1
|
Mateo D, Carrión N, Cabrera C, Heredia L, Marquès M, Forcadell-Ferreres E, Pino M, Zaragoza J, Moral A, Cavallé L, González-de-Echávarri JM, Vicens P, Domingo JL, Torrente M. Gut Microbiota Alterations in Alzheimer's Disease: Relation with Cognitive Impairment and Mediterranean Lifestyle. Microorganisms 2024; 12:2046. [PMID: 39458354 PMCID: PMC11510339 DOI: 10.3390/microorganisms12102046] [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: 09/25/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Recently, an association between dysbiosis of the gut microbiota (GMB) and the development of several diseases, such as Alzheimer's disease (AD), has been proposed. Dysbiosis involves changes in microbial diversity influenced by environmental factors, like diet or lifestyle. In this study, we investigated the role of GMB parameters in Spanish AD patients, assessing the impact of adherence to the Mediterranean lifestyle (ML), as well as to characterize these parameters in relation to neuropsychological, neuropsychiatric, emotional, and functionality parameters. A case-control study was conducted to investigate the association between the composition of the GMB and cognitive, emotional, neuropsychiatric, and functionality status in Spanish AD patients, along with a shotgun metagenomics approach. Richness and alpha-diversity were significantly lower in the AD group compared to the controls. PERMANOVA and ANOSIM tests of Bray-Curtis dissimilarity, Aitchison distance, and Jaccard similarity did not showed significant differences in beta-diversity between the two groups. Moreover, associations between various phyla of the AD group and orientation performance, food consumption, and activities of daily living were identified. Dysbiosis observed in Spanish AD patients is characterized by reductions in richness and alpha-diversity, alongside alterations in GMB composition, which may be linked to adherence to the ML and cognitive and functionality symptoms.
Collapse
Affiliation(s)
- David Mateo
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Nerea Carrión
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
- Department of Psychology, Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Cristian Cabrera
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
- Department of Psychology, Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Institute Lerin Neurocognitive, Alzheimer and Other Neurocognitive Disorders Association, 43205 Reus, Spain
| | - Luis Heredia
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- Department of Psychology, Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Research Center for Behaviour Assessment (CRAMC), Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Department of Research Methods and Diagnosis in Education, Universidad Internacional de la Rioja, 26006 Logroño, Spain
| | - Montse Marquès
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Eva Forcadell-Ferreres
- Neurology, Hospital Verge de la Cinta de Tortosa, 43500 Tortosa, Spain; (E.F.-F.); (J.Z.)
| | - Maria Pino
- Cognitive Impairment Unit, University Hospital Sant Joan de Reus, 43204 Reus, Spain;
| | - Josep Zaragoza
- Neurology, Hospital Verge de la Cinta de Tortosa, 43500 Tortosa, Spain; (E.F.-F.); (J.Z.)
| | - Alfons Moral
- Neurology, Xarxa Santa Tecla, 43003 Tarragona, Spain;
| | - Lluís Cavallé
- Neurology, University Hospital Joan XXIII, 43005 Tarragona, Spain; (L.C.); (J.M.G.-d.-E.)
| | | | - Paloma Vicens
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- Department of Psychology, Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Research Center for Behaviour Assessment (CRAMC), Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - José L. Domingo
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
| | - Margarita Torrente
- Laboratory of Toxicology and Environmental Health (LSTM), Centre for Environmental, Food and Toxicological Technology (TECNATOX), Universitat Rovira i Virgili, 43201 Reus, Spain; (D.M.); (N.C.); (C.C.); (L.H.); (M.M.); (P.V.); (J.L.D.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
- Department of Psychology, Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
- Institute Lerin Neurocognitive, Alzheimer and Other Neurocognitive Disorders Association, 43205 Reus, Spain
- Research Center for Behaviour Assessment (CRAMC), Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| |
Collapse
|
2
|
Hu Y, Zhu T, Yuan M, Zhu H, Zhang W. Longitudinal association of depressive symptoms with cognition and neuroimaging biomarkers in cognitively unimpaired older adults, mild cognitive impairment, and Alzheimer's disease. Cereb Cortex 2024; 34:bhae423. [PMID: 39441024 DOI: 10.1093/cercor/bhae423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/11/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
We aimed to longitudinally examine the relationship between depression and cognitive function and investigate the mediating effects of imaging indicators in this relationship. 2,251 subjects with longitudinal assessment of geriatric depression scale, Mini-Mental State Examination, Montreal Cognitive Assessment, Clinical Dementia Rating-Sum of Boxes (CDRSB), Alzheimer's Disease Assessment Scale11, Alzheimer's Disease Assessment Scale13 and imaging of 3DT1, diffusion tensor imaging, fluid-attenuated inversion recovery, arterial spin labeling, fluorodeoxyglucose positron emission tomography, 18F-AV45-PET, and 18F-AV1451-PET were included from the Alzheimer's Disease Neuroimaging Initiative database. The multivariate mixed-effects models were employed to analyze the correlation between geriatric depression scale scores, cognitive function, and imaging indicators. The sgmediation software package was utilized to analyze the mediating effects of imaging indicators. The geriatric depression scale was negatively correlated with Mini-Mental State Examination and Montreal Cognitive Assessment, and positively correlated with CDRSB, Alzheimer's Disease Assessment Scale11, and Alzheimer's Disease Assessment Scale13 when the subjects were not grouped. The geriatric depression scale was negatively correlated with Montreal Cognitive Assessment and positively correlated with Alzheimer's Disease Assessment Scal13 in groups with baseline diagnosis of early mild cognitive impairment and late mild cognitive impairment. Furthermore, depression was associated with regional imaging indicators, while cognitive function was linked to broad imaging indicators. Some of these indicators were related to both depression and cognitive function, playing a mediating role in their relationship. Depression was related to cognitive function, especially in subjects with mild cognitive impairment. Some imaging indicators may represent the underlying basis for the association between depression and cognitive function.
Collapse
Affiliation(s)
- Ying Hu
- Department of Radiology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Ting Zhu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Hongru Zhu
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| | - Wei Zhang
- West China Biomedical Big Data Center of West China Hospital, Med-X Center for Informatics, Mental Health Center of West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Wang AY, Hu HY, Huang LY, Xiao CY, Li QY, Tan L, Hu H. Risk factors for cognitive decline in non-demented elders with amyloid-beta positivity. Alzheimers Res Ther 2024; 16:189. [PMID: 39160609 PMCID: PMC11331665 DOI: 10.1186/s13195-024-01554-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 08/11/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND As a currently incurable but preventable disease, the prevention and early diagnosis of Alzheimer's disease (AD) has long been a research hotspot. Amyloid deposition has been shown to be a major pathological feature of AD. Notably, not all the people with amyloid-beta (Aβ) pathology will have significant cognitive declines and eventually develop AD. Therefore, the aim of this study was to explore the risk factors for cognitive decline in Aβ-positive participants. METHODS We included 650 non-demented participants who were Aβ-positive at baseline from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Mixed effects and COX regression models were applied to assess 37 potential risk factors. Mixed effects models were employed to assess the temporal associations between potential risk factors and four cognitive assessment scales. COX regression models were used to assess the impact of potential risk factors on cognitive diagnosis conversion. Univariate and multivariate analyses were applied to the above models. Additionally, we used the Cochran-Armitage trend test to examine whether the incidence of cognitive decline increased with the number concurrent of risk factors. RESULTS Six factors (low diastolic pressure, low body mass index, retired status, a history of drug abuse, Parkinsonism, and depression) were the identified risk factors and four factors (a history of urinary disease, musculoskeletal diseases, no major surgical history, and no prior dermatologic-connective tissue diseases) were found to be suggestive risk factors. The incidence of cognitive decline in the Aβ-positive participants gradually increased as the number of concurrent risk factors increased (p for trend = 0.0005). CONCLUSIONS Our study may facilitate the understanding of the potential pathological processes in AD and provide novel targets for the prevention of cognitive decline among participants with Aβ positivity.
Collapse
Affiliation(s)
- An-Yi Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Chu-Yun Xiao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| |
Collapse
|
4
|
Tyrakis P, Agridi C, Kourti M. A Comprehensive Exploration of the Multifaceted Neuroprotective Role of Cannabinoids in Alzheimer's Disease across a Decade of Research. Int J Mol Sci 2024; 25:8630. [PMID: 39201317 PMCID: PMC11354546 DOI: 10.3390/ijms25168630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/15/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, manifests through dysregulation of brain function and subsequent loss of bodily control, attributed to β-amyloid plaque deposition and TAU protein hyperphosphorylation and aggregation, leading to neuronal death. Concurrently, similar cannabinoids to the ones derived from Cannabis sativa are present in the endocannabinoid system, acting through receptors CB1R and CB2R and other related receptors such as Trpv-1 and GPR-55, and are being extensively investigated for AD therapy. Given the limited efficacy and adverse effects of current available treatments, alternative approaches are crucial. Therefore, this review aims to identify effective natural and synthetic cannabinoids and elucidate their beneficial actions for AD treatment. PubMed and Scopus databases were queried (2014-2024) using keywords such as "Alzheimer's disease" and "cannabinoids". The majority of natural (Δ9-THC, CBD, AEA, etc.) and synthetic (JWH-133, WIN55,212-2, CP55-940, etc.) cannabinoids included showed promise in improving memory, cognition, and behavioral symptoms, potentially via pathways involving antioxidant effects of selective CB1R agonists (such as the BDNF/TrkB/Akt pathway) and immunomodulatory effects of selective CB2R agonists (TLR4/NF-κB p65 pathway). Combining anticholinesterase properties with a cannabinoid moiety may enhance therapeutic responses, addressing cholinergic deficits of AD brains. Thus, the positive outcomes of the vast majority of studies discussed support further advancing cannabinoids in clinical trials for AD treatment.
Collapse
Affiliation(s)
| | | | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus; (P.T.); (C.A.)
| |
Collapse
|
5
|
Wang B, Cui C, Chen Y, Liang Z. The mediating effect of 18F-FDG metabolism in right caudate between depressive symptoms and cognitive function in Alzheimer's disease. Front Aging Neurosci 2024; 16:1328143. [PMID: 38511197 PMCID: PMC10950943 DOI: 10.3389/fnagi.2024.1328143] [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: 10/26/2023] [Accepted: 02/14/2024] [Indexed: 03/22/2024] Open
Abstract
Purpose The objective of this study was to investigate the accumulation of 18F-fluorodeoxyglucose (18F-FDG) in the whole brain between Alzheimer's disease (AD) with depressive (ADD) symptoms compared with AD without depressive (ADND) symptoms using positron emission tomography/magnetic resonance imaging (PET/MRI). Additionally, this study aimed to explore the associations among the accumulation of 18F-FDG in the brain, depressive symptoms, and cognitive function in ADD patients. Methods In this study, 25 AD patients and 22 healthy controls were enrolled. The AD patients were stratified into two groups, namely ADD and ADND, based on their scores of the Hamilton Depression Scale (HAMD). Both AD patients and healthy controls underwent an 18F-FDG PET/MRI scan. A standardized uptake value ratio (SUVR) was calculated to examine the accumulation of 18F-FDG in the brain. A simple mediation model was employed to examine the mediation effect between SUVR, depressive symptoms and cognitive function in ADD patients. Results The ADD group exhibited significant cognitive impairment compared to the ADND group (p < 0.001) and healthy controls (p < 0.001). The ADD patients exhibited the reduced SUVR (0.228 ± 0.126) in the right caudate (the voxel level p < 0.005, cluster level p < 0.05, after false discovery rate (FDR) correction) compared to ADND patients (0.459 ± 0.064) and healthy controls (0.706 ± 0.122). The SUVR of the right caudate was correlated with the HAMD scores (r = -0.792, p < 0.001) and mini-mental state examination (MMSE) (r = 0.738, p < 0.01). The relationship between depressive symptoms and the cognitive function in ADD patients is mediated by the right caudate SUVR (total effects = -0.385, direct effects = -0.02, total indirect effects = -0.405). Conclusion The ADD group exhibited the reduced SUVR in the right caudate compared to the ADND group and healthy controls. The relationship between depressive symptoms and the cognitive ability of AD patients was mediated by the right caudate SUVR. The results contribute to a deeper understanding of the neurobiological mechanisms related to AD with depressive symptoms.
