1
|
Tampi RR, Joshi P, Jeste DV. Psychosis associated with dementia: evaluation and management. Schizophr Res 2025; 281:82-90. [PMID: 40319614 DOI: 10.1016/j.schres.2025.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
Dementia is one of the most common neurodegenerative disorders in the world, and 34-63 % of individuals with dementia have psychotic symptoms. Neurobiological correlates of dementia with psychosis include significantly increased densities of senile plaques and neurofibrillary tangles and higher D3 receptor density. Limitations of proposed diagnostic criteria include a lack of specificity for psychotic symptoms in individuals with dementia, a lack of consistent differentiation between symptoms, late recognition, and not accounting for comorbid depression or agitation that may be the primary symptom, which makes diagnosis challenging. This review aims to provide clinicians, researchers, and policymakers with a comprehensive evaluation of psychosis in dementia, covering its epidemiology, neurobiology, diagnosis, and treatment strategies. We review both psychosocial and pharmacological interventions for dementia-related psychosis. Psychosocial treatments such as meaningful communication between persons with dementia and their caregivers, simplifying the living environment, and optimizing tasks can help reduce the adverse impact of psychosis. Evidence from meta-analyses indicates modest efficacy for cholinesterase inhibitors, antidepressants, and antipsychotics for psychosis in dementia. The use of antipsychotic medications is limited by increased risks for serious adverse effects including cerebrovascular events and death. Emerging therapies such as xanomeline-trospium present promising avenues for treatment. By synthesizing current evidence and clinical guidelines, this review provides a framework for improving diagnosis and treatment of psychosis in dementia, helping clinicians and researchers refine patient care strategies while informing future research directions.
Collapse
Affiliation(s)
- Rajesh R Tampi
- Department of Psychiatry, Creighton University School of Medicine, 7710 Mercy Road, Suite 601, Omaha, NE 68124, USA; Department of Psychiatry, Yale School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA.
| | - Pallavi Joshi
- Banner Alzheimer's Institute, 901 E Willetta St, Phoenix, AZ 85006, USA; Department of Psychiatry, University of Arizona College of Medicine-Phoenix, 475 N 5th, Phoenix, AZ 85004, USA
| | - Dilip V Jeste
- Global Research Network on Social Determinants of Mental Health and Exposomics, La Jolla, CA, USA
| |
Collapse
|
2
|
Liu H, Xie Y, Ji Y, Zhou Y, Xu J, Tang J, Liu N, Ding H, Qin W, Liu F, Yu C. Identification of genetic architecture shared between schizophrenia and Alzheimer's disease. Transl Psychiatry 2025; 15:150. [PMID: 40240757 PMCID: PMC12003746 DOI: 10.1038/s41398-025-03348-w] [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: 07/10/2024] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Both schizophrenia (SCZ) and Alzheimer's disease (AD) are highly heritable brain disorders. Despite of the observed comorbidity and shared psychosis and cognitive decline between the two disorders, the genetic risk architecture shared by SCZ and AD remains largely unknown. Based on summary statistics of the currently available largest genome-wide association studies for SCZ (n = 130,644) and AD (n = 455,258) in individuals of European ancestry, we conducted conditional/conjunctional false discovery rate (FDR) analysis to enhance the statistical power for discovering more genetic associations with SCZ or AD and to detect the common genetic variants shared by both disorders. We found shared genetic architecture in SCZ conditioned on AD and vice versa across different levels of significance, indicating polygenic overlap. We found 268 (78 novel) SCZ-only and 125 (55 novel) AD-only SNPs at conditional FDR < 0.01, and 16 lead SNPs shared by SCZ and AD at conjunctional FDR < 0.05. Only half of the shared SNPs showed concordant effect direction, which was consistent with the modest genetic correlation (r = 0.097; P = 0.026) between the two disorders. This study provides evidence for polygenic overlap between SCZ and AD, suggesting the existence of the shared molecular genetic mechanisms, which may inform therapeutic targets that are applicable for both disorders.
Collapse
Affiliation(s)
- Huaigui Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingying Xie
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuan Ji
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujing Zhou
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Tang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Nana Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Ding
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Wen Qin
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Feng Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
- State Key Laboratory of Experimental Hematology, Tianjin, China.
| |
Collapse
|
3
|
Gómez-Acero L, Sánchez-Fernández N, Subirana P, Ciruela F, Aso E. Striatal dopamine D 2, adenosine A 2A and cannabinoid CB 1 receptors balance as a target against non-cognitive symptoms in a mouse model of Alzheimer's disease. Pharmacol Biochem Behav 2025; 249:173970. [PMID: 39956277 DOI: 10.1016/j.pbb.2025.173970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/03/2024] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Behavioral and psychological symptoms of dementia are almost ubiquitous in Alzheimer's disease (AD) but current therapies are not fully effective and safe. In this study, we aim to evaluate the role played by the interplay among striatal D2, adenosine A2A (A2AR) and cannabinoid CB1 (CB1R) receptors in some of these non-cognitive impairments in a well-established animal model of AD, the double transgenic APP/PS1 mice. Our results reveal that the alterations existing in the ratios between these three receptors significantly correlate with the sensorimotor gating and the social interaction impairments occurring in APP/PS1 mice at 12 months of age. Moreover, the pharmacological stimulation of A2AR and CB1R blunted the sensorimotor gating deficiencies in APP/PS1 mice. To note, we observed some age-dependent differences among male and female mice. In conclusion, the present study provides evidence for the contribution of an altered interplay between dopaminergic, adenosinergic and endocannabinoid systems in the sensorimotor gating deficits and social withdrawal occurring in AD and points to A2AR and CB1R as a potential target to reverse these non-cognitive symptoms in AD patients.
