Department of Psychiatry, Division of Geriatric PsychiatryInstitut Universitaire en Santé Mentale de MontréalMontréalQCCanada
| | - Doris Clerc
- Division of Geriatric PsychiatryInstitut Universitaire de Gériatrie de MontréalMontréalQCCanada
| | - Philippe Huot
- Department of Neurology and NeurosurgeryMcGill UniversityMontréalQCCanada
- Neurodegenerative Disease GroupMontreal Neurological Institute HospitalMontréalQCCanada
| | - Marie‐France Forget
- Department of Medicine, Division of GeriatricsCentre Hospitalier de l'Université de MontréalMontréalQCCanada
| | - Quoc Dinh Nguyen
- Department of Medicine, Division of GeriatricsCentre Hospitalier de l'Université de MontréalMontréalQCCanada
- Innovation Hub, Centre de Recherche du Centre Hospitalier de l'Université de MontréalMontréalQCCanada
| | - Richard Camicioli
- Department of Medicine and Neuroscience and Mental Health InstituteUniversity of AlbertaEdmontonABCanada
| | - Philippe Desmarais
- Department of Medicine, Division of GeriatricsCentre Hospitalier de l'Université de MontréalMontréalQCCanada
- Innovation Hub, Centre de Recherche du Centre Hospitalier de l'Université de MontréalMontréalQCCanada
| |
Collapse
|
295
|
de Oliveira FF, Miraldo MC, de Castro-Neto EF, de Almeida SS, de Andrade Matas SL, Bertolucci PHF, da Graça Naffah-Mazzacoratti M. Anthropometric and Demographic Features Affect the Interpretation of Cerebrospinal Fluid Biomarkers in Patients with Different Dementia Syndromes and Cognitively Healthy Adults. Neuromolecular Med 2024; 26:43. [PMID: 39487345 DOI: 10.1007/s12017-024-08810-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024]
Abstract
Clinical distinction between dementia with Lewy bodies (DLB) and late-onset Alzheimer's disease (AD) is difficult, while several features might affect the analyses of biomarkers. This study aimed to verify associations of anthropometric and demographic features with cerebrospinal fluid biomarkers, their ratios, and restructured traditional regression formulas in patients with DLB and AD, as well as in cognitively healthy controls. Consecutive outpatients with DLB were paired with outpatients with AD according to sex, dementia stage, and cognitive status, and with controls according to sex and age to investigate associations of sex, age, dementia duration, total sleep time, body mass index, alcohol use, smoking, sanitation, and APOE-ε4 alleles on the measurement of cerebrospinal fluid α-synuclein, biomarker ratios, and restructured traditional regression formulas involving amyloid-β (Aβ42,Aβ40,Aβ38), tau, and phospho-tau Thr181. Overall, 81 participants were included with DLB (n = 27;11 APOE-ε4 +) or AD (n = 27;12 APOE-ε4 +), and controls (n = 27;4 APOE-ε4 +); two thirds were women. Cerebrospinal fluid evidence of amyloidosis and tauopathy was more prevalent among women with AD, while Aβ42/Aβ38 could also discriminate men with DLB from men with AD. Restructured traditional regression formulas had higher diagnostic accuracy for women with AD. Aging, higher body mass index, and APOE-ε4 alleles were associated with amyloidosis in DLB, while only in AD were higher body mass index associated with lower tau pathology load, and more alcohol use associated with higher phospho-tau Thr181/Aβ42. These findings confirm the effects of anthropometric and demographic features on cerebrospinal fluid biomarkers, and also differences in aberrant amyloidosis and tauopathy between DLB and AD.
Collapse
Affiliation(s)
- Fabricio Ferreira de Oliveira
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, Vila Clementino, São Paulo, SP, CEP 04023-900, Brazil.