Collapse
Affiliation(s)
- Bojun Wang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chunlei Cui
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yifan Chen
- Department of Radiology, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Zhigang Liang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Zhang MZ, Sun Y, Chen YM, Guo F, Gao PY, Tan L, Tan MS. Associations of Multimorbidity with Cerebrospinal Fluid Biomarkers for Neurodegenerative Disorders in Early Parkinson's Disease: A Crosssectional and Longitudinal Study. Curr Alzheimer Res 2024; 21:201-213. [PMID: 39041277 DOI: 10.2174/0115672050314397240708060314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 07/24/2024]
Abstract
OBJECT The study aims to determine whether multimorbidity status is associated with cerebrospinal fluid (CSF) biomarkers for neurodegenerative disorders. METHODS A total of 827 patients were enrolled from the Parkinson's Progression Markers Initiative (PPMI) database, including 638 patients with early-stage Parkinson's disease (PD) and 189 healthy controls (HCs). Multimorbidity status was evaluated based on the count of long-term conditions (LTCs) and the multimorbidity pattern. Using linear regression models, cross-sectional and longitudinal analyses were conducted to assess the associations of multimorbidity status with CSF biomarkers for neurodegenerative disorders, including α-synuclein (αSyn), amyloid-β42 (Aβ42), total tau (t-tau), phosphorylated tau (p-tau), glial fibrillary acidic protein (GFAP), and neurofilament light chain protein (NfL). RESULTS At baseline, the CSF t-tau (p = 0.010), p-tau (p = 0.034), and NfL (p = 0.049) levels showed significant differences across the three categories of LTC counts. In the longitudinal analysis, the presence of LTCs was associated with lower Aβ42 (β < -0.001, p = 0.020), and higher t-tau (β = 0.007, p = 0.026), GFAP (β = 0.013, p = 0.022) and NfL (β = 0.020, p = 0.012); Participants with tumor/musculoskeletal/mental disorders showed higher CSF levels of t-tau (β = 0.016, p = 0.011) and p-tau (β = 0.032, p = 0.044) than those without multimorbidity. CONCLUSION Multimorbidity, especially severe multimorbidity and the pattern of mental/musculoskeletal/ tumor disorders, was associated with CSF biomarkers for neurodegenerative disorders in early-stage PD patients, suggesting that multimorbidity might play a crucial role in aggravating neuronal damage in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming-Zhan Zhang
- School of Clinical Medicine, Shandong Second Medical University (formerly Weifang Medical University), Weifang 261000, Shandong, China
| | - Yan Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan-Ming Chen
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Fan Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Meng-Shan Tan
- School of Clinical Medicine, Shandong Second Medical University (formerly Weifang Medical University), Weifang 261000, Shandong, China
- Department of Neurology, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
7
|
Johnson CE, Duncan MJ, Murphy MP. Sex and Sleep Disruption as Contributing Factors in Alzheimer's Disease. J Alzheimers Dis 2024; 97:31-74. [PMID: 38007653 PMCID: PMC10842753 DOI: 10.3233/jad-230527] [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: 11/27/2023]
Abstract
Alzheimer's disease (AD) affects more women than men, with women throughout the menopausal transition potentially being the most under researched and at-risk group. Sleep disruptions, which are an established risk factor for AD, increase in prevalence with normal aging and are exacerbated in women during menopause. Sex differences showing more disrupted sleep patterns and increased AD pathology in women and female animal models have been established in literature, with much emphasis placed on loss of circulating gonadal hormones with age. Interestingly, increases in gonadotropins such as follicle stimulating hormone are emerging to be a major contributor to AD pathogenesis and may also play a role in sleep disruption, perhaps in combination with other lesser studied hormones. Several sleep influencing regions of the brain appear to be affected early in AD progression and some may exhibit sexual dimorphisms that may contribute to increased sleep disruptions in women with age. Additionally, some of the most common sleep disorders, as well as multiple health conditions that impair sleep quality, are more prevalent and more severe in women. These conditions are often comorbid with AD and have bi-directional relationships that contribute synergistically to cognitive decline and neuropathology. The association during aging of increased sleep disruption and sleep disorders, dramatic hormonal changes during and after menopause, and increased AD pathology may be interacting and contributing factors that lead to the increased number of women living with AD.
Collapse
Affiliation(s)
- Carrie E. Johnson
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
| | - Marilyn J. Duncan
- University of Kentucky, College of Medicine, Department of Neuroscience, Lexington, KY, USA
| | - M. Paul Murphy
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY, USA
| |
Collapse
|
8
|
Sinclair LI, Mohr A, Morisaki M, Edmondson M, Chan S, Bone-Connaughton A, Turecki G, Love S. Is later-life depression a risk factor for Alzheimer's disease or a prodromal symptom: a study using post-mortem human brain tissue? Alzheimers Res Ther 2023; 15:153. [PMID: 37700368 PMCID: PMC10496415 DOI: 10.1186/s13195-023-01299-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/24/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Depression and dementia are both common diseases. Although new cases of depression are more common in younger adults, there is a second peak at the age of 50 years suggesting a different pathological process. Late-life depression (LLD) is associated with dementia. However, it remains unclear whether depression represents a dementia prodrome or is a true risk factor for its development. LLD is thought to have a vascular component and this may be a possible link between depression and dementia. We hypothesised that later-life depression is a prodromal manifestation of dementia and would therefore be associated with more AD, and/or ischaemic brain abnormalities that are present in earlier-life depression or in age- and sex-matched controls. METHODS We assessed post-mortem orbitofrontal cortex and dorsolateral pre-frontal cortex from 145 individuals in 4 groups: 28 18-50-year-olds with depression, 30 older individuals (ages 51-90) with depression, 28 with early AD (Braak tangle stages III-IV) and 57 matched controls (17 early-life, 42 later-life). Levels of Aβ, phospho-tau and α-synuclein were assessed by immunohistochemistry and ELISA. To quantify chronic ischaemia, VEGF, MAG and PLP1 were measured by ELISA. To assess pericyte damage, PDGFRB was measured by ELISA. For blood-brain barrier leakiness, JAM-A, claudin 5 and fibrinogen were measured by ELISA. To quantity endothelial activation, the ratio of ICAM1:collagen IV was assessed by immunohistochemistry. RESULTS There was no evidence of chronic cerebral hypoperfusion or increased Aβ/tau in either depression group. There was also no indication of pericyte damage, increased blood-brain barrier leakiness or endothelial activation in the OFC or DLPFC in the depression groups. CONCLUSIONS Contrary to some previous findings, we have not found evidence of impaired vascular function or increased Aβ in LLD. Our study had a relatively small sample size and limitations in the availability of clinical data. These results suggest that depression is a risk factor for dementia rather than an early manifestation of AD or a consequence of cerebral vascular insufficiency.
Collapse
Affiliation(s)
- Lindsey I Sinclair
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Level 1 Learning & Research Building, Bristol, BS10 5NB, UK.
| | - Asher Mohr
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Level 1 Learning & Research Building, Bristol, BS10 5NB, UK
| | - Mizuki Morisaki
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Level 1 Learning & Research Building, Bristol, BS10 5NB, UK
| | - Martin Edmondson
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Level 1 Learning & Research Building, Bristol, BS10 5NB, UK
| | - Selina Chan
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Level 1 Learning & Research Building, Bristol, BS10 5NB, UK
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, Canada
| | - A Bone-Connaughton
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Level 1 Learning & Research Building, Bristol, BS10 5NB, UK
| | - Gustavo Turecki
- Department of Life Sciences, Warwick University, Warwick, UK
| | - Seth Love
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Level 1 Learning & Research Building, Bristol, BS10 5NB, UK
| |
Collapse
|
9
|
Cannizzaro A, Ronat L, El Haffaf LM, Hanganu A. Associations between neuropsychiatric symptoms of affective and vegetative domains and brain morphology in aging people with mild cognitive impairment and Alzheimer's disease. Int J Geriatr Psychiatry 2023; 38:e5952. [PMID: 37351584 DOI: 10.1002/gps.5952] [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: 01/29/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
OBJECTIVE Neuropsychiatric symptoms (NPS) are common in mild cognitive impairment (MCI) and even more in Alzheimer's disease (AD). The symptom-based cluster including nighttime disturbances, depression, appetite changes, anxiety, and apathy (affective and vegetative symptoms) was associated with an increased risk of dementia in MCI and has common neuroanatomical associations. Our objective was to investigate the differences in brain morphology associations with affective and vegetative symptoms between three groups: cognitively normal older adults (CN), MCI and AD. MATERIAL AND METHODS Alzheimer's Disease Neuroimaging Initiative data of 223 CN, 367 MCI and 175 AD, including cortical volumes, surface areas and thicknesses and severity scores of the five NPS were analyzed. A whole-brain vertex-wise general linear model was performed to test for intergroup differences (CN-MCI, CN-AD, AD-MCI) in brain morphology associations with five NPS. Multiple regressions were conducted to investigate cortical change as a function of NPS severity in the AD-MCI contrast. RESULTS We found (1) signature differences in nighttime disturbances associations with prefrontal regions in AD-MCI, (2) signature differences in NPS associations with temporal regions in AD-MCI for depression and in CN-AD for anxiety, (3) decreased temporal metrics in MCI as nighttime disturbances and depression severity increased, (4) decreased pars triangularis metrics in AD as nighttime disturbances and apathy severity increased. CONCLUSION Each NPS seems to have a signature on brain morphology. Affective and vegetative NPS were primarily associated with prefrontal and temporal regions. These signatures open the possibility of potential future assessments of links between brain morphology and NPS on an individual level.
Collapse
Affiliation(s)
- Adriana Cannizzaro
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, Quebec, Canada
- Faculté des Arts et des Sciences, Département de Psychologie, Université de Montréal, Montreal, Quebec, Canada
| | - Lucas Ronat
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, Quebec, Canada
- Faculté de Médecine, Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Lyna Mariam El Haffaf
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, Quebec, Canada
- Faculté des Arts et des Sciences, Département de Psychologie, Université de Montréal, Montreal, Quebec, Canada
| | - Alexandru Hanganu
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montreal, Quebec, Canada
- Faculté des Arts et des Sciences, Département de Psychologie, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Imai A, Matsuoka T, Narumoto J. Emotional Dysregulation in Mild Behavioral Impairment Is Associated with Reduced Cortical Thickness in the Right Supramarginal Gyrus. J Alzheimers Dis 2023; 93:521-532. [PMID: 37038811 DOI: 10.3233/jad-220948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
BACKGROUND Mild behavioral impairment (MBI) has attracted attention as a possible precursor symptom of dementia, but its neural basis has not been fully investigated. OBJECTIVE We aimed to investigate the relationship between MBI and surface area, cortical thickness, and volume in the temporal and parietal lobes, which are strongly associated with dementia and emotional disorders. METHODS This retrospective study evaluated 123 participants: 90 with mild cognitive impairment (MCI), 13 with subjective cognitive decline (SCD), and 20 cognitively healthy (CH). Using analysis of covariance (ANCOVA) with sex, age, and MMSE score as covariates, cortical thickness, surface area, and volume in 10 regions were compared between groups with and without MBI. Groups with MBI emotional dysregulation were also compared with groups without MBI. RESULTS ANCOVA revealed significantly smaller cortical thickness in the MBI group's right parahippocampal (p = 0.01) and supramarginal gyri (p = 0.002). After multiple comparison correction, only the right supramarginal gyrus was significantly smaller (p = 0.02). When considering only MBI emotional dysregulation, the right parahippocampal and supramarginal gyrus' cortical thicknesses were significantly smaller in this MBI group (p = 0.03, 0.01). However, multiple comparison correction identified no significant differences (p = 0.14, 0.11). CONCLUSION Overall MBI and the emotional dysregulation domains were associated with reduced cortical thickness in the right parahippocampal and supramarginal gyri. Since neurodegeneration in the medial temporal and parietal lobe precedes early Alzheimer's disease (AD), MBI, particularly emotion dysregulation, may predict early AD below the diagnostic threshold.
Collapse
Affiliation(s)
- Ayu Imai
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Teruyuki Matsuoka
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jin Narumoto
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
11
|
Nowrangi MA, Outen JD, Kim J, Avramopoulos D, Lyketsos CG, Rosenberg PB. Neuropsychiatric Symptoms of Alzheimer's Disease: An Anatomic-Genetic Framework for Treatment Development. J Alzheimers Dis 2023; 95:53-68. [PMID: 37522204 DOI: 10.3233/jad-221247] [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: 08/01/2023]
Abstract
BACKGROUND Despite the burden on patients and caregivers, there are no approved therapies for the neuropsychiatric symptoms of Alzheimer's disease (NPS-AD). This is likely due to an incomplete understanding of the underlying mechanisms. OBJECTIVE To review the neurobiological mechanisms of NPS-AD, including depression, psychosis, and agitation. METHODS Understanding that genetic encoding gives rise to the function of neural circuits specific to behavior, we review the genetics and neuroimaging literature to better understand the biological underpinnings of depression, psychosis, and agitation. RESULTS We found that mechanisms involving monoaminergic biosynthesis and function are likely key elements of NPS-AD and while current treatment approaches are in line with this, the lack of effectiveness may be due to contributions from additional mechanisms including neurodegenerative, vascular, inflammatory, and immunologic pathways. CONCLUSION Within an anatomic-genetic framework, development of novel effective biological targets may engage targets within these pathways but will require a better understanding of the heterogeneity in NPS-AD.