Collapse
Affiliation(s)
- Laura Gómez-Acero
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Nuria Sánchez-Fernández
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Paula Subirana
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
4
|
Alexander L, Beck K, Bocharova M, Young AH, Stewart R, Carter R, Mueller C. Late-life affective disorders and risk of progression to dementia: retrospective cohort study of patients in secondary care. Br J Psychiatry 2025:1-7. [PMID: 40160115 PMCID: PMC7617556 DOI: 10.1192/bjp.2025.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
BACKGROUND Late-life affective disorders (LLADs) are common and are projected to increase by 2050. There have been several studies linking late-life depression to an increased risk of dementia, but it is unclear if bipolar affective disorder or anxiety disorders pose a similar risk. AIMS We aimed to compare the risk of LLADs progressing to all-cause dementia, and the demographic and clinical variables mediating the risk. METHODS We used the South London and Maudsley National Health Service Foundation Trust Clinical Records Interactive Search system to identify patients aged 60 years or older with a diagnosis of any affective disorder. Cox proportional hazard models were used to determine differences in dementia risk between late-life anxiety disorders versus late-life depression, and late-life bipolar disorder versus late-life depression. Demographic and clinical characteristics associated with the risk of dementia were investigated. RESULTS Some 5695 patients were identified and included in the final analysis. Of these, 388 had a diagnosis of bipolar affective disorder, 1365 had a diagnosis of an anxiety disorder and 3942 had a diagnosis of a depressive disorder. Bipolar affective disorder was associated with a lower hazard of developing dementia compared to depression (adjusted model including demographics and baseline cognition, hazard ratio: 0.60; 95% CI: 0.41-0.87). Anxiety disorders had a similar hazard of developing dementia (adjusted hazard ratio: 1.05; 95% CI: 0.90-1.22). A prior history of a depressive disorder reduced the risk of late-life depression progressing to dementia - suggesting the new onset of a depressive disorder in later life is associated with higher risk - but a prior history of anxiety disorders or bipolar affective disorder did not alter risk. CONCLUSIONS LLADs have a differential risk of developing all-cause dementia, with demographic- and illness-related factors influencing the risk. Further prospective cohort studies are needed to explore the link between LLADs and dementia development, and mediators of the lower risk of dementia associated with late-life bipolar disorder compared to late-life depression.
Collapse
Affiliation(s)
- Laith Alexander
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Katherine Beck
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Mariia Bocharova
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Allan H. Young
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Robert Stewart
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Rowena Carter
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Christoph Mueller
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
5
|
Fan J, Churchill N, Fadhel A, Fornazzari LR, De Luca V, Ismail Z, Munoz DG, Schweizer TA, Fischer CE. Determining the Role of Sex and APOE4 status on Psychosis in Alzheimer's Disease. J Geriatr Psychiatry Neurol 2025; 38:8919887241313225. [PMID: 40128188 PMCID: PMC12022363 DOI: 10.1177/08919887241313225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/06/2024] [Accepted: 12/22/2024] [Indexed: 03/26/2025]
Abstract
BackgroundPsychosis occurs in approximately 41% of patients living with Alzheimer's disease. Previous findings from our group based on analyses of a neuropathological cohort suggest that among AD patients with Lewy Body pathology, female APOE4 homozygotes are at significantly greater risk of psychosis. This study aims to replicate this finding in a clinical cohort using data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset.MethodsOur group used data from a sample of patients with AD in the ADNI database from the ADNI1, ADNI2, ADNI3, and ADNIGO studies. We defined psychosis status as experiencing hallucinations or delusions at one time point based on the Neuropsychiatric Inventory. We then used forward binary logistic regression to determine if sex and APOE4 status are predictors of AD + P.ResultsIn total there were 204 participants who met the inclusion criteria, 133 of which were male, and 71 of which were female. Fifty-six patients were APOE4 non-carriers, 109 patients were APOE4 heterozygote carriers, and 39 were APOE4 homozygote carriers. In total, there were 59 patients with psychosis. When adjusting for mini mental state examination score, adjusted hippocampal volume, and age, we demonstrate that female APOE4 homozygotes have a significantly increased risk of psychosis compared to other groups (P = 0.0264, OR = 19.50).DiscussionThe results of our study demonstrate a significant association between psychosis risk and female APOE4 homozygotes, thus corroborating findings from a neuropathological cohort. The effects of APOE ε4 on psychosis risk are significant only in females, and not in males.