| | - Marjorie Câmara Miraldo
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, Vila Clementino, São Paulo, SP, CEP 04023-900, Brazil
| | - Eduardo Ferreira de Castro-Neto
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, Vila Clementino, São Paulo, SP, CEP 04023-900, Brazil
| | - Sandro Soares de Almeida
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Sandro Luiz de Andrade Matas
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, Vila Clementino, São Paulo, SP, CEP 04023-900, Brazil
| | - Paulo Henrique Ferreira Bertolucci
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, Vila Clementino, São Paulo, SP, CEP 04023-900, Brazil
| | - Maria da Graça Naffah-Mazzacoratti
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), Rua Botucatu 740, Vila Clementino, São Paulo, SP, CEP 04023-900, Brazil
| |
Collapse
|
296
|
Jean KR, Dotson VM. Dementia: Common Syndromes and Modifiable Risk and Protective Factors. Neurol Clin 2024; 42:793-807. [PMID: 39343475 DOI: 10.1016/j.ncl.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Dementia is an umbrella term for multiple conditions that lead to progressive cognitive decline and impaired activities of daily living. Neuropsychological evaluation is essential for characterizing the distinct cognitive and behavioral profile that can aid in the diagnostic process and treatment planning for dementia. Modifiable risk factors for dementia such as nutrition, physical activity, sleep, cognitive and social engagement, and stress provide important avenues for prevention. Neurologists and other health care providers can help patients reduce their risk for dementia by providing them with education about modifiable factors and connecting them to resources to empower them to engage in brain-healthy behavior.
Collapse
Affiliation(s)
- Kharine R Jean
- Department of Psychology, Georgia State University, PO Box 5010, Atlanta, GA 30302-5010, USA
| | - Vonetta M Dotson
- Department of Psychology, Georgia State University, PO Box 5010, Atlanta, GA 30302-5010, USA; Gerontology Institute, Georgia State University, PO Box 3984, Atlanta, GA 30302-3984, USA.
| |
Collapse
|
297
|
Malaty GR, Decourt B, Shill HA, Sabbagh MN. Biomarker Assessment in Parkinson's Disease Dementia and Dementia with Lewy Bodies by the Immunomagnetic Reduction Assay and Clinical Measures. J Alzheimers Dis Rep 2024; 8:1361-1371. [PMID: 39493956 PMCID: PMC11530035 DOI: 10.3233/adr-240110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/14/2024] [Indexed: 11/05/2024] Open
Abstract
Background Plasma biomarker assays provide an opportunity to reassess whether Alzheimer's disease, Parkinson's disease dementia (PDD), and dementia with Lewy bodies (DLB) plasma biomarkers are diagnostically useful. Objective We hypothesized that immunomagnetic reduction (IMR) of plasma biomarkers could differentiate between patients with PDD and DLB and healthy patients when combined with established clinical testing measures. Methods Plasma samples from 61 participants (12 PDD, 12 DLB, 37 controls) were analyzed using IMR to quantify amyloid-β 42 (Aβ42), total tau (t-tau), phosphorylated tau at threonine 181 (p-tau181), and α-synuclein (α-syn). Receiver operating characteristic curve (ROC) analysis was used to obtain sensitivity, specificity, and area under the ROC curve. Biomarker results were combined with clinical measures from the Unified Parkinson's Disease Rating Scale (UPDRS), Montreal Cognitive Assessment, and Hoehn-Yahr stage to optimize diagnostic test performance. Results Participants with PDD had higher α-syn than those with DLB and healthy participants and were distinguishable by their biomarker products Aβ42×p-tau181 and Aβ42×α-syn. Patients with DLB had higher p-tau181 than those with PDD and healthy participants and were distinguishable by their concentrations of α-syn×p-tau181. Plasma α-syn plus UPDRS versus either test alone increased sensitivity, specificity, and AUC when healthy patients were compared with those with PDD and DLB. Combined clinical examination scores and plasma biomarker products demonstrated utility in differentiating PDD from DLB when p-tau181 was combined with UPDRS, α-syn was combined with UPDRS, and α-syn×p-tau181 was combined with UPDRS. Conclusions In this pilot study, IMR plasma p-tau181 and α-syn may discriminate between PDD and DLB when used in conjunction with clinical testing.