Collapse
Affiliation(s)
- Milap A Nowrangi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - John D Outen
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitrios Avramopoulos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Constantine G Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| |
Collapse
|
12
|
Pinus halepensis Essential Oil Ameliorates Aβ1-42-Induced Brain Injury by Diminishing Anxiety, Oxidative Stress, and Neuroinflammation in Rats. Biomedicines 2022; 10:biomedicines10092300. [PMID: 36140401 PMCID: PMC9496595 DOI: 10.3390/biomedicines10092300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/04/2022] [Accepted: 09/12/2022] [Indexed: 01/18/2023] Open
Abstract
The Pinus L. genus comprises around 250 species, being popular worldwide for their medicinal and aromatic properties. The present study aimed to evaluate the P. halepensis Mill. essential oil (PNO) in an Alzheimer’s disease (AD) environment as an anxiolytic and antidepressant agent. The AD-like symptoms were induced in Wistar male rats by intracerebroventricular administration of amyloid beta1-42 (Aβ1-42), and PNO (1% and 3%) was delivered to Aβ1-42 pre-treated rats via inhalation route for 21 consecutive days, 30 min before behavioral assessments. The obtained results indicate PNO’s potential to relieve anxious–depressive features and to restore redox imbalance in the rats exhibiting AD-like neuropsychiatric impairments. Moreover, PNO presented beneficial effects against neuroinflammation and neuroapoptosis in the Aβ1-42 rat AD model.
Collapse
|
13
|
Anxiety and depression in Alzheimer's disease: a systematic review of pathogenetic mechanisms and relation to cognitive decline. Neurol Sci 2022; 43:4107-4124. [PMID: 35461471 PMCID: PMC9213384 DOI: 10.1007/s10072-022-06068-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/06/2022] [Indexed: 12/13/2022]
Abstract
Objectives To explore the pathogenetic hypothesis provided to explain the comorbidity of anxious and depressive symptomatology and AD and to assess the association between anxious and depressive symptoms and the AD-related cognitive impairment. Methods In October 2020 and March 2021, PsycINFO, Embase, Ovid, and CINAHL were searched for peer-reviewed original articles investigating anxiety and/or depression in AD. Results A total of 14,760 studies were identified and 34 papers on AD patients were included in the review. Suggested biological causes of depression and anxiety in AD include higher strychnine-sensitive glycine receptor (GlyRS) functioning and selective reduction of N-methyl-d-aspartate (NMDA) receptor NR2A density, cortical and limbic atrophy, lower resting cortical metabolism, lower CSF Aβ42 and higher t-tau and p-tau levels, and neuritic plaques. At the same time, dysthymia arises in the early stages of AD as an emotional reaction to the progressive cognitive decline and can cause it; anxiety can appear as an initial compensating behaviour; and depression might be related to AD awareness and loss of functional abilities. Affective symptoms and the expression of the depressive symptoms tend to reduce as AD progresses. Conclusion The neurodegeneration of areas and circuits dealing with emotions can elicit anxiety and depression in AD. In the early stages of the disease, anxiety and depression could arise as a psychological reaction to AD and due to coping difficulties. In late AD stages, the cognitive impairment reduces the emotional responses and their expression. Anxiety and depression are more intense in early-onset AD, due to the major impact of AD on the individual. Supplementary Information The online version contains supplementary material available at 10.1007/s10072-022-06068-x.
Collapse
|
14
|
Yang L, Shu J, Yan A, Yang F, Xu Z, Wei W. White matter hyperintensities-related cortical changes and correlation with mild behavioral impairment. Adv Med Sci 2022; 67:241-249. [PMID: 35780532 DOI: 10.1016/j.advms.2022.06.002] [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: 01/19/2022] [Revised: 04/16/2022] [Accepted: 06/09/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE The aim of this study was to analyze cortical thickness and gray matter volume (GMV) changes in white matter hyperintensities (WMH) which were associated brain regions and their association with mild behavioral impairment (MBI) by means of voxel- and surface-based morphology (VBM and SBM). METHODS A total of 60 patients underwent 3T MRI scan and MBI checklist (MBI-C) assessment and were divided into two groups: lower WMH (LWMH) and higher WMH (HWMH). After adjusting for confounding factors i.e. age, gender, education, and total intracranial volume, we found a GMV decrease in the left anterior insula (AIns), right middle frontal gyrus, right central operculum, right fusiform gyrus, left cerebellum exterior, and thalamus proper in the HWMH group based VBM, while in the HWMH group based SBM we found cortical thickness decrease in the left lingual, right posterior cingulate cortex (rPCC), right precentral, left superior frontal, right medial orbitofrontal gyrus, and left pars opercularis. RESULTS The HWMH group had higher MBI-C scores. The GMV in the left AIns and thalamus proper and the thickness of rPCC negatively correlated with the MBI-C scores. The mediation analysis suggested that WMH may partially mediate MBI-C scores by reducing the GMV and cortical thickness of the mentioned brain regions. CONCLUSIONS In WMH patients, the occurrence of MBI is associated with atrophy of gray matter and cortex. The occurrence of MBI may be partially mediated by WMH through gray matter and cortical atrophy. It provides a new insight into the relationship between WMH and dementia.
Collapse
Affiliation(s)
- Lu Yang
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jun Shu
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Aijuan Yan
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Fuxia Yang
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Ziwei Xu
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Wenshi Wei
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Barca ML, Alnæs D, Engedal K, Persson K, Eldholm RS, Siafarikas N, Selseth Almdahl I, Stylianou-Korsnes M, Saltvedt I, Selbæk G, Westlye LT. Brain Morphometric Correlates of Depressive Symptoms among Patients with and without Dementia. Dement Geriatr Cogn Dis Extra 2022; 12:107-114. [PMID: 35950148 PMCID: PMC9251457 DOI: 10.1159/000521114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 12/02/2022] Open
Abstract
Introduction Findings regarding brain morphometry among patients with dementia and concomitant depressive symptoms have been inconsistent. Thus, the aim of the present study was to test the hypothesis that dementia and concomitant depressive symptoms are associated with structural brain changes in the temporal lobe measured with structural magnetic resonance imaging (MRI). Methods A sample of 492 patients from Norwegian memory clinics (n = 363) and Old Age Psychiatry services (n = 129) was studied. The assessment included the Cornell Scale for Depression in Dementia (CSDD), Instrumental Activities of Daily Living Scale, Mini Mental State Examination, and MRI of the brain, processed with FreeSurfer to derive ROI measures of cortical thickness, volume, and area using the Desikan-Killiany parcellation, as well as subcortical volumes. Dementia was diagnosed according to ICD-10 research criteria. Correlates of brain morphometry using multiple linear regression were examined. Results Higher scores on the CSDD were associated with larger cortical volume (β = 0.125; p value = 0.003) and area of the left isthmus of the cingulate gyrus (β = 0.151; p value = <0.001) across all patients. Inclusion of an interaction term (dementia × CSDD) revealed a smaller area in the left temporal pole (β = −0.345; p value = 0.001) and right-transverse temporal cortex (β = −0.321; p value = 0.001) in patients with dementia and depressive symptoms. Discussion/Conclusion We confirm the previous findings of structural brain changes in temporal regions among patients with dementia and concomitant depressive symptoms. This may contribute to a better understanding of the mechanisms underlying depression in dementia. To the best of our knowledge, this is the largest study conducted on this topic to date.
Collapse
Affiliation(s)
- Maria Lage Barca
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
- *Maria Lage Barca,
| | - Dag Alnæs
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Knut Engedal
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Karin Persson
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Rannveig Sakshaug Eldholm
- Department of Neuromedicine and Movement Science, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nikias Siafarikas
- Department of Geriatric Psychiatry, Akershus University Hospital, Lørenskog, Norway
| | - Ina Selseth Almdahl
- Department of Old Age Psychiatry, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Maria Stylianou-Korsnes
- Department of Old Age Psychiatry, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ingvild Saltvedt
- Department of Neuromedicine and Movement Science, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Geriatric Department, St. Olav Hospital, University Hospital of Trondheim, Trondheim, Norway
| | - Geir Selbæk
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| |
Collapse
|
16
|
Moulinet I, Touron E, Mézenge F, Dautricourt S, De La Sayette V, Vivien D, Marchant NL, Poisnel G, Chételat G. Depressive Symptoms Have Distinct Relationships With Neuroimaging Biomarkers Across the Alzheimer’s Clinical Continuum. Front Aging Neurosci 2022; 14:899158. [PMID: 35795235 PMCID: PMC9251580 DOI: 10.3389/fnagi.2022.899158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023] Open
Abstract
Background Depressive and anxiety symptoms are frequent in Alzheimer’s disease and associated with increased risk of developing Alzheimer’s disease in older adults. We sought to examine their relationships to Alzheimer’s disease biomarkers across the preclinical and clinical stages of the disease. Method Fifty-six healthy controls, 35 patients with subjective cognitive decline and 56 amyloid-positive cognitively impaired patients on the Alzheimer’s continuum completed depression and anxiety questionnaires, neuropsychological tests and neuroimaging assessments. We performed multiple regressions in each group separately to assess within group associations of depressive and anxiety symptoms with either cognition (global cognition and episodic memory) or neuroimaging data (gray matter volume, glucose metabolism and amyloid load). Results Depressive symptoms, but not anxiety, were higher in patients with subjective cognitive decline and cognitively impaired patients on the Alzheimer’s continuum compared to healthy controls. Greater depressive symptoms were associated with higher amyloid load in subjective cognitive decline patients, while they were related to higher cognition and glucose metabolism, and to better awareness of cognitive difficulties, in cognitively impaired patients on the Alzheimer’s continuum. In contrast, anxiety symptoms were not associated with brain integrity in any group. Conclusion These data show that more depressive symptoms are associated with greater Alzheimer’s disease biomarkers in subjective cognitive decline patients, while they reflect better cognitive deficit awareness in cognitively impaired patients on the Alzheimer’s continuum. Our findings highlight the relevance of assessing and treating depressive symptoms in the preclinical stages of Alzheimer’s disease.
Collapse
Affiliation(s)
- Inès Moulinet
- Physiopathology and Imaging of Neurological Disorders (PhIND), Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Université de Caen Normandie, Caen, France
| | - Edelweiss Touron
- Physiopathology and Imaging of Neurological Disorders (PhIND), Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Université de Caen Normandie, Caen, France
| | - Florence Mézenge
- Physiopathology and Imaging of Neurological Disorders (PhIND), Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Université de Caen Normandie, Caen, France
| | - Sophie Dautricourt
- Physiopathology and Imaging of Neurological Disorders (PhIND), Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Université de Caen Normandie, Caen, France
- CHU de Caen, Service de Neurologie, Caen, France
| | | | - Denis Vivien
- Physiopathology and Imaging of Neurological Disorders (PhIND), Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Université de Caen Normandie, Caen, France
- Département de Recherche Clinique, CHU de Caen-Normandie, Caen, France
| | | | - Géraldine Poisnel
- Physiopathology and Imaging of Neurological Disorders (PhIND), Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Université de Caen Normandie, Caen, France
| | - Gaël Chételat
- Physiopathology and Imaging of Neurological Disorders (PhIND), Institut National de la Santé et de la Recherche Médicale, Blood and Brain @ Caen-Normandie, GIP Cyceron, Université de Caen Normandie, Caen, France
- *Correspondence: Gaël Chételat,
| |
Collapse
|
17
|
Jellinger KA. Pathomechanisms of Vascular Depression in Older Adults. Int J Mol Sci 2021; 23:ijms23010308. [PMID: 35008732 PMCID: PMC8745290 DOI: 10.3390/ijms23010308] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023] Open
Abstract
Depression in older individuals is a common complex mood disorder with high comorbidity of both psychiatric and physical diseases, associated with high disability, cognitive decline, and increased mortality The factors predicting the risk of late-life depression (LLD) are incompletely understood. The reciprocal relationship of depressive disorder and age- and disease-related processes has generated pathogenic hypotheses and provided various treatment options. The heterogeneity of depression complicates research into the underlying pathogenic cascade, and factors involved in LLD considerably differ from those involved in early life depression. Evidence suggests that a variety of vascular mechanisms, in particular cerebral small vessel disease, generalized microvascular, and endothelial dysfunction, as well as metabolic risk factors, including diabetes, and inflammation that may induce subcortical white and gray matter lesions by compromising fronto-limbic and other important neuronal networks, may contribute to the development of LLD. The "vascular depression" hypothesis postulates that cerebrovascular disease or vascular risk factors can predispose, precipitate, and perpetuate geriatric depression syndromes, based on their comorbidity with cerebrovascular lesions and the frequent development of depression after stroke. Vascular burden is associated with cognitive deficits and a specific form of LLD, vascular depression, which is marked by decreased white matter integrity, executive dysfunction, functional disability, and poorer response to antidepressive therapy than major depressive disorder without vascular risk factors. Other pathogenic factors of LLD, such as neurodegeneration or neuroimmune regulatory dysmechanisms, are briefly discussed. Treatment planning should consider a modest response of LLD to antidepressants, while vascular and metabolic factors may provide promising targets for its successful prevention and treatment. However, their effectiveness needs further investigation, and intervention studies are needed to assess which interventions are appropriate and effective in clinical practice.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150 Vienna, Austria
| |
Collapse
|
18
|
Neuropsychiatric symptoms and brain morphology in patients with mild cognitive impairment and Alzheimer's disease with dementia. Int Psychogeriatr 2021; 33:1217-1228. [PMID: 34399870 DOI: 10.1017/s1041610221000934] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
UNLABELLED We present associations between neuropsychiatric symptoms (NPS) and brain morphology in a large sample of patients with mild cognitive impairment (MCI) and Alzheimer's disease with dementia (AD dementia).Several studies assessed NPS factor structure in MCI and AD dementia, but we know of no study that tested for associations between NPS factors and brain morphology. The use of factor scores increases parsimony and power. For transparency, we performed an additional analysis with selected Neuropsychiatric Inventory - Questionnaire (NPI-Q) items. Including regional cortical thickness, cortical and subcortical volumes, we examined associations between NPS and brain morphology across the whole brain in an unbiased fashion. We reported both statistical significance and effect sizes, using linear models adjusted for multiple comparisons by false discovery rate (FDR). Moreover, we included an interaction term for diagnosis and could thereby compare associations of NPS and brain morphology between MCI and AD dementia.We found an association between the factor elation and thicker right anterior cingulate cortex across MCI and AD dementia. Associations between the factors depression to thickness of the banks of the left superior temporal sulcus and psychosis to the left post-central volume depended on diagnosis: in MCI these associations were positive, in AD dementia negative.Our findings indicate that NPS in MCI and AD dementia are not exclusively associated with atrophy and support previous findings of associations between NPS and mainly frontotemporal brain structures. OBJECTIVES Neuropsychiatric symptoms (NPS) are common in mild cognitive impairment (MCI) and Alzheimer’s disease with dementia (AD dementia), but their brain structural correlates are unknown. We tested for associations between NPS and MRI-based cortical and subcortical morphometry in patients with MCI and AD dementia. DESIGN Cross-sectional. SETTINGS Conducted in Norway. PARTICIPANTS Patients with MCI (n = 102) and AD dementia (n = 133) from the Memory Clinic and the Geriatric Psychiatry Unit at Oslo University Hospital. MEASUREMENTS Neuropsychiatric Inventory – Questionnaire (NPI-Q) severity indices were reduced using principal component analysis (PCA) and tested for associations with 170 MRI features using linear models and false discovery rate (FDR) adjustment. We also tested for differences between groups. For transparency, we added analyses with selected NPI-Q items. RESULTS PCA revealed four factors: elation, psychosis, depression, and motor behavior.FDR adjustment revealed a significant positive association (B = 0.20, pFDR < 0.005) between elation and thickness of the right caudal anterior cingulate cortex (ACC) across groups, and significant interactions between diagnosis and psychosis (B = −0.48, pFDR < 0.0010) on the left post-central volume and between diagnosis and depression (B = −0.40, pFDR < 0.005) on the thickness of the banks of the left superior temporal sulcus. Associations of apathy, anxiety, and nighttime behavior to the left temporal lobe were replicated. CONCLUSIONS The positive association between elation and ACC thickness suggests that mechanisms other than atrophy underly elation. Interactions between diagnosis and NPS on MRI features suggest different mechanisms of NPS in our MCI and AD dementia samples. The results contribute to a better understanding of NPS brain mechanisms in MCI and AD dementia.