Collapse
Affiliation(s)
- Jonathan Fan
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Nathan Churchill
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
| | - Ayad Fadhel
- Faculty of Science, York University, Toronto, ON, Canada
| | - Luis R. Fornazzari
- Faculty of Medicine, Department of Neurology, University of Toronto, Toronto, ON, Canada
| | - Vincenzo De Luca
- Faculty of Medicine, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Zahinoor Ismail
- Cumming School of Medicine, Department of Psychiatry, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - David G. Munoz
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Division of Pathology, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tom A. Schweizer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Corinne E. Fischer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Faculty of Medicine, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
6
|
Soncu Büyükişcan E. Neuropsychology of Alzheimer's disease: From preclinical phase to dementia. APPLIED NEUROPSYCHOLOGY. ADULT 2025:1-9. [PMID: 39982692 DOI: 10.1080/23279095.2025.2469236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by significant cognitive and functional decline, initially presenting with episodic memory impairment. A thorough neuropsychological assessment is essential for AD diagnosis, particularly in the early stages in which interventions may be more effective. This paper reviews the neuropsychology of Alzheimer's disease, highlighting the cognitive progression of the disease. In the typical forms of AD, episodic memory appears to be the first and foremost affected cognitive domain. As AD progresses, cognitive impairments extend beyond memory to affect various domains such as attention, executive functions, language, and visuospatial abilities. Neuropsychiatric issues, such as depression and anxiety, which often accompany cognitive decline, are also common, especially at the advanced stages of the disease. While episodic memory impairment is the earliest and most prominent feature in typical AD cases, comprehensive assessments, including social cognition and neuropsychiatric evaluations, are crucial for accurate diagnosis and treatment planning.
Collapse
|
7
|
Miranda O, Jiang C, Qi X, Kofler J, Sweet RA, Wang L. Exploring Potential Medications for Alzheimer's Disease with Psychosis by Integrating Drug Target Information into Deep Learning Models: A Data-Driven Approach. Int J Mol Sci 2025; 26:1617. [PMID: 40004081 PMCID: PMC11855865 DOI: 10.3390/ijms26041617] [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: 12/13/2024] [Revised: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Approximately 50% of Alzheimer's disease (AD) patients develop psychotic symptoms, leading to a subtype known as psychosis in AD (AD + P), which is associated with accelerated cognitive decline compared to AD without psychosis. Currently, no FDA-approved medication specifically addresses AD + P. This study aims to improve psychosis predictions and identify potential therapeutic agents using the DeepBiomarker deep learning model by incorporating drug-target interactions. Electronic health records from the University of Pittsburgh Medical Center were analyzed to predict psychosis within three months of AD diagnosis. AD + P patients were classified as those with either a formal psychosis diagnosis or antipsychotic prescriptions post-AD diagnosis. Two approaches were employed as follows: (1) a drug-focused method using individual medications and (2) a target-focused method pooling medications by shared targets. The updated DeepBiomarker model achieved an area under the receiver operating curve (AUROC) above 0.90 for psychosis prediction. A drug-focused analysis identified gabapentin, amlodipine, levothyroxine, and others as potentially beneficial. A target-focused analysis highlighted significant proteins, including integrins, calcium channels, and tyrosine hydroxylase, confirming several medications linked to these targets. Integrating drug-target information into predictive models improves the identification of medications for AD + P risk reduction, offering a promising strategy for therapeutic development.
Collapse
Affiliation(s)
- Oshin Miranda
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (O.M.); (C.J.); (X.Q.)
| | - Chen Jiang
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (O.M.); (C.J.); (X.Q.)
| | - Xiguang Qi
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (O.M.); (C.J.); (X.Q.)
| | - Julia Kofler
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Alzheimer Disease Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Robert A. Sweet
- Alzheimer Disease Research Center, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lirong Wang
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA; (O.M.); (C.J.); (X.Q.)
| |
Collapse
|
8
|
Vattathil SM, Blostein F, Miller-Fleming TW, Davis LK, Wingo TS, Wingo AP. GWAS links APOE to neuropsychiatric symptoms in mild cognitive impairment and dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.31.25321498. [PMID: 39974048 PMCID: PMC11838693 DOI: 10.1101/2025.01.31.25321498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Neuropsychiatric symptoms in dementia (NPS) collectively refer to behavioral and psychological symptoms affecting individuals with mild cognitive impairment (MCI) or Alzheimer's disease or related dementia (ADRD). NPS are among the most troubling aspects of living with dementia and their treatments have limited efficacy. We aim to investigate genetic variants contributing to NPS to identify new therapeutic targets. METHODS We performed a genome-wide association study (GWAS) for nine NPS domains measured by the NPI-Q in 12,800 participants of European ancestry with MCI or ADRD recruited by Alzheimer's disease research centers across the U.S. RESULTS We found genome-wide significant signals for agitation, anxiety, apathy, delusions, and hallucinations in the APOE locus that were driven by the APOE ε4 allele. We replicated these findings in two independent datasets. Mediation analyses revealed that MCI/ADRD severity only partially mediated the GWAS signals, except for apathy. DISCUSSION These findings suggest the APOE ε4 allele influences NPS independently of and beyond its effect on ADRD.