Collapse
Affiliation(s)
- Giovanni R. Malaty
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Boris Decourt
- Department of Pharmacology and Neurosciences, Health Sciences Center, Texas Tech University, Lubbock, TX, USA
| | - Holly A. Shill
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| |
Collapse
|
298
|
Tikkanen V, Krüger J, Heikkinen AL, Hänninen T, Hublin C, Koivisto AM, Virkkala J, Saari TT, Remes AM, Paajanen TI. A Novel Computerized Flexible Attention Test in Detecting Executive Dysfunction of Patients with Early-Onset Cognitive Impairment and Dementia. Arch Clin Neuropsychol 2024; 39:817-828. [PMID: 38581151 DOI: 10.1093/arclin/acae026] [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: 09/28/2023] [Revised: 01/06/2024] [Accepted: 03/18/2024] [Indexed: 04/08/2024] Open
Abstract
OBJECTIVE The number of computer-based cognitive tests has increased in recent years, but there is a need for tests focusing on the assessment of executive function (EF), as it can be crucial for the identification of early-onset neurodegenerative disorders. This study aims to examine the ability of the Flexible Attention Test (FAT), a new computer-based test battery for detecting executive dysfunction of early-onset cognitive impairment and dementia patients. METHOD We analyzed the FAT subtask results in memory clinic patients with cognitive symptom onset at ≤65 years. The patients were divided into four groups: early onset dementia (EOD, n = 48), mild cognitive impairment due to neurological causes (MCI-n, n = 34), MCI due to other causes (MCI-o, n = 99), and subjective cognitive decline (SCD, n = 14). The test accuracy to distinguish EOD patients from other groups was examined, as well as correlations with pen-and-paper EF tests. We also reported the 12-months follow-up results. RESULTS The EOD and MCI-n patients performed significantly poorer (p ≤ .002) than those in the MCI-o and SCD groups in most of the FAT subtasks. The accuracies of the FAT subtasks to detect EOD from other causes were mainly moderate (0.34 ≤ area under the curve < 0.74). The FAT subtasks correlated logically with corresponding pen-and-paper EF tests (.15 ≤ r ≤ .75). No systematic learning effects were detected in the FAT performance at follow-up. CONCLUSIONS The FAT appears to be a promising method for the precise evaluation of EF and applicable distinguishing early-onset neurodegenerative disorders from patients with other causes of cognitive problems.
Collapse
Affiliation(s)
- Veera Tikkanen
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
| | - Johanna Krüger
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
| | - Anna-Leena Heikkinen
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
- Work Ability and Working Careers Unit, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Tuomo Hänninen
- Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Christer Hublin
- Work Ability and Working Careers Unit, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Anne M Koivisto
- Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Geriatrics, Helsinki University Hospital, Helsinki, Finland
- Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Jussi Virkkala
- Work Ability and Working Careers Unit, Finnish Institute of Occupational Health, Helsinki, Finland
- Clinical Neurophysiology and Clinical Neurosciences, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Toni T Saari
- Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Anne M Remes
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
- Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Teemu I Paajanen
- Work Ability and Working Careers Unit, Finnish Institute of Occupational Health, Helsinki, Finland
| |
Collapse
|
299
|
Orso B, Mattioli P, Yoon EJ, Kim YK, Kim H, Shin JH, Kim R, Famà F, Brugnolo A, Massa F, Chiaravalloti A, Fernandes M, Spanetta M, Placidi F, Pardini M, Bauckneht M, Morbelli S, Lee JY, Liguori C, Arnaldi D. Progression trajectories from prodromal to overt synucleinopathies: a longitudinal, multicentric brain [ 18F]FDG-PET study. NPJ Parkinsons Dis 2024; 10:200. [PMID: 39448609 PMCID: PMC11502916 DOI: 10.1038/s41531-024-00813-z] [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: 07/01/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024] Open
Abstract
The phenoconversion trajectory from idiopathic/isolated Rapid eye movement (REM) sleep behavior disorder (iRBD) towards either Parkinson's Disease (PD) or Dementia with Lewy Bodies (DLB) is currently uncertain. We investigated the capability of baseline brain [18F]FDG-PET in differentiating between iRBD patients eventually phenoconverting to PD or DLB, by deriving the denovoPDRBD-related pattern (denovoPDRBD-RP) from 32 de novo PD patients; and the denovoDLBRBD-RP from 30 de novo DLB patients, both with evidence of RBD at diagnosis. To explore [18F]FDG-PET phenoconversion trajectories prediction power, we applied these two patterns on a group of 115 iRBD patients followed longitudinally. At follow-up (25.6 ± 17.2 months), 42 iRBD patients progressed through overt alpha-synucleinopathy (21 iRBD-PD and 21 iRBD-DLB converters), while 73 patients remained stable at the last follow-up visit (43.2 ± 27.6 months). At survival analysis, both patterns were significantly associated with the phenoconversion trajectories. Brain [18F]FDG-PET is a promising biomarker to study progression trajectories in the alpha-synucleinopathy continuum.