Collapse
|
19
|
Lee M, Nam JH, Yi E, Bhimla A, Nelson J, Ma GX. Differences in Subjective Memory Impairment, Depressive Symptoms, Sleep, and Physical Activity in African American and Asian American Elderly. Brain Sci 2021; 11:1155. [PMID: 34573176 PMCID: PMC8472213 DOI: 10.3390/brainsci11091155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 01/11/2023] Open
Abstract
Background: Subjective memory impairment (SMI) is associated with negative health outcomes including mild cognitive impairment and Alzheimer's disease. However, ethnic differences in SMI and disparities in risk factors associated with SMI among minority populations are understudied. The study examined the ethnic differences in SMI, whether SMI was associated with depressive symptoms, sleep, and physical activity (PA), and whether the associations vary across racial/ethnic groups. Methods: Participants included 243 African and Asian Americans (including Chinese, Vietnamese, and Korean Americans) aged 50 or older. Demographic information, SMI, depressive symptoms, daily sleeping hours, and PA levels were assessed. Results: Vietnamese Americans reported the highest SMI score. Depressive symptoms, sleeping hours, and PA levels were significantly associated with SMI. Depressive symptoms were the only significant factor across all ethnic groups. Significant interaction effects were found between ethnicity and health behaviors in predicting SMI. In particular, Vietnamese American participants with greater depressive symptoms and physical inactivity were significantly more likely to experience SMI compared to other ethnic groups Conclusions: Our findings demonstrate ethnic differences in SMI and its association with depressive symptoms, sleep, and PA, which highlight the importance of considering the unique cultural and historical backgrounds across different racial/ethnic groups when examining cognitive functioning in elderly.
Collapse
Affiliation(s)
- Minsun Lee
- Center for Asian Health, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.L.); (J.-H.N.); (E.Y.); (A.B.)
| | - Jin-Hyeok Nam
- Center for Asian Health, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.L.); (J.-H.N.); (E.Y.); (A.B.)
| | - Elizabeth Yi
- Center for Asian Health, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.L.); (J.-H.N.); (E.Y.); (A.B.)
| | - Aisha Bhimla
- Center for Asian Health, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.L.); (J.-H.N.); (E.Y.); (A.B.)
| | - Julie Nelson
- Philadelphia Senior Center, Philadelphia, PA 19147, USA;
| | - Grace X. Ma
- Center for Asian Health, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (M.L.); (J.-H.N.); (E.Y.); (A.B.)
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
20
|
Monereo-Sánchez J, Schram MT, Frei O, O’Connell K, Shadrin AA, Smeland OB, Westlye LT, Andreassen OA, Kaufmann T, Linden DEJ, van der Meer D. Genetic Overlap Between Alzheimer's Disease and Depression Mapped Onto the Brain. Front Neurosci 2021; 15:653130. [PMID: 34290577 PMCID: PMC8288283 DOI: 10.3389/fnins.2021.653130] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Alzheimer's disease (AD) and depression are debilitating brain disorders that are often comorbid. Shared brain mechanisms have been implicated, yet findings are inconsistent, reflecting the complexity of the underlying pathophysiology. As both disorders are (partly) heritable, characterising their genetic overlap may provide aetiological clues. While previous studies have indicated negligible genetic correlations, this study aims to expose the genetic overlap that may remain hidden due to mixed directions of effects. Methods: We applied Gaussian mixture modelling, through MiXeR, and conjunctional false discovery rate (cFDR) analysis, through pleioFDR, to genome-wide association study (GWAS) summary statistics of AD (n = 79,145) and depression (n = 450,619). The effects of identified overlapping loci on AD and depression were tested in 403,029 participants of the UK Biobank (UKB) (mean age 57.21, 52.0% female), and mapped onto brain morphology in 30,699 individuals with brain MRI data. Results: MiXer estimated 98 causal genetic variants overlapping between the 2 disorders, with 0.44 concordant directions of effects. Through pleioFDR, we identified a SNP in the TMEM106B gene, which was significantly associated with AD (B = -0.002, p = 9.1 × 10-4) and depression (B = 0.007, p = 3.2 × 10-9) in the UKB. This SNP was also associated with several regions of the corpus callosum volume anterior (B > 0.024, p < 8.6 × 10-4), third ventricle volume ventricle (B = -0.025, p = 5.0 × 10-6), and inferior temporal gyrus surface area (B = 0.017, p = 5.3 × 10-4). Discussion: Our results indicate there is substantial genetic overlap, with mixed directions of effects, between AD and depression. These findings illustrate the value of biostatistical tools that capture such overlap, providing insight into the genetic architectures of these disorders.
Collapse
Affiliation(s)
- Jennifer Monereo-Sánchez
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Miranda T. Schram
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Internal Medicine, School for Cardiovascular Disease (CARIM), Maastricht University, Maastricht, Netherlands
- Heart and Vascular Centre, Maastricht University Medical Center, Maastricht, Netherlands
| | - Oleksandr Frei
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Informatics, Centre for Bioinformatics, University of Oslo, Oslo, Norway
| | - Kevin O’Connell
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A. Shadrin
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav B. Smeland
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars T. Westlye
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - David E. J. Linden
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Dennis van der Meer
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
21
|
Chen Y, Dang M, Zhang Z. Brain mechanisms underlying neuropsychiatric symptoms in Alzheimer's disease: a systematic review of symptom-general and -specific lesion patterns. Mol Neurodegener 2021; 16:38. [PMID: 34099005 PMCID: PMC8186099 DOI: 10.1186/s13024-021-00456-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/11/2021] [Indexed: 12/16/2022] Open
Abstract
Neuropsychiatric symptoms (NPSs) are common in patients with Alzheimer's disease (AD) and are associated with accelerated cognitive impairment and earlier deaths. This review aims to explore the neural pathogenesis of NPSs in AD and its association with the progression of AD. We first provide a literature overview on the onset times of NPSs. Different NPSs occur in different disease stages of AD, but most symptoms appear in the preclinical AD or mild cognitive impairment stage and develop progressively. Next, we describe symptom-general and -specific patterns of brain lesions. Generally, the anterior cingulate cortex is a commonly damaged region across all symptoms, and the prefrontal cortex, especially the orbitofrontal cortex, is also a critical region associated with most NPSs. In contrast, the anterior cingulate-subcortical circuit is specifically related to apathy in AD, the frontal-limbic circuit is related to depression, and the amygdala circuit is related to anxiety. Finally, we elucidate the associations between the NPSs and AD by combining the onset time with the neural basis of NPSs.
Collapse
Affiliation(s)
- Yaojing Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875 China
- BABRI Centre, Beijing Normal University, Beijing, 100875 China
| | - Mingxi Dang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875 China
- BABRI Centre, Beijing Normal University, Beijing, 100875 China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875 China
- BABRI Centre, Beijing Normal University, Beijing, 100875 China
| |
Collapse
|
22
|
Cai LY, Kerley CI, Yu C, Aboud KS, Beason-Held LL, Shafer AT, Resnick SM, Jordan LC, Anderson AW, Schilling KG, Lyu I, Landman BA. Joint cortical surface and structural connectivity analysis of Alzheimer's Disease. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2021; 11596:1159630. [PMID: 34354323 PMCID: PMC8336655 DOI: 10.1117/12.2580956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prior neuroimaging studies have demonstrated isolated structural and connectivity changes in the brain due to Alzheimer's Disease (AD). However, how these changes relate to each other is not well understood. We present a preliminary study to begin to fill this gap by leveraging joint independent component analysis (jICA). We explore how jICA performs in an analysis of T1 and diffusion weighted MRI by characterizing the joint changes of complex cortical surface and structural connectivity metrics in AD in subjects from the Baltimore Longitudinal Study of Aging. We calculate 588 region-based cortical metrics and 4,753 fractional anisotropy-based connectivity metrics and project them into a low-dimensional manifold with principal component analysis. We perform jICA on the manifold and subsequently backproject the independent components to the original data space. We demonstrate component stability with 3-fold cross validation and find differential component loadings between 776 cognitively unimpaired control subjects and 23 with AD that generalizes across folds. In addition, we perform the same analysis on the surface and connectivity metrics separately and find that the joint approach identifies both novel and similar components to the separate approaches. To illustrate the joint approach's primary utility, we provide an example hypothesis for how surface and connectivity components may vary together with AD. These preliminary results suggest jointly varying independent cortical surface and structural connectivity components can be consistently extracted from MRI data and provide a data-driven way for generating novel hypotheses about AD that may not be captured by separate analyses.
Collapse
Affiliation(s)
- Leon Y Cai
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Cailey I Kerley
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Chang Yu
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Katherine S Aboud
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Lori L Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Andrea T Shafer
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Lori C Jordan
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam W Anderson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
| | - Kurt G Schilling
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
| | - Ilwoo Lyu
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Bennett A Landman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
23
|
Li R, Zhang Y, Zhuo Z, Wang Y, Jia Z, Sun M, Zhang Y, Li W, Duan Y, Yao Z, Weng H, Wei J, Liu Y, Xu J. Altered Cerebral Blood Flow in Alzheimer's Disease With Depression. Front Psychiatry 2021; 12:687739. [PMID: 34305683 PMCID: PMC8295555 DOI: 10.3389/fpsyt.2021.687739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Depression is common in Alzheimer's disease (AD) with an unclear neural mechanism. This study aimed to investigate the underlying cerebral perfusion associated with depression in AD and evaluate its clinical significance. Method: Twenty-one AD patients and 21 healthy controls (HCs) were enrolled in this study. The depressive symptom was defined according to the Hamilton Depression Rating Scale (HAMD). Nine patients were diagnosed as AD with depression symptoms (HAMD >7). Three-dimensional pseudocontinuous arterial spin labeling MR imaging was conducted to measure regional cerebral blood flow (CBF). Neuropsychological tests covered cognition and depressive scores. Between-group comparisons on clinical variables and regional CBFs, relationship between regional CBF and depressive score, and identification of AD patients with depression were performed using covariance analysis, linear regression, and receiver operating characteristic (ROC) analysis, respectively. Results: Compared with HCs, AD patients without depression exhibited lower gray matter CBF (p = 0.016); compared with AD patients without depression, AD patients with depression had higher CBF in the right supplementary motor area (39.23 vs. 47.91 ml/100 g/min, p = 0.017) and right supramarginal gyrus (35.54 vs. 43.85 ml/100 g/min, p = 0.034). CBF in the right supplementary motor area was correlated with depressive score (β = 0.46, p = 0.025). The combination of CBF in the right supplementary motor area and supramarginal gyrus and age could identify AD patients with depression from those without depression with a specificity of 100%, sensitivity of 66.67%, accuracy of 85.71%, and area under the curve of 0.87. Conclusions: Our findings suggested that hyperperfusion of the right supplementary motor area and right supramarginal gyrus were associated with depression syndrome in AD, which could provide a potential neuroimaging marker to evaluate the depression state in AD.