Collapse
Affiliation(s)
| | - Freida Blostein
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Genomic Medicine Training Program, Vanderbilt University Medical Center, Nashville, TN
| | - Tyne W. Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Lea K. Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | | | - Thomas S. Wingo
- Department of Neurology, University of California, Davis, Davis, CA
- Alzheimer’s Disease Research Center, University of California, Davis, Davis CA
| | - Aliza P. Wingo
- Department of Psychiatry, University of California, Davis, Davis, CA
- VA Northern California Health Care System, Davis, CA
| |
Collapse
|
9
|
Kouhsar M, Weymouth L, Smith AR, Imm J, Bredemeyer C, Wedatilake Y, Torkamani A, Bergh S, Selbæk G, Mill J, Ballard C, Sweet RA, Kofler J, Creese B, Pishva E, Lunnon K. A brain DNA co-methylation network analysis of psychosis in Alzheimer's disease. Alzheimers Dement 2025; 21:e14501. [PMID: 39936280 DOI: 10.1002/alz.14501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The presence of psychosis in Alzheimer's disease (AD) is suggested to be associated with distinct molecular and neuropathological profiles in the brain. METHODS We assessed brain DNA methylation in AD donors with psychosis (AD+P) and without psychosis (AD-P) using the EPIC array. Weighted gene correlation network analysis identified modules of co-methylated genes in a discovery cohort (PITT-ADRC: N = 113 AD+P, N = 40 AD-P), with validation in an independent cohort (BDR: N = 79 AD+P, N = 117 AD-P), with Gene Ontology and cell-type enrichment analysis. Genetic data were integrated to identify methylation quantitative trait loci (mQTLs), which were co-localized with GWAS for related traits. RESULTS We replicated one AD+P associated module, which was enriched for synaptic pathways and in excitatory and inhibitory neurons. mQTLs in this module co-localized with variants associated with schizophrenia and educational attainment. DISCUSSION This represents the largest epigenetic study of AD+P to date, identifying pleiotropic relationships between AD+P and related traits. HIGHLIGHTS DNA methylation was assessed in the prefrontal cortex in subjects with AD+P and AD-P. WGCNA identified six modules of co-methylated loci associated with AD+P in a discovery cohort. One of the modules was replicated in an independent cohort. This module was enriched for synaptic genes and in excitatory and inhibitory neurons. mQTLs mapping to genes in the module co-localized with GWAS loci for schizophrenia and educational attainment.
Collapse
Affiliation(s)
- Morteza Kouhsar
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| | - Luke Weymouth
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| | - Adam R Smith
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| | - Jennifer Imm
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| | - Claudia Bredemeyer
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| | - Yehani Wedatilake
- Norwegian National Centre for Aging and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Research Centre for Age-related Functional Decline and Disease, Innlandet Hospital Trust, Ottestad, Norway
| | - Ali Torkamani
- The Scripps Research Institute, La Jolla, California, USA
| | - Sverre Bergh
- Norwegian National Centre for Aging and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Research Centre for Age-related Functional Decline and Disease, Innlandet Hospital Trust, Ottestad, Norway
| | - Geir Selbæk
- Norwegian National Centre for Aging and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Nydalen, Oslo, Norway
| | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| | - Clive Ballard
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Byron Creese
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
- Division of Psychology, Department of Life Sciences, Brunel University London, Uxbridge, UK
| | - Ehsan Pishva
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Katie Lunnon
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon, UK
| |
Collapse
|
10
|
Imbimbo C, Cotta Ramusino M, Leone S, Mazzacane F, De Franco V, Gatti A, Perini G, Costa A. Emerging Pharmacological Approaches for Psychosis and Agitation in Alzheimer's Disease. CNS Drugs 2025; 39:143-160. [PMID: 39623197 PMCID: PMC11769872 DOI: 10.1007/s40263-024-01133-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 01/27/2025]
Abstract
Psychosis and agitation are among the most distressing neuropsychiatric symptoms (NPSs) of Alzheimer's disease (AD), linked to faster disease progression and earlier admission to nursing homes. While nonpharmacological treatments may alleviate mild behavioral symptoms, more severe syndromes often require pharmacological intervention. Brexpiprazole is the only medication approved for agitation in AD, although its limited clinical efficacy has raised criticism. No drugs have been approved for treating psychosis in AD, highlighting the critical need for new, effective, and safe treatments. Recent studies have elucidated part of the neurobiological basis of NPSs in the AD brain, offering insights for testing repurposed and novel drugs. We conducted a comprehensive nonsystematic literature review, aiming to provide a critical overview of both current treatments and emerging pharmacological interventions under clinical development for treating psychosis and agitation in AD. Additionally, we present strategies to optimize the clinical development of new drug candidates. We identify three promising compounds that are currently in phase 3 trials: xanomeline-trospium for AD psychosis, and dextromethorphan-bupropion and dexmedetomidine for agitation in AD. We propose that biomarkers linked to the neuropsychiatric traits of AD patients should be identified in dedicated studies and then included in phase 2 dose-range-finding studies with novel compounds to establish biological engagement. Furthermore, phase 3 placebo-controlled studies should be carried out in AD biomarker-confirmed subjects with narrower cognitive impairment ranges and precise NPS severity at screening. Alternative study designs, such as sequential phase approaches, may also be adopted.