Collapse
Grants
- MNESYS (PE0000006) Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN 2022, Protocol N. 20228XKKCM_001 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- MNESYS (PE0000006) Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- Fondi per la Ricerca Corrente Ministero della Salute (Ministry of Health, Italy)
- PNRR POC Ministero della Salute (Ministry of Health, Italy)
- 5x1000 founding scheme Ministero della Salute (Ministry of Health, Italy)
- NRF-2022R1A2C4001834 National Research Foundation of Korea (NRF)
Collapse
Affiliation(s)
- Beatrice Orso
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.
| | - Pietro Mattioli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Neurophysiopathology Unit, IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| | - Eun-Jin Yoon
- Memory Network Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Heejung Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Jung Hwan Shin
- Department of Neurology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ryul Kim
- Department of Neurology, Inha University Hospital, Incheon, Republic of Korea
| | - Francesco Famà
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Neurophysiopathology Unit, IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| | - Andrea Brugnolo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Clinical Psychology Unit, IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Clinical Neurology Unit, IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| | - Agostino Chiaravalloti
- IRCCS Neuromed, Pozzilli, Italy
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Mariana Fernandes
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Fabio Placidi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Sleep Medicine Center, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Clinical Neurology Unit, IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| | - Matteo Bauckneht
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine Unit, AOU Città Della Salute e Della Scienza di Torino, Torino, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Jee-Young Lee
- Department of Neurology, Seoul National University College of Medicine and Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Sleep Medicine Center, Neurology Unit, University Hospital "Tor Vergata", Rome, Italy
| | - Dario Arnaldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Neurophysiopathology Unit, IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| |
Collapse
|
300
|
Schroeder BE, Rivera-Rivera LA, Barger MR, de Chavez ER, Ospina-Romero M, Langhough RE, Teague JP, Carey FJ, Asthana S, Johnson SC, Johnson KM, Eisenmenger L, Wieben O, Salamat MS, Betthauser TJ. Cerebral artery and brain pathology correlates of antemortem cerebral artery 4D flow MRI. IMAGING NEUROSCIENCE (CAMBRIDGE, MASS.) 2024; 2:1-19. [PMID: 40092424 PMCID: PMC11908694 DOI: 10.1162/imag_a_00322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Large-scale clinical research studies often incorporate neuroimaging biomarkers to understand underlying pathologic changes that occur in aging and neurodegenerative disease and are associated with cognitive decline and clinical impairment. Of particular interest are neuroimaging methods designed to understand various aspects of cerebrovascular disease that can lead to dementia and also co-occur with neurodegenerative diseases such as Alzheimer's disease. Neurovascular 4D flow magnetic resonance imaging is one such method that measures hemodynamic characteristics of medium-large cerebral vessels, but it remains unclear how measures derived from 4D flow imaging including pulsatility index, cerebral blood flow, and cross-sectional area relate to underlying pathologic changes in cerebral arteries and downstream cerebrovascular pathology. For example, pulsatility index is thought to be a marker of vessel compliance, which may be due to fibrotic and/or atherosclerotic changes. This observational study investigates imaging-pathologic correlates of cerebral artery 4D flow MRI in 20 initial brain donors (mean (SD) age at death 78.2 (10.3) years; 3.2 (1.4) years from MRI to autopsy) from the Wisconsin Alzheimer's Disease Research Center that underwent antemortem imaging and postmortem assessment of cerebral artery and brain pathology to identify possible pathologic correlates of 4D flow MRI. Our results suggest that 4D flow MRI measures recapitulate expected hemodynamic and structural relationships across cerebral arteries, but also that measures like MRI cross-sectional area may reflect arterial fibrosis whereas mean blood flow may indicate downstream cerebrovascular disease including white matter rarefaction and arteriolosclerosis. In contrast, associations were minimal with pulsatility index and cerebral artery or brain pathology across participants but were moderate across arterial segments. To our knowledge, this is the first study to investigate pathologic correlates of antemortem 4D flow MRI in cerebral arteries. These results provide preliminary insights regarding the pathologic processes contributing to cerebral artery hemodynamics measured with 4D flow MRI that will help inform interpretation of large-scale clinical aging and dementia studies utilizing this method. Future work with larger samples is needed to confirm these findings.
Collapse
Affiliation(s)
- Brooke E Schroeder
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Duke University School of Medicine, Durham, North Carolina, USA
| | - Leonardo A Rivera-Rivera
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Madeleine R Barger
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Elena Ruiz de Chavez
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Monica Ospina-Romero
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jordan P Teague
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Finnuella J Carey
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Veterans Hospital, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kevin M Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Laura Eisenmenger
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Oliver Wieben
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - M Shahriar Salamat
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|