Collapse
Affiliation(s)
- Runzhi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanling Zhang
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhizheng Zhuo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanli Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ziyan Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunyun Duan
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zeshan Yao
- AnImage Technology (Beijing) Co., Ltd., Beijing, China
| | - Haoyi Weng
- Bioinformatics Department, Shenzhen WeGene Clinical Laboratory, Shenzhen, China
| | - Juan Wei
- GE Healthcare, MR Research China, Beijing, China
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Cognitive Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Loureiro JC, Stella F, Pais MV, Radanovic M, Canineu PR, Joaquim HPG, Talib LL, Forlenza OV. Cognitive impairment in remitted late-life depression is not associated with Alzheimer's disease-related CSF biomarkers. J Affect Disord 2020; 272:409-416. [PMID: 32553384 DOI: 10.1016/j.jad.2020.03.166] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/23/2020] [Accepted: 03/29/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Cognitive impairment is a common feature of late-life depression (LLD). Early studies using Alzheimer's disease (AD) biomarkers inferred a biological link between AD pathology and LLD, but recent findings have challenged this association. The aim of this investigation was to determine a panel of AD-related cerebrospinal fluid (CSF) biomarkers in a cross-section of elders with mild cognitive impairment (MCI) with and without LLD. METHODS Subjects comprised 102 older adults: 27 with 'pure' amnestic MCI (aMCI), 53 with major depression and cognitive impairment - encompassing 22 late-onset (LOD) and 31 early-onset depression (EOD), and 22 euthymic elders without cognitive impairment (controls). Participants underwent lumbar puncture for determination of CSF concentrations of Aβ1-42, T-tau, and P-tau. Cut-off scores for suspected AD were: Aβ1-42 < 416p g/mL, P-tau > 36.1 pg/mL and Aβ/P-tau ratio < 9.53 (O. V. Forlenza et al. 2015). Statistical analyses consisted of analyses of variance (ANOVA), analyses of covariance (ANCOVA), Bonferroni post-hoc tests, and Pearson's chi-squared tests. RESULTS ANCOVA (age and schooling as covariates) displayed statistically significant results with respect to CSF biomarkers' profiles regardless of the socio-demographic divergencies previously identified by one-way ANOVA. Mean Aβ1-42 values (pg/mL) were: aMCI, 360.3 (p < 0.001); LOD, 486.6 (p < 0.001); EOD, 494.2 (p < 0.001); controls, 528.3 (p < 0.001); p< 0.05. Mean Aβ1-42/P-tau ratio: aMCI, 7.9 (p < 0.001); LOD 14.2 (p < 0.001); EOD, 15.3 (p < 0.001); controls, 17.1 (p < 0.001); p < 0.05. Post-hoc tests indicated that patients with aMCI showed significant differences in biomarker profile compatible with AD signature. LIMITATION The main limitation is the relatively small sample. CONCLUSION Our findings suggest that, distinctively from aMCI, cognitive impairment in LLD is not associated with AD's CSF pathological signature.
Collapse
Affiliation(s)
- Júlia C Loureiro
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil.
| | - Florindo Stella
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil; UNESP- Universidade Estadual Paulista, Instituto de Biociências, Rio Claro, SP, Brasil
| | - Marcos V Pais
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Marcia Radanovic
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Paulo R Canineu
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil; Programa de Gerontologia, Pontifícia Universidade Católica de São Paulo, São Paulo, SP, Brasil
| | - Helena P G Joaquim
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Leda L Talib
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Orestes V Forlenza
- Laboratorio de Neurociencias LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| |
Collapse
|
25
|
Wu Y, Wu X, Wei Q, Wang K, Tian Y. Differences in Cerebral Structure Associated With Depressive Symptoms in the Elderly With Alzheimer's Disease. Front Aging Neurosci 2020; 12:107. [PMID: 32477094 PMCID: PMC7236549 DOI: 10.3389/fnagi.2020.00107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/30/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Alzheimer's disease (AD) is characterized by global deterioration in multiple cognitive domains. In addition to cognitive impairment, depressive symptoms are common issues that trouble AD patients. The neuroanatomical basis of depressive symptoms in AD patients has yet to be elucidated. Method: Twenty AD patients and 22 healthy controls (HCs) were recruited for the present study. Depressive symptoms in AD patients and HCs were assessed according to the Hamilton Depression Rating Scale (HDRS). Anatomical structural differences were assessed between AD patients and HCs using voxel-based morphometry (VBM) and surface-based morphometry (SBM). Correlation analyses were conducted to investigate relationships between depressive symptoms and structural altered regions. Multiple pattern analysis using linear support vector machine (SVM) was performed in another independent cohort, which was collected from Alzheimer's Disease Neuroimaging Initiative (ADNI) data and contained 20 AD patients and 20 HCs, to distinguish AD patients from HCs. Results: Compared with HCs, AD patients exhibited global cerebral atrophy in gray matter volume (GMV) and cortical thickness, including frontal, parietal, temporal, occipital, and insular lobes. In addition, insular GMV was negatively correlated with depressive symptoms. Moreover, SVM-based classification achieved an accuracy of 77.5%, a sensitivity of 70%, and a specificity of 85% by leave-one-out cross-validation. Conclusion: GMV of the insula displayed atrophy among AD patients, which is associated with depressive symptoms. Our observations provide a potential neural substrate for analysis to examine the co-occurrence of AD with depressive symptoms.
Collapse
Affiliation(s)
- Yue Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
| | - Qiang Wei
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
- Department of Medical Psychology, Chaohu Clinical Medical College, Anhui Medical University, Hefei, China
| | - Yanghua Tian
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| |
Collapse
|
26
|
Brzezińska A, Bourke J, Rivera-Hernández R, Tsolaki M, Woźniak J, Kaźmierski J. Depression in Dementia or Dementia in Depression? Systematic Review of Studies and Hypotheses. Curr Alzheimer Res 2020; 17:16-28. [DOI: 10.2174/1567205017666200217104114] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/09/2020] [Accepted: 01/18/2020] [Indexed: 01/21/2023]
Abstract
The majority of research works to date suggest that Major Depressive Disorder (MDD) is a
risk factor for dementia and may predispose to cognitive decline in both early and late onset variants.
The presence of depression may not, however, reflect the cause, rather, an effect: it may be a response to
cognitive impairment or alters the threshold at which cognitive impairment might manifest or be detected.
An alternative hypothesis is that depression may be part of a prodrome to Alzheimer’s Disease
(AD), suggesting a neurobiological association rather than one of psychological response alone. Genetic
polymorphisms may explain some of the variances in shared phenomenology between the diagnoses, the
instance, when the conditions arise comorbidly, the order in which they are detected that may depend on
individual cognitive and physical reserves, as well as the medical history and individual vulnerability.
This hypothesis is biologically sound but has not been systematically investigated to date. The current
review highlights how genetic variations are involved in the development of both AD and MDD, and the
risk conferred by these variations on the expression of these two disorders comorbidly is an important
consideration for future studies of pathoaetiological mechanisms and in the stratification of study samples
for randomised controlled trials.
Collapse
Affiliation(s)
- Agnieszka Brzezińska
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - Julius Bourke
- Centre for Psychiatry, Wolfson Institute for Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London E14NS, United Kingdom
| | - Rayito Rivera-Hernández
- Department of Psychiatry, Psychology, Legal Medicine and History of Medicine, University of Salamanca, Salamanca, Spain
| | - Magda Tsolaki
- 3rd Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece, “George Papanicolaou” Hospital, Thessaloniki, Greece
| | - Joanna Woźniak
- Central Clinical Hospital of Medical University of Lodz, Lodz, Poland
| | - Jakub Kaźmierski
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
27
|
Clement A, Wiborg O, Asuni AA. Steps Towards Developing Effective Treatments for Neuropsychiatric Disturbances in Alzheimer's Disease: Insights From Preclinical Models, Clinical Data, and Future Directions. Front Aging Neurosci 2020; 12:56. [PMID: 32210790 PMCID: PMC7068814 DOI: 10.3389/fnagi.2020.00056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/18/2020] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide. It is mostly known for its devastating effect on memory and learning but behavioral alterations commonly known as neuropsychiatric disturbances (NPDs) are also characteristics of the disease. These include apathy, depression-like behavior, and sleep disturbances, and they all contribute to an increased caregiver burden and earlier institutionalization. The interaction between NPDs and AD pathology is not well understood, but the consensus is that they contribute to disease progression and faster decline. Consequently, recognizing and treating NPDs might improve AD pathology and increase the quality of life for both patients and caregivers. In this review article, we examine previous and current literature on apathy, depressive symptoms, and sleep disturbances in AD patients and preclinical AD mechanistic models. We hypothesize that tau accumulation, beta-amyloid (Aβ) aggregation, neuroinflammation, mitochondrial damage, and loss of the locus coeruleus (LC)-norepinephrine (NE) system all collectively impact the development of NPDs and contribute synergistically to AD pathology. Targeting more than one of these processes might provide the most optimal strategy for treating NPDs and AD. The development of such clinical approaches would be preceded by preclinical studies, for which robust and reliable mechanistic models of NPD-like behavior are needed. Thus, developing effective preclinical research models represents an important step towards a better understanding of NPDs in AD.
Collapse
Affiliation(s)
- Amalie Clement
- Laboratory of Neurobiology, Department of Health, Science and Technology, Aalborg University, Aalborg, Denmark
- Department of Physiology and Symptoms, H. Lundbeck A/S, Copenhagen, Denmark
| | - Ove Wiborg
- Laboratory of Neurobiology, Department of Health, Science and Technology, Aalborg University, Aalborg, Denmark
| | - Ayodeji A. Asuni
- Department of Physiology and Symptoms, H. Lundbeck A/S, Copenhagen, Denmark
| |
Collapse
|
28
|
Correlation between potentially inappropriate medication and Alzheimer’s disease among the elderly. Arch Gerontol Geriatr 2020; 87:103842. [DOI: 10.1016/j.archger.2019.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/22/2019] [Accepted: 03/11/2019] [Indexed: 02/05/2023]
|
29
|
Verdaguer ES, Stafford J, Tuijt R, Orgeta V. Minor and subthreshold depressive disorders in Alzheimer's disease: A systematic review and meta-analysis of prevalence studies. J Affect Disord 2020; 263:728-734. [PMID: 31787425 DOI: 10.1016/j.jad.2019.11.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 10/02/2019] [Accepted: 11/10/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Depressive symptoms are common in Alzheimer's disease (AD) and negatively impact patient well-being. The main aim of the present study was to establish summary estimates for the prevalence of minor depressive disorder (MinD) and subthreshold depression in AD and synthesise evidence on prognosis and management of these symptoms in order to inform clinical guidelines. METHODS Systematic review and meta-analysis of cross-sectional and longitudinal studies of prevalence, prognosis, and treatments for minor and subthreshold depression in AD. We searched MEDLINE, Embase, PsycINFO and CINAHL. We included studies that reported prevalence of subthreshold depressive disorders and those reporting data on validity of diagnostic criteria, mechanisms, or randomised controlled clinical trials (RCTs) testing effectiveness of interventions. Estimates of prevalence were pooled using random-effects meta-analyses. Two authors screened articles and independently extracted data on study characteristics. RESULTS We reviewed 5671 abstracts, retrieved 621 full text articles and included a total of 15 studies. Pooling data from 10 studies showed that prevalence for MinD in AD was 22.0% (95% CI 16.0 to 28.0). Prevalence for a clinical diagnosis of MinD (DSM-III-R and DSM-IV) was 26.0% (95% CI 20.0 to 32.0; 6 studies). People with MinD experienced higher levels of neuropsychiatric symptoms, functional and cognitive decline, although studies remain cross-sectional. Neither sertraline nor a carer intervention were effective in reducing symptoms. CONCLUSION This review finds that MinD is prevalent in people with a diagnosis of AD and requires clinical attention. Research is warranted to develop effective interventions to treat and prevent these symptoms.