Collapse
Affiliation(s)
- Camillo Imbimbo
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy.
| | - Silvia Leone
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Federico Mazzacane
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
| | - Valentino De Franco
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
| | - Alberto Gatti
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
11
|
Nabizadeh F, Sheykhlou S, Mahmoodi S, Khalili E, Zafari R, Hosseini H. Neuroimaging Findings of Psychosis in Alzheimer's Disease: A Systematic Review. Brain Behav 2025; 15:e70205. [PMID: 39740792 DOI: 10.1002/brb3.70205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 11/15/2024] [Accepted: 11/23/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Previous studies on neuroimaging findings in Alzheimer's disease (AD) patients with hallucinations and delusions have yielded inconsistent results. We aimed to systematically review neuroimaging findings of delusions and hallucinations in AD patients to describe the most prominent neuroimaging features. METHODS We performed a comprehensive search in three online databases, including PubMed, Scopus, and Web of Science in June 2023. We included studies that reported neuroimaging features of AD patients with delusion, hallucination, or psychosis. RESULTS After the screening, 34 studies with 2241 AD patients were eligible to be included in our qualitative synthesis. On the basis of the included studies, there are significant changes in the volume and perfusion levels of broad brain areas, including the hippocampus, amygdala, insula, cingulate, occipital, frontal, prefrontal, orbitofrontal, temporal, and parietal cortices in these patients. Moreover, AD patients with psychosis, hallucinations, or delusions reflected different EEG waves compared to AD patients without these disorders. CONCLUSION The results of our review provided evidence about the neuroimaging alterations in AD patients suffering from psychosis, hallucinations, and delusions using different imaging methods. AD patients with psychosis, hallucinations, or delusions have significant differences in the volume and perfusion levels of various brain regions along with alterations in EEG waves and biological molecules compared to patients with only AD.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Alzheimer's Disease Institute, Tehran, Iran
| | - Shadi Sheykhlou
- Medical Laboratory Department, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Mahmoodi
- Medical Laboratory Department, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Khalili
- Universal Scientific Education and Research Network (USERN), Bandar Abbas, Hormozgan, Iran
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Rasa Zafari
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Helia Hosseini
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Westheimer JL, Smith RP, Iacobelli P, Oh H, Tezino L, Khan R, Broussard J, Meltzer G, Obeid N, Cunningham S, Boland RJ, Patriquin MA. The state of (mis) trust: Human-centered technology development & implementation in intensive mental health settings. J Affect Disord 2024; 367:318-323. [PMID: 39226937 DOI: 10.1016/j.jad.2024.08.206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024]
Abstract
Innovative technology-based solutions in mental healthcare promise significant improvements in care quality and clinical outcomes. However, their successful implementation is profoundly influenced by the levels of trust patients hold toward their treatment providers, organizations, and the technology itself. This paper delves into the complexities of building and assessing patient trust within the intensive mental health care context, focusing on inpatient settings. We explore the multifaceted nature of trust, including interpersonal, institutional, and technological trust. We highlight the crucial role of therapeutic trust, which comprises both interpersonal trust between patients and providers, and institutional trust in treatment organizations. The manuscript identifies potential key barriers to trust, from sociocultural background to a patient's psychopathology. Furthermore, it examines the concept of technological trust, emphasizing the influence of digital literacy, socio-economic status, and user experience on patients' acceptance of digital health innovations. By emphasizing the importance of assessing and addressing the state of trust among patients, the overarching goal is to leverage digital innovations to enhance mental healthcare outcomes within intensive mental health settings.
Collapse
Affiliation(s)
| | | | | | - Hyuntaek Oh
- The Menninger Clinic, Houston, TX, USA; Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | | | | - Robert J Boland
- The Menninger Clinic, Houston, TX, USA; Baylor College of Medicine, Houston, TX, USA
| | - Michelle A Patriquin
- The Menninger Clinic, Houston, TX, USA; Baylor College of Medicine, Houston, TX, USA; Michael E. DeBakey VA Medical Center, Houston, TX, USA.