Collapse
|
30
|
Banning LCP, Ramakers IHGB, Deckers K, Verhey FRJ, Aalten P. Affective symptoms and AT(N) biomarkers in mild cognitive impairment and Alzheimer's disease: A systematic literature review. Neurosci Biobehav Rev 2019; 107:346-359. [PMID: 31525387 DOI: 10.1016/j.neubiorev.2019.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) biomarkers such as amyloid, p-tau and neuronal injury markers have been associated with affective symptoms in cognitively impaired individuals, but results are conflicting. METHODS CINAHL, Embase, PsycINFO and PubMed were searched for studies evaluating AD biomarkers with affective symptoms in mild cognitive impairment and AD dementia. Studies were classified according to AT(N) research criteria. RESULT Forty-five abstracts fulfilled eligibility criteria, including in total 8,293 patients (41 cross-sectional studies and 7 longitudinal studies). Depression and night-time behaviour disturbances were not related to AT(N) markers. Apathy was associated with A markers (PET, not CSF). Mixed findings were reported for the association between apathy and T(N) markers; anxiety and AT(N) markers; and between agitation and irritability and A markers. Agitation and irritability were not associated with T(N) markers. DISCUSSION Whereas some AD biomarkers showed to be associated with affective symptoms in AD, most evidence was inconsistent. This is likely due to differences in study design or heterogeneity in affective symptoms. Directions for future research are given.
Collapse
Affiliation(s)
- Leonie C P Banning
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Inez H G B Ramakers
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Kay Deckers
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Frans R J Verhey
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| | - Pauline Aalten
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands.
| |
Collapse
|
31
|
Capogna E, Manca R, De Marco M, Hall A, Soininen H, Venneri A. Understanding the effect of cognitive/brain reserve and depression on regional atrophy in early Alzheimer's disease. Postgrad Med 2019; 131:533-538. [PMID: 31478419 DOI: 10.1080/00325481.2019.1663127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Introduction: Depression in patients with mild cognitive impairment (MCI) and dementia of the Alzheimer's type (AD) is associated with worse prognosis. Indeed, depressed MCI patients have worse cognitive performance and greater loss of gray-matter volume in several brain areas. To date, knowledge of the factors that can mitigate this detrimental effect is still limited. The aim of the present study was to understand in what way cognitive reserve/brain reserve and depression interact and are linked to regional atrophy in early stage AD. Methods: Depression was evaluated with the Patient Health Questionnaire-9 in 90 patients with early AD, and a cutoff of ≥ 5 was used to separate depressed (n = 44) from non-depressed (n = 46) patients. Each group was further stratified into high/low cognitive reserve/brain reserve. Cognitive reserve was calculated using years of education as proxy, while normalized parenchymal volumes were used to estimate brain reserve. Voxel-based morphometry was carried out to extract and analyze gray-matter maps. 2 × 2 ANCOVAs were run to test the effect of the reserve-by-depression interaction on gray matter. Age and hippocampal ratio were used as covariates. Composite indices of major cognitive domains were also analyzed with comparable models. Results: No reserve-by-depression interaction was found in the analytical models of gray matter. Depression was associated with less gray matter volume in the cerebellum and parahippocampal gyrus. The brain reserve-by-depression interaction was a significant predictor of executive functioning. Among those with high brain reserve, depressed patients had poorer executive skills. No significant results were found in association with cognitive reserve. Conclusion: These findings suggest that brain reserve may modulate the association between neurodegeneration and depression in patients with MCI and dementia of the AD type, influencing in particular executive functioning.
Collapse
Affiliation(s)
- Elettra Capogna
- Department of Neuroscience, University of Sheffield , Sheffield , UK
| | - Riccardo Manca
- Department of Neuroscience, University of Sheffield , Sheffield , UK
| | - Matteo De Marco
- Department of Neuroscience, University of Sheffield , Sheffield , UK
| | - Anette Hall
- Institute of Clinical Medicine, Neurology, University of Eastern Finland , Kuopio , Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland , Kuopio , Finland
| | - Annalena Venneri
- Department of Neuroscience, University of Sheffield , Sheffield , UK
| |
Collapse
|
32
|
The power of sample size through a multi-scanner approach in MR neuroimaging regression analysis: evidence from Alzheimer's disease with and without depression. AUSTRALASIAN PHYSICAL & ENGINEERING SCIENCES IN MEDICINE 2019; 42:563-571. [PMID: 31054027 DOI: 10.1007/s13246-019-00758-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 04/27/2019] [Indexed: 10/26/2022]
Abstract
The inconsistency of volumetric results often seen in MR neuroimaging studies can be partially attributed to small sample sizes and variable data analysis approaches. Increased sample size through multi-scanner studies can tackle the former, but combining data across different scanner platforms and field-strengths may introduce a variability factor capable of masking subtle statistical differences. To investigate the sample size effect on regression analysis between depressive symptoms and grey matter volume (GMV) loss in Alzheimer's disease (AD), a retrospective multi-scanner investigation was conducted. A cohort of 172 AD patients, with or without comorbid depressive symptoms, was studied. Patients were scanned with different imaging protocols in four different MRI scanners operating at either 1.5 T or 3.0 T. Acquired data were uniformly analyzed using the computational anatomy toolbox (CAT12) of the statistical parametric mapping (SPM12) software. Single- and multi-scanner regression analyses were applied to identify the anatomical pattern of correlation between GM loss and depression severity. A common anatomical pattern of correlation between GMV loss and increased depression severity, mostly involving sensorimotor areas, was identified in all patient subgroups imaged in different scanners. Analysis of the pooled multi-scanner data confirmed the above finding employing a more conservative statistical criterion. In the retrospective multi-scanner setting, a significant correlation was also exhibited for temporal and frontal areas. Increasing the sample size by retrospectively pooling multi-scanner data, irrespective of the acquisition platform and parameters employed, can facilitate the identification of anatomical areas with a strong correlation between GMV changes and depression symptoms in AD patients.
Collapse
|
33
|
Association of depression with malnutrition, grip strength and impaired cognitive function among senior trauma patients. J Affect Disord 2019; 247:175-182. [PMID: 30684891 DOI: 10.1016/j.jad.2019.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/17/2018] [Accepted: 01/13/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Depression is common among senior adults, yet understudied among trauma patients. The purpose of this study was to assess the prevalence of depressive symptoms among seniors hospitalized in acute trauma care, to compare patients with depressive symptoms vs. those without, and to evaluate whether depression symptoms affects discharge destination. METHODS This cross-sectional and prospective analysis was conducted among community-dwelling patients ≥70 years old, hospitalized at the Senior Trauma Center of the University Hospital Zurich, Switzerland. We used the Geriatric Depression Scale (GDS-15) to assess presence of depressive symptoms. Using a cutoff value of 5 points, we compared age- and gender-adjusted characteristics of patients with and without depressive symptoms. Multinomial logistic regression models were used to estimate the odds of returning home vs. not adjusting for age, gender, nutritional status, cognitive function and others. RESULTS Of the 273 seniors enrolled, 104 (38.1%) were men and the mean age was 79.4 (SD = 6.5) years. We identified 52 (19.0%) patients with depressive symptoms. These patients were more likely to be older (p = 0.04), at risk for malnutrition (p<0.0001), at least pre-frail (p = 0.005), and have decreased cognitive function (p = 0.001). They were also more than twice as likely to be discharged to acute geriatric care compared to home (OR = 2.28 (CI = 1.12-4.68)). LIMITATIONS Depressive symptoms were assessed during acute care without data before hospitalization. CONCLUSIONS Senior trauma patients with depressive symptoms during acute care were more likely to be at higher risk of malnutrition, have cognitive decline and are more likely to receive additional geriatric care.
Collapse
|
34
|
Avedisova AS, Samotaeva IS, Luzin RV, Semenovyh NS, Sergunova KA, Akzhigitov RG, Zakharova RV. Apathy in depression: a morphometric analysis. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:141-147. [DOI: 10.17116/jnevro2019119051141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
35
|
Kuring JK, Mathias JL, Ward L. Prevalence of Depression, Anxiety and PTSD in People with Dementia: a Systematic Review and Meta-Analysis. Neuropsychol Rev 2018; 28:393-416. [PMID: 30536144 DOI: 10.1007/s11065-018-9396-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022]
Abstract
There appears to be a link between depression/anxiety/PTSD and dementia, although the evidence is incomplete and the reason is unclear. Mental illness may cause dementia or may be prodromal or comorbid with dementia, or dementia may trigger a relapse of symptoms in individuals with a history of mental illness. This study examined the link between depression/anxiety/PTSD and dementia by evaluating the prevalence of these disorders in people with dementia, relative to their healthy peers. Existing meta-analyses have examined the prevalence of clinically-significant depression and anxiety in Alzheimer's disease (AD), and depression in frontotemporal dementia (FTD), but have not considered vascular dementia (VaD), dementia with Lewy bodies (DLB), PTSD, or anxiety in FTD. The current meta-analysis compared the prevalence of clinically-significant depression, anxiety and PTSD in the four most common types of dementia (AD, VaD, DLB, FTD) and in unspecified dementia to that of healthy controls (PROSPERO number: CRD42017082086). PubMed, EMBASE, PsycINFO and CINAHL database searches identified 120 eligible studies. Prevalence rates were calculated for depression and anxiety in AD, VaD, DLB, FTD, unspecified dementia, and controls. PTSD data were only available for unspecified dementia. Subgroup analyses indicated that depression, but not anxiety, was more prevalent in people with dementia compared to controls; however, the anxiety analyses were probably under-powered. The results support a link between depression and dementia; however, the link between anxiety or PTSD and dementia remains unclear due to insufficient data. Longitudinal data is now needed to clarify whether depression/anxiety/PTSD may be risk factors for dementia.
Collapse
Affiliation(s)
- J K Kuring
- School of Psychology, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - J L Mathias
- School of Psychology, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia.
| | - L Ward
- School of Psychology, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| |
Collapse
|
36
|
Alterations in structural rich-club connectivity of the precuneus are associated with depressive symptoms among individuals with subjective memory complaints. COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2018; 19:73-87. [PMID: 30298425 DOI: 10.3758/s13415-018-0645-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The association between subjective memory complaints (SMCs) and depressive symptoms has been widely reported and both have been regarded as risk factors for dementia, such as Alzheimer's disease (AD). Although SMCs arise as early as in middle age, the exact neural correlates of comorbid depressive symptoms among individuals who are middle-aged and with SMCs have not yet been well investigated. Because rich-club organization of the brain plays a key role in the pathophysiology of various neuropsychiatric disorders, the investigation of rich club organization may provide insight regarding the neurobiological mechanisms of depressive symptoms in SMCs. In the current study, we compared the rich-club organization in the structural brain connectivity between individuals who have SMCs along with depressive symptoms (SMCD) and individuals with SMCs but without depressive symptoms (SMCO). A total of 53 individuals with SMCD and 91 individuals with SMCO participated in the study. For all participants, high-resolution, T1-weighted images and diffusion tensor images were obtained, and the network analysis was performed. Individuals with SMCD had lower connectivity strength between the precuneus and other rich-club nodes than those with SMCO, which was significant after adjusting for potential confounders. Our findings suggest that disruptions of rich-club connectivity strength of the precuenus are associated with depressive symptoms in middle-aged individuals with SMCs. Given that the precuneus is one of the commonly affected regions in the early stages of AD, our findings may imply that the concomitant depressive symptoms in middle-aged individuals with SMCs could reflect structural alterations related to AD.
Collapse
|
37
|
Brendel M, Sauerbeck J, Greven S, Kotz S, Scheiwein F, Blautzik J, Delker A, Pogarell O, Ishii K, Bartenstein P, Rominger A. Serotonin Selective Reuptake Inhibitor Treatment Improves Cognition and Grey Matter Atrophy but not Amyloid Burden During Two-Year Follow-Up in Mild Cognitive Impairment and Alzheimer’s Disease Patients with Depressive Symptoms. J Alzheimers Dis 2018; 65:793-806. [DOI: 10.3233/jad-170387] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
| | - Julia Sauerbeck
- Department of Nuclear Medicine, University of Munich, Germany
| | - Sonja Greven
- Department of Statistics, University of Munich, Germany
| | - Sebastian Kotz
- Department of Nuclear Medicine, University of Munich, Germany
| | | | | | - Andreas Delker
- Department of Nuclear Medicine, University of Munich, Germany
| | | | - Kazunari Ishii
- Department of Radiology, Kindai University Faculty of Medicine, Osakasayama City, Osaka, Japan
| | | | - Axel Rominger
- Department of Nuclear Medicine, University of Munich, Germany
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Switzerland
| | | |
Collapse
|
38
|
Moon B, Kim S, Park YH, Lim JS, Youn YC, Kim S, Jang JW. Depressive Symptoms are Associated with Progression to Dementia in Patients with Amyloid-Positive Mild Cognitive Impairment. J Alzheimers Dis 2018; 58:1255-1264. [PMID: 28550264 DOI: 10.3233/jad-170225] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Depressive symptoms are prevalent in patients with mild cognitive impairment (MCI) and are considered to be a risk factor for progression to dementia. OBJECTIVE The purpose of this study was to evaluate whether depressive symptoms in MCI promote disease progression in a manner related to amyloid status, and to determine the relationship between depressive symptoms and longitudinal cerebral structural changes. METHODS Baseline data for 336 patients with MCI (75 with depression and 261 without) from the Alzheimer's Disease Neuroimaging Initiative study were analyzed. All participants underwent comprehensive cognitive testing, volumetric magnetic resonance imaging (MRI), and [18F]AV45 positron emission tomography amyloid imaging. Depressive symptoms were measured using the Neuropsychiatric Inventory Questionnaire. A voxel-based morphometric analysis using volumetric brain MRI data was used to compare longitudinal structural changes related to depressive symptoms. RESULTS The conversion rate to dementia was different between patients with and without depression in amyloid-positive MCI (40.8% versus 19.7%, respectively; p = 0.006). Patients who were amyloid-positive at baseline also exhibited a greater degree of 2-year cognitive decline. Depression in amyloid-positive MCI was associated with longitudinal cortical atrophy in the left cingulate gyrus. CONCLUSION Our study indicates that the presence of depressive symptoms in patients with amyloid-positive MCI is associated with higher progression to dementia and longitudinal cortical atrophy.