| |
Collapse
|
13
|
Rao S, Forns J, Danysh HE, Calingaert B, Dempsey C, Aquilina T, Pathak S, Anthony MS, Layton JB. Natural history and clinical outcomes in patients with Alzheimer's disease-related psychosis by antipsychotic treatment status in the United States. J Alzheimers Dis 2024; 102:1260-1270. [PMID: 39604278 DOI: 10.1177/13872877241297361] [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: 11/29/2024]
Abstract
BACKGROUND While some literature on clinical outcomes in persons with dementia-related psychosis exists, little is known regarding Alzheimer's disease-related psychosis (ADP). OBJECTIVE Describe demographic/clinical characteristics of adults with ADP and estimate incidence of clinical events by antipsychotic treatment status. METHODS This cohort study identified adults ≥65 years with Alzheimer's disease and incident psychosis (US Medicare database [2013-2018]) and no prior exposure to antipsychotics. Two nonmutually exclusive ADP subcohorts included: patients who initiated treatment with antipsychotic medications (antipsychotic users) and those who remained untreated (antipsychotic nonusers). Baseline characteristics were evaluated before psychosis diagnosis in untreated patients and before antipsychotic initiation in treated patients. Incidence rates were estimated for falls and fractures (composite and separately), seizure/epilepsy (new onset and any), and mortality. RESULTS 145,333 ADP antipsychotic nonusers and 49,452 antipsychotic users were identified. Both cohorts had similar baseline demographics; however, antipsychotic users versus nonusers had higher baseline skilled nursing facility use (40.3% and 27.8%), mood (72.7% and 62.1%) and anxiety (70.9% and 57.3%) disorders, falls/fractures (39.5% and 33.8%), urinary tract infections (55.1% and 47.0%), and frailty index scores (76.0% and 69.7%). Crude incidence rates (95% confidence interval)/100 person-years in antipsychotic users and nonusers were 70.0 (68.9-71.2) and 55.8 (55.4-56.1) (falls/fractures composite), 69.0 (67.9-70.1) and 54.9 (54.5-55.2) (falls), 38.6 (38.1-39.0) and 33.0 (32.7-33.2) (mortality), and 45.8 (44.9-46.7) and 54.2 (53.9-54.6) (any seizure/epilepsy). CONCLUSIONS Antipsychotic initiators with ADP had a higher burden of some baseline comorbidities; experienced higher incidence of falls, fractures, and mortality; and had lower incidence of seizure/epilepsy than antipsychotic nonusers.
Collapse
Affiliation(s)
- Sapna Rao
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Research Triangle Park, NC, USA
| | - Joan Forns
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Barcelona, Spain
| | - Heather E Danysh
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Waltham, MA, USA
| | - Brian Calingaert
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Research Triangle Park, NC, USA
| | - Colleen Dempsey
- Drug Safety and Pharmacovigilance, ACADIA Pharmaceuticals Inc., Princeton, NJ, USA
| | - Thomas Aquilina
- Drug Safety and Pharmacovigilance, ACADIA Pharmaceuticals Inc., Princeton, NJ, USA
| | - Sanjeev Pathak
- Clinical Development, ACADIA Pharmaceuticals Inc., Princeton, NJ, USA
| | - Mary S Anthony
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Research Triangle Park, NC, USA
| | - J Bradley Layton
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Research Triangle Park, NC, USA
| |
Collapse
|
14
|
Scarfo S, Marsella AMA, Grigoriadou L, Moshfeghi Y, McGeown WJ. Neuroanatomical correlates and predictors of psychotic symptoms in Alzheimer's disease: A systematic review and meta-analysis. Neuropsychologia 2024; 204:109006. [PMID: 39326784 DOI: 10.1016/j.neuropsychologia.2024.109006] [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: 05/21/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Psychotic symptoms (hallucinations and delusions) are a type of neuropsychiatric symptom found during Alzheimer's Disease (AD). OBJECTIVE This systematic review aims to comprehensively capture, analyse, and evaluate the body of evidence that has investigated associations between brain regions/networks and psychotic symptoms in AD. METHODS The protocol, created according to the PRISMA guidelines, was pre-registered on OSF (https://osf.io/tg8xp/). Searches were performed using PubMed, Web of Science and PsycInfo. A partial coordinate-based meta-analysis (CBMA) was performed based on data availability. RESULTS Eighty-two papers were selected: delusions were found to be associated mainly with right fronto-temporal brain regions and the insula; hallucinations mainly with fronto-occipital areas; both were frequently associated with the anterior cingulate cortex. The CBMA, performed on the findings of fourteen papers on delusions, identified a cluster in the frontal lobe, one in the putamen, and a smaller one in the insula. CONCLUSIONS The available evidence highlights that key brain regions, predominantly in the right frontal lobe, the anterior cingulate cortex, and temporo-occipital areas, appear to underpin the different manifestations of psychotic symptoms in AD and MCI. The fronto-temporal areas identified in relation to delusions may underpin a failure to assimilate correct information and consider alternative possibilities (which might generate and maintain the delusional belief), and dysfunction within the salience network (anterior cingulate cortex and insula) may suggest a contribution for how internal and external stimuli are identified; the fronto-occipital areas linked to hallucinations may indicate diminished sensory processing and non-optimal predictive processing, that together contribute to misinterpretation of stimuli and misperceptions; the fronto-temporal and occipital areas, as well as the anterior cingulate cortex were linked to the psychotic cluster.