Collapse
Affiliation(s)
- Byungseung Moon
- College of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Seongheon Kim
- College of Medicine, Kangwon National University, Chuncheon, Republic of Korea.,Department of Neurology, Kangwon National University Hospital, Chuncheon, Republic of Korea
| | - Young Ho Park
- Clinical Neuroscience Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Department of Neurology, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Sung Lim
- Department of Neurology, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - SangYun Kim
- Clinical Neuroscience Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Department of Neurology, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Won Jang
- College of Medicine, Kangwon National University, Chuncheon, Republic of Korea.,Department of Neurology, Kangwon National University Hospital, Chuncheon, Republic of Korea
| | | |
Collapse
|
39
|
Brown EE, Iwata Y, Chung JK, Gerretsen P, Graff-Guerrero A. Tau in Late-Life Depression: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2018; 54:615-33. [PMID: 27497481 DOI: 10.3233/jad-160401] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A lifetime history of major depressive disorder (MDD) increases the risk of developing Alzheimer's disease, of which neurofibrillary tangles due to abnormal tau proteins are a hallmark. We systematically reviewed the literature on tau in MDD and identified 49 relevant articles spanning a number of modalities, including cerebrospinal fluid (CSF) analysis, positron emission tomography, and clinicopathological correlation. We compared CSF total and phosphorylated tau proteins in MDD and controls using a meta-analytic approach. We found no difference in total or phosphorylated tau in MDD. We also found no difference in a comparison of a subgroup excluding studies with significant age differences. Positron emission tomography studies lacked specificity. Clinicopathological studies failed to associate neurofibrillary tangles with MDD. The available data on tau in MDD is limited. The involvement of tau in a subset of MDD cannot be ruled out and requires prospective exploration.
Collapse
Affiliation(s)
- Eric E Brown
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Multimodal Imaging Group-Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Yusuke Iwata
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Multimodal Imaging Group-Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jun Ku Chung
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Multimodal Imaging Group-Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Philip Gerretsen
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Multimodal Imaging Group-Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Ariel Graff-Guerrero
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Multimodal Imaging Group-Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
40
|
Zhu Q, Lin M, Bi S, Ni Z, Zhao J, Chen B, Fan G, Shang X. Impaired Frontolimbic Connectivity and Depressive Symptoms in Patients with Alzheimer's Disease. Dement Geriatr Cogn Disord 2018; 41:281-91. [PMID: 27331920 DOI: 10.1159/000447056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Depressive symptoms are commonly observed in Alzheimer's disease (AD). The underlying mechanisms of depressive symptoms in AD remain unclear; frontolimbic circuitry dysfunction may play a role. We aimed to investigate the microstructural integrity of frontolimbic connectivity of specific fiber tracts in AD patients with and without depressive symptoms using diffusion tensor imaging (DTI). METHODS Eleven AD patients with depressive symptoms (dep-AD), 18 AD patients without depressive symptoms (nondep-AD), and 18 normal control (NC) subjects were included. The cingulum bundle (CB), uncinate fasciculus (UF), and fornix, mainly frontolimbic connectivity, were measured by DTI tractography and the metrics of fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity, and radial diffusivity (RD) were calculated. RESULTS Compared with NC subjects, both dep-AD and nondep-AD patients showed significant differences for all indices in the fornix and significantly decreased FA and increased MD and RD in the bilateral CB and UF. When compared to nondep-AD patients, dep-AD patients showed significantly increased MD and RD in the bilateral CB and right UF. CONCLUSION Depressive symptoms in AD patients may be involved in greater microstructural abnormalities of frontolimbic connectivity and myelin injury in the bilateral CB and right UF might contribute to the pathophysiology of depressive symptoms in AD.
Collapse
Affiliation(s)
- Qingyong Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sperling SA, Tsang S, Williams IC, Park MH, Helenius IM, Manning CA. Subjective Memory Change, Mood, and Cerebrovascular Risk Factors in Older African Americans. J Geriatr Psychiatry Neurol 2017; 30:324-330. [PMID: 28954594 PMCID: PMC5772652 DOI: 10.1177/0891988717732153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Subjective memory change (SMC) in older individuals may represent a harbinger of cognitive decline. This study examined the factors associated with SMC in older African Americans (AA), who have greater risk of developing dementia. We predicted that symptoms of depression and anxiety, as well as the total number of cerebrovascular risk factors (tCVRFs), but not performances on objective memory measures, would be positively associated with SMC. METHODS Ninety-six AA completed brief cognitive testing and answered questions about mood and memory at their primary care appointment. Vascular data were obtained from medical records. RESULTS Symptoms of depression and anxiety, but not performances on objective memory measures, were positively associated with SMC, t(χ2(1) = 16.55 and 12.94, respectively, both P < .001). In nondepressed participants, the tCVRF was important in distinguishing between those with and without SMC. CONCLUSIONS In older AA, symptoms of depression or anxiety were associated with SMC. In nondepressed AA, the tCVRFs were important in distinguishing between those with and without SMC.
Collapse
Affiliation(s)
- Scott A. Sperling
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Siny Tsang
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Ishan C. Williams
- Department of Nursing, University of Virginia, Charlottesville, VA, USA
| | - Moon Ho Park
- Department of Neurology, Korea University College of Medicine, Seoul, South Korea
| | - Ira M. Helenius
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Carol A. Manning
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
42
|
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green RC, Harvey D, Jack CR, Jagust W, Morris JC, Petersen RC, Saykin AJ, Shaw LM, Toga AW, Trojanowski JQ. Recent publications from the Alzheimer's Disease Neuroimaging Initiative: Reviewing progress toward improved AD clinical trials. Alzheimers Dement 2017; 13:e1-e85. [PMID: 28342697 DOI: 10.1016/j.jalz.2016.11.007] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The Alzheimer's Disease Neuroimaging Initiative (ADNI) has continued development and standardization of methodologies for biomarkers and has provided an increased depth and breadth of data available to qualified researchers. This review summarizes the over 400 publications using ADNI data during 2014 and 2015. METHODS We used standard searches to find publications using ADNI data. RESULTS (1) Structural and functional changes, including subtle changes to hippocampal shape and texture, atrophy in areas outside of hippocampus, and disruption to functional networks, are detectable in presymptomatic subjects before hippocampal atrophy; (2) In subjects with abnormal β-amyloid deposition (Aβ+), biomarkers become abnormal in the order predicted by the amyloid cascade hypothesis; (3) Cognitive decline is more closely linked to tau than Aβ deposition; (4) Cerebrovascular risk factors may interact with Aβ to increase white-matter (WM) abnormalities which may accelerate Alzheimer's disease (AD) progression in conjunction with tau abnormalities; (5) Different patterns of atrophy are associated with impairment of memory and executive function and may underlie psychiatric symptoms; (6) Structural, functional, and metabolic network connectivities are disrupted as AD progresses. Models of prion-like spreading of Aβ pathology along WM tracts predict known patterns of cortical Aβ deposition and declines in glucose metabolism; (7) New AD risk and protective gene loci have been identified using biologically informed approaches; (8) Cognitively normal and mild cognitive impairment (MCI) subjects are heterogeneous and include groups typified not only by "classic" AD pathology but also by normal biomarkers, accelerated decline, and suspected non-Alzheimer's pathology; (9) Selection of subjects at risk of imminent decline on the basis of one or more pathologies improves the power of clinical trials; (10) Sensitivity of cognitive outcome measures to early changes in cognition has been improved and surrogate outcome measures using longitudinal structural magnetic resonance imaging may further reduce clinical trial cost and duration; (11) Advances in machine learning techniques such as neural networks have improved diagnostic and prognostic accuracy especially in challenges involving MCI subjects; and (12) Network connectivity measures and genetic variants show promise in multimodal classification and some classifiers using single modalities are rivaling multimodal classifiers. DISCUSSION Taken together, these studies fundamentally deepen our understanding of AD progression and its underlying genetic basis, which in turn informs and improves clinical trial design.
Collapse
Affiliation(s)
- Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA.
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, CA, USA
| | - Laurel A Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert C Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | | | - William Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - John C Morris
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, CA, USA
| | | | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute of Neuroimaging and Informatics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Alzheimer's Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Udall Parkinson's Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
43
|
Zhang J, Guo Z, Liu X, Jia X, Li J, Li Y, Lv D, Chen W. Abnormal functional connectivity of the posterior cingulate cortex is associated with depressive symptoms in patients with Alzheimer's disease. Neuropsychiatr Dis Treat 2017; 13:2589-2598. [PMID: 29066900 PMCID: PMC5644530 DOI: 10.2147/ndt.s146077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Depressive symptoms are significant and very common psychiatric complications in patients with Alzheimer's disease (AD), which can aggravate the decline in social function. However, changes in the functional connectivity (FC) of the brain in AD patients with depressive symptoms (D-AD) remain unclear. OBJECTIVE To investigate whether any differences exist in the FC of the posterior cingulate cortex (PCC) between D-AD patients and non-depressed AD patients (nD-AD). MATERIALS AND METHODS We recruited 15 D-AD patients and 17 age-, sex-, educational level-, and Mini-Mental State Examination (MMSE)-matched nD-AD patients to undergo tests using the Neuropsychiatric Inventory, Hamilton Depression Rating Scale, and 3.0T resting-state functional magnetic resonance imaging. Bilateral PCC were selected as the regions of interest and between-group differences in the PCC FC network were assessed using Student's t-test. RESULTS Compared with the nD-AD group, D-AD patients showed increased PCC FC in the right amygdala, right parahippocampus, right superior temporal pole, right middle temporal lobe, right middle temporal pole, and right hippocampus (AlphaSim correction; P<0.05). In the nD-AD group, MMSE scores were positively correlated with PCC FC in the right superior temporal pole and right hippocampus (false discovery rate corrected; P<0.05). CONCLUSION Differences were detected in PCC FC between nD-AD and D-AD patients, which may be related to depressive symptoms. Our study provides a significant enhancement to our understanding of the functional mechanisms underlying D-AD.
Collapse
Affiliation(s)
- Jiangtao Zhang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and the Collaborative Innovation Center for Brain Science, Hangzhou, Zhejiang, China.,Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhongwei Guo
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiaozheng Liu
- China-USA Neuroimaging Research Institute & Department of Radiology, the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xize Jia
- Center for Cognitive Brain Disorders & Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou Normal University, Hangzhou, China
| | - Jiapeng Li
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yaoyao Li
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and the Collaborative Innovation Center for Brain Science, Hangzhou, Zhejiang, China.,Key Laboratory of Medical Neurobiology of Chinese Ministry of Health, Hangzhou, Zhejiang, China
| | - Danmei Lv
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and the Collaborative Innovation Center for Brain Science, Hangzhou, Zhejiang, China.,Key Laboratory of Medical Neurobiology of Chinese Ministry of Health, Hangzhou, Zhejiang, China
| | - Wei Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and the Collaborative Innovation Center for Brain Science, Hangzhou, Zhejiang, China.,Key Laboratory of Medical Neurobiology of Chinese Ministry of Health, Hangzhou, Zhejiang, China
| |
Collapse
|
44
|
Neurochemical correlation between major depressive disorder and neurodegenerative diseases. Life Sci 2016; 158:121-9. [DOI: 10.1016/j.lfs.2016.06.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/14/2016] [Accepted: 06/27/2016] [Indexed: 12/13/2022]
|
45
|
Targowska-Duda KM, Wnorowski A, Budzynska B, Jozwiak K, Biala G, Arias HR. The positive allosteric modulator of α7 nicotinic acetylcholine receptors, 3-furan-2-yl-N-p-tolyl-acrylamide, enhances memory processes and stimulates ERK1/2 phosphorylation in mice. Behav Brain Res 2016; 302:142-51. [DOI: 10.1016/j.bbr.2016.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/31/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023]
|
46
|
Dyke JP, Sondhi D, Voss HU, Yohay K, Hollmann C, Mancenido D, Kaminsky SM, Heier LA, Rudser KD, Kosofsky B, Casey BJ, Crystal RG, Ballon D. Brain Region-Specific Degeneration with Disease Progression in Late Infantile Neuronal Ceroid Lipofuscinosis (CLN2 Disease). AJNR Am J Neuroradiol 2016; 37:1160-9. [PMID: 26822727 DOI: 10.3174/ajnr.a4669] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 11/30/2015] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Late infantile neuronal ceroid lipofuscinosis (CLN2 disease) is a uniformly fatal lysosomal storage disease resulting from mutations in the CLN2 gene. Our hypothesis was that regional analysis of cortical brain degeneration may identify brain regions that are affected earliest and most severely by the disease. MATERIALS AND METHODS Fifty-two high-resolution 3T MR imaging datasets were prospectively acquired on 38 subjects with CLN2. A retrospective cohort of 52 disease-free children served as a control population. The FreeSurfer software suite was used for calculation of cortical thickness. RESULTS An increased rate of global cortical thinning in CLN2 versus control subjects was the primary finding in this study. Three distinct patterns were observed across brain regions. In the first, subjects with CLN2 exhibited differing rates of cortical thinning versus age. This was true in 22 and 26 of 34 regions in the left and right hemispheres, respectively, and was also clearly discernable when considering brain lobes as a whole and Brodmann regions. The second pattern exhibited a difference in thickness from healthy controls but with no discernable change with age (9 left hemispheres, 5 right hemispheres). In the third pattern, there was no difference in either the rate of cortical thinning or the mean cortical thickness between groups (3 left hemispheres, 3 right hemispheres). CONCLUSIONS This study demonstrates that CLN2 causes differential rates of degeneration across the brain. Anatomic and functional regions that degenerate sooner and more severely than others compared with those in healthy controls may offer targets for directed therapies. The information gained may also provide neurobiologic insights regarding the mechanisms underlying disease progression.