Collapse
Affiliation(s)
- Sara Scarfo
- Department of Psychological Sciences and Health, University of Strathclyde, Glasgow, UK
| | | | - Loulouda Grigoriadou
- Department of Psychological Sciences and Health, University of Strathclyde, Glasgow, UK
| | - Yashar Moshfeghi
- Computer and Information Sciences, University of Strathclyde, Glasgow, UK
| | - William J McGeown
- Department of Psychological Sciences and Health, University of Strathclyde, Glasgow, UK.
| |
Collapse
|
15
|
Kwon KJ, Kim HY, Han SH, Shin CY. Future Therapeutic Strategies for Alzheimer's Disease: Focus on Behavioral and Psychological Symptoms. Int J Mol Sci 2024; 25:11338. [PMID: 39518892 PMCID: PMC11547068 DOI: 10.3390/ijms252111338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive, degenerative brain disorder that impairs memory and thinking skills, leading to significant economic and humanistic burdens. It is associated with various neuropsychiatric symptoms (NPS) such as anxiety, agitation, depression, aggression, apathy, and psychosis. NPSs are common in patients with AD, affecting up to 97% of individuals diagnosed with AD. The severity of NPS is linked to disease progression and cognitive decline. NPS in Alzheimer's disease leads to increased morbidity, mortality, caregiver burden, earlier nursing home placement, and higher healthcare costs. Despite their significant impact, clinical research on NPS in AD is limited. In clinical settings, accurately distinguishing and diagnosing NPS related to AD remains a challenge. Additionally, conventional treatments for NPS in AD are often ineffective, highlighting the need for new therapies that target these specific symptoms. Understanding these comorbidities can aid in early diagnosis and better management of AD. In this review, we provide a summary of the various neurological and psychiatric symptoms (NPS) associated with AD and new candidates under development for the treatment of NPS based on their therapeutic targets and mechanisms. On top of the conventional NPS studied so far, this review adds recent advancements in the understanding of social functional impairment in AD. This review also provides information that can contribute to the advancement of studies and translational research in this field by emphasizing therapeutic targets and mechanisms of action focused on AD-related NPS rather than conventional mechanisms targeted in AD drug development. Above all, considering the relative lack of research in this new field despite the importance of clinical, medical, and translational research, it may increase interest in NPS in AD, its pathophysiological mechanisms, and potential therapeutic candidates such as molecules with antioxidant potential.
Collapse
Affiliation(s)
- Kyoung Ja Kwon
- Department of Pharmacology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea;
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.Y.K.); (S.-H.H.)
- Department of Neurology, Konkuk Hospital Medical Center, 120-1 Neungdong-ro, Gwangjin-Gu, Seoul 05030, Republic of Korea
| | - Hahn Young Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.Y.K.); (S.-H.H.)
- Department of Neurology, Konkuk Hospital Medical Center, 120-1 Neungdong-ro, Gwangjin-Gu, Seoul 05030, Republic of Korea
| | - Seol-Heui Han
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.Y.K.); (S.-H.H.)
- Department of Neurology, Konkuk Hospital Medical Center, 120-1 Neungdong-ro, Gwangjin-Gu, Seoul 05030, Republic of Korea
| | - Chan Young Shin
- Department of Pharmacology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea;
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.Y.K.); (S.-H.H.)
| |
Collapse
|
16
|
Zamboni G, Mattioli I, Arya Z, Tondelli M, Vinceti G, Chiari A, Jenkinson M, Huey ED, Grafman J. Multimodal nonlinear correlates of behavioural symptoms in frontotemporal dementia. Brain Imaging Behav 2024; 18:1226-1238. [PMID: 39243355 PMCID: PMC11582133 DOI: 10.1007/s11682-024-00913-7] [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] [Accepted: 08/23/2024] [Indexed: 09/09/2024]
Abstract
Studies exploring the brain correlates of behavioral symptoms in the frontotemporal dementia spectrum (FTD) have mainly searched for linear correlations with single modality neuroimaging data, either structural magnetic resonance imaging (MRI) or fluoro-deoxy-D-glucose positron emission tomography (FDG-PET). We aimed at studying the two imaging modalities in combination to identify nonlinear co-occurring patterns of atrophy and hypometabolism related to behavioral symptoms. We analyzed data from 93 FTD patients who underwent T1-weighted MRI, FDG-PET imaging, and neuropsychological assessment including the Neuropsychiatric Inventory, Frontal Systems Behavior Scale, and Neurobehavioral Rating Scale. We used a data-driven approach to identify the principal components underlying behavioral variability, then related the identified components to brain variability using a newly developed method fusing maps of grey matter volume and FDG metabolism. A component representing apathy, executive dysfunction, and emotional withdrawal was associated with atrophy in bilateral anterior insula and putamen, and with hypometabolism in the right prefrontal cortex. Another component representing the disinhibition versus depression/mutism continuum was associated with atrophy in the right striatum and ventromedial prefrontal cortex for disinhibition, and hypometabolism in the left fronto-opercular region and sensorimotor cortices for depression/mutism. A component representing psychosis was associated with hypometabolism in the prefrontal cortex and hypermetabolism in auditory and visual cortices. Behavioral symptoms in FTD are associated with atrophy and altered metabolism of specific brain regions, especially located in the frontal lobes, in a hierarchical way: apathy and disinhibition are mostly associated with grey matter atrophy, whereas psychotic symptoms are mostly associated with hyper-/hypo-metabolism.