Collapse
Affiliation(s)
- J P Dyke
- From the Departments of Radiology (J.P.D., H.U.V., L.A.H., D.B.)
| | - D Sondhi
- Genetic Medicine (D.S., C.H., D.M., S.M.K., R.G.C., D.B.)
| | - H U Voss
- From the Departments of Radiology (J.P.D., H.U.V., L.A.H., D.B.)
| | | | - C Hollmann
- Genetic Medicine (D.S., C.H., D.M., S.M.K., R.G.C., D.B.)
| | - D Mancenido
- Genetic Medicine (D.S., C.H., D.M., S.M.K., R.G.C., D.B.)
| | - S M Kaminsky
- Genetic Medicine (D.S., C.H., D.M., S.M.K., R.G.C., D.B.)
| | - L A Heier
- From the Departments of Radiology (J.P.D., H.U.V., L.A.H., D.B.)
| | - K D Rudser
- Division of Biostatistics (K.D.R.), Clinical and Translational Science Institute, University of Minnesota, Minneapolis, Minnesota
| | | | - B J Casey
- Psychiatry (B.J.C.), Weill Cornell Medical College, New York, New York
| | - R G Crystal
- Genetic Medicine (D.S., C.H., D.M., S.M.K., R.G.C., D.B.)
| | - D Ballon
- From the Departments of Radiology (J.P.D., H.U.V., L.A.H., D.B.) Genetic Medicine (D.S., C.H., D.M., S.M.K., R.G.C., D.B.)
| |
Collapse
|
47
|
DeLorenzo C, Sovago J, Gardus J, Xu J, Yang J, Behrje R, Kumar JSD, Devanand DP, Pelton GH, Mathis CA, Mason NS, Gomez-Mancilla B, Aizenstein H, Mann JJ, Parsey RV. Characterization of brain mGluR5 binding in a pilot study of late-life major depressive disorder using positron emission tomography and [¹¹C]ABP688. Transl Psychiatry 2015; 5:e693. [PMID: 26645628 PMCID: PMC5068588 DOI: 10.1038/tp.2015.189] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/05/2015] [Accepted: 10/26/2015] [Indexed: 01/06/2023] Open
Abstract
The metabotropic glutamate receptor subtype 5 (mGluR5) has been implicated in the pathophysiology of mood and anxiety disorders and is a potential treatment target in major depressive disorder (MDD). This study compared brain mGluR5 binding in elderly patients suffering from MDD with that in elderly healthy volunteers using positron emission tomography (PET) and [(11)C]ABP688. Twenty elderly (mean age: 63.0 ± 6.3) subjects with MDD and twenty-two healthy volunteers in the same age range (mean age: 66.4 ± 7.3) were examined with PET after a single bolus injection of [(11)C]ABP688, with many receiving arterial sampling. PET images were analyzed on a region of interest and a voxel level to compare mGluR5 binding in the brain between the two groups. Differences in [(11)C]ABP688 binding between patients with early- and late-onset depression were also assessed. In contrast to a previously published report in a younger cohort, no significant difference in [(11)C]ABP688 binding was observed between elderly subjects with MDD and healthy volunteers. [(11)C]ABP688 binding was also similar between subgroups with early- or late-onset depression. We believe this is the first study to examine mGluR5 expression in depression in the elderly. Although future work is required, results suggest potential differences in the pathophysiology of elderly depression versus depression earlier in life.
Collapse
Affiliation(s)
- C DeLorenzo
- Department of Psychiatry, Columbia University, New York, NY, USA,Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA,Department of Psychiatry, Stony Brook University, HSC-T-10, Room 40D, Stony Brook, NY 11794, USA. E-mail:
| | - J Sovago
- Novartis Institute for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - J Gardus
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - J Xu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - J Yang
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - R Behrje
- Novartis Pharmaceuticals Corporations, East Hanover, NJ, USA
| | - J S D Kumar
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - D P Devanand
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - G H Pelton
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - C A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - N S Mason
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - B Gomez-Mancilla
- Novartis Institute for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - H Aizenstein
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - J J Mann
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - R V Parsey
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
48
|
Brendel M, Pogarell O, Xiong G, Delker A, Bartenstein P, Rominger A. Depressive symptoms accelerate cognitive decline in amyloid-positive MCI patients. Eur J Nucl Med Mol Imaging 2015; 42:716-24. [PMID: 25631614 PMCID: PMC5849231 DOI: 10.1007/s00259-014-2975-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Late-life depression even in subsyndromal stages is strongly associated with Alzheimer's disease (AD). Furthermore, brain amyloidosis is an early biomarker in subjects who subsequently suffer from AD and can be sensitively detected by amyloid PET. Therefore, we aimed to compare amyloid load and glucose metabolism in subsyndromally depressed subjects with mild cognitive impairment (MCI). METHODS [(18)F]AV45 PET, [(18)F]FDG PET and MRI were performed in 371 MCI subjects from the Alzheimer's Disease Neuroimaging Initiative Subjects were judged β-amyloid-positive (Aβ+; 206 patients) or β-amyloid-negative (Aβ-; 165 patients) according to [(18)F]AV45 PET. Depressive symptoms were assessed by the Neuropsychiatric Inventory Questionnaire depression item 4. Subjects with depressive symptoms (65 Aβ+, 41 Aβ-) were compared with their nondepressed counterparts. Conversion rates to AD were analysed (mean follow-up time 21.5 ± 9.1 months) with regard to coexisting depressive symptoms and brain amyloid load. RESULTS Aβ+ depressed subjects showed large clusters with a higher amyloid load in the frontotemporal and insular cortices (p < 0.001) with coincident hypermetabolism (p < 0.001) in the frontal cortices than nondepressed subjects. Faster progression to AD was observed in subjects with depressive symptoms (p < 0.005) and in Aβ+ subjects (p < 0.001). Coincident depressive symptoms additionally shortened the conversion time in all Aβ+ subjects (p < 0.005) and to a greater extent in those with a high amyloid load (p < 0.001). CONCLUSION Our results clearly indicate that Aβ+ MCI subjects with depressive symptoms have an elevated amyloid load together with relative hypermetabolism of connected brain areas compared with cognitively matched nondepressed individuals. MCI subjects with high amyloid load and coexistent depressive symptoms are at high risk of faster conversion to AD.
Collapse
Affiliation(s)
| | | | - Guoming Xiong
- Dept. of Nuclear Medicine, University of Munich, Germany
| | - Andreas Delker
- Dept. of Nuclear Medicine, University of Munich, Germany
| | | | - Axel Rominger
- Dept. of Nuclear Medicine, University of Munich, Germany
| |
Collapse
|
49
|
Auning E, Selnes P, Grambaite R, Šaltytė Benth J, Haram A, Løvli Stav A, Bjørnerud A, Hessen E, Hol PK, Muftuler løndalen A, Fladby T, Aarsland D. Neurobiological correlates of depressive symptoms in people with subjective and mild cognitive impairment. Acta Psychiatr Scand 2015; 131:139-47. [PMID: 25346330 DOI: 10.1111/acps.12352] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2014] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To test the hypothesis that depressive symptoms correlate with Alzheimer's disease (AD) type changes in CSF and structural and functional imaging including hippocampus volume, cortical thickness, white matter lesions, Diffusion tensor imaging (DTI), and fluoro-deoxy-glucose positron emission tomography (FDG-PET) in patient with subjective (SCI) and mild (MCI) cognitive impairment. METHOD In 60 patients, depressive symptoms were assessed using the Geriatric Depression Scale. The subjects underwent MRI, 18F-FDG PET imaging, and lumbar CSF extraction. RESULTS Subjects with depressive symptoms (n=24) did not have more pathological AD biomarkers than non-depressed. Uncorrected there were trends towards larger hippocampal volumes (P=0.06), less orbital WM damage measured by DTI (P=0.10), and higher orbital glucose metabolism (P=0.02) in the depressed group. The findings were similar when SCI and MCI were analyzed separately. Similarly, in patients with pathological CSF biomarkers (i.e., predementia AD, n=24), we found that correlations between scores on GDS and CSF Aß42 and P-tau indicated less severe AD-specific CSF changes with increasing depression. CONCLUSION Depressive symptoms are common in SCI/MCI, but are not associated with pathological imaging or CSF biomarkers of AD. Depression can explain cognitive impairment in SCI/MCI or add to cognitive impairment leading to an earlier clinical investigation in predementia AD.
Collapse
Affiliation(s)
- E Auning
- Department of Geriatric Psychiatry, Akershus University Hospital, Ahus campus, Lørenskog, Norway; Institute of Clinical Medicine, Ahus campus University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fujishima M, Maikusa N, Nakamura K, Nakatsuka M, Matsuda H, Meguro K. Mild cognitive impairment, poor episodic memory, and late-life depression are associated with cerebral cortical thinning and increased white matter hyperintensities. Front Aging Neurosci 2014; 6:306. [PMID: 25426066 PMCID: PMC4224123 DOI: 10.3389/fnagi.2014.00306] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022] Open
Abstract
In various independent studies to date, cerebral cortical thickness and white matter hyperintensity (WMH) volume have been associated with episodic memory, depression, and mild cognitive impairment (MCI). The aim of this study was to uncover variations in cortical thickness and WMH volume in association with episodic memory, depressive state, and the presence of MCI simultaneously in a single study population. The participants were 186 individuals with MCI (clinical dementia rating [CDR] of 0.5) and 136 healthy elderly controls (HCs; CDR of 0) drawn from two community-based cohort studies in northern Japan. We computed cerebral cortical thickness and WMH volume by using MR scans and statistically analyzed differences in these indices between HCs and MCI participants. We also assessed the associations of these indices with memory performance and depressive state in participants with MCI. Compared with HCs, MCI participants exhibited thinner cortices in the temporal and inferior parietal lobes and greater WMH volumes in the corona radiata and semioval center. In MCI participants, poor episodic memory was associated with thinner cortices in the left entorhinal region and increased WMH volume in the posterior periventricular regions. Compared with non-depressed MCI participants, depressed MCI participants showed reduced cortical thickness in the anterior medial temporal lobe and gyrus adjacent to the amygdala bilaterally, as well as greater WMH volume as a percentage of the total intracranial volume (WMHr). A higher WMHr was associated with cortical thinning in the frontal, temporal, and parietal regions in MCI participants. These results demonstrate that episodic memory and depression are associated with both cortical thickness and WMH volume in MCI participants. Additional longitudinal studies are needed to clarify the dynamic associations and interactions among these indices.
Collapse
Affiliation(s)
- Motonobu Fujishima
- Department of Nuclear Medicine, Saitama Medical University International Medical Center Hidaka, Japan ; Integrative Brain Imaging Center, National Center of Neurology and Psychiatry (NCNP), Kodaira Tokyo, Japan
| | - Norihide Maikusa
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry (NCNP), Kodaira Tokyo, Japan
| | - Kei Nakamura
- Division of Geriatric Behavioral Neurology, Cyclotron and Radioisotope Center, Tohoku University Sendai, Japan
| | - Masahiro Nakatsuka
- Division of Geriatric Behavioral Neurology, Cyclotron and Radioisotope Center, Tohoku University Sendai, Japan
| | - Hiroshi Matsuda
- Department of Nuclear Medicine, Saitama Medical University International Medical Center Hidaka, Japan ; Integrative Brain Imaging Center, National Center of Neurology and Psychiatry (NCNP), Kodaira Tokyo, Japan
| | - Kenichi Meguro
- Division of Geriatric Behavioral Neurology, Cyclotron and Radioisotope Center, Tohoku University Sendai, Japan
| |
Collapse
|