Collapse
Affiliation(s)
- Giovanna Zamboni
- Università di Modena e Reggio Emilia, Modena, Italy.
- Azienda Ospedaliero Universitaria di Modena, Modena, Italy.
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giardini 1355, Modena, 41126, Italy.
| | | | - Zobair Arya
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Giulia Vinceti
- Università di Modena e Reggio Emilia, Modena, Italy
- Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | | | - Mark Jenkinson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Edward D Huey
- Departments of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, USA
| | - Jordan Grafman
- Shirley Ryan AbilityLab & Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
17
|
Lee HJ, Kim HY, Oh SJ, Son Y, Kang KJ, Nam KR, Choi JY. Administration of Aripiprazole Alleviates Memory Impairment and Restores Damaged Glutamatergic System in 5xFAD Mice. Mol Imaging Biol 2024; 26:879-887. [PMID: 39174788 DOI: 10.1007/s11307-024-01944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/20/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
PURPOSE Many patients with Alzheimer's disease (AD) also have psychosis, and it has been reported that these patients have more severely impaired cognitive functions than patients without psychosis. The glutamatergic system in the brain is known to play an important role in memory and learning in the neural circuits. However, there has been limited research on how antipsychotic drugs affect the glutamatergic system of AD. Therefore, we aimed to investigate the effects of aripiprazole on the glutamatergic system in an animal model of AD using functional molecular imaging. PROCEDURES In this study, 5xFAD mice were used as the animal model. At the age of 5 months, the mice were divided into wild-type, vehicle control, and aripiprazole-treated groups (n = 6 per group). The aripiprazole-treated group was administered aripiprazole for 2 months at a dose of 1 mg·kg-1·day-1. At 7 months of age, the animals underwent behavioral tests and glutamate positron emission tomography (PET) scans. RESULTS The aripiprazole-treated group exhibited alleviated memory impairment in a novel object recognition test. Moreover, this group displayed 7-8% higher binding in the glutamate PET scan than the vehicle-treated 5xFAD group. Postmortem examination confirmed the recovery of glutamatergic damage. CONCLUSIONS The administration of aripiprazole alleviated memory impairment and restored the damaged glutamatergic system in 5xFAD mice. Although the use of aripiprazole in AD patients may be a constraint in terms of safety, we confirmed the possibility that the administration of antipsychotic drugs can be effective in AD.
Collapse
Affiliation(s)
- Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Hyun-Yong Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Yeonghoon Son
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.
| |
Collapse
|
18
|
Swann P, Mirza-Davies A, O'Brien J. Associations Between Neuropsychiatric Symptoms and Inflammation in Neurodegenerative Dementia: A Systematic Review. J Inflamm Res 2024; 17:6113-6141. [PMID: 39262651 PMCID: PMC11389708 DOI: 10.2147/jir.s385825] [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: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Background Neuropsychiatric symptoms are common in dementia and linked to adverse outcomes. Inflammation is increasingly recognized as playing a role as a driver of early disease progression in Alzheimer's disease (AD) and related dementias. Inflammation has also been linked to primary psychiatric disorders, however its association with neuropsychiatric symptoms in neurodegenerative dementias remains uncertain. Methods We conducted a systematic literature review investigating associations between inflammation and neuropsychiatric symptoms in neurodegenerative dementias, including AD, Lewy body, Frontotemporal, Parkinson's (PD) and Huntington's disease dementias. Results Ninety-nine studies met our inclusion criteria, and the majority (n = 59) investigated AD and/or mild cognitive impairment (MCI). Thirty-five studies included PD, and only 6 investigated non-AD dementias. Inflammation was measured in blood, CSF, by genotype, brain tissue and PET imaging. Overall, studies exhibited considerable heterogeneity and evidence for specific inflammatory markers was inconsistent, with lack of replication and few longitudinal studies with repeat biomarkers. Depression was the most frequently investigated symptom. In AD, some studies reported increases in peripheral IL-6, TNF-a associated with depressive symptoms. Preliminary investigations using PET measures of microglial activation found an association with agitation. In PD, studies reported positive associations between TNF-a, IL-6, CRP, MCP-1, IL-10 and depression. Conclusion Central and peripheral inflammation may play a role in neuropsychiatric symptoms in neurodegenerative dementias; however, the evidence is inconsistent. There is a need for multi-site longitudinal studies with detailed assessments of neuropsychiatric symptoms combined with replicable peripheral and central markers of inflammation.
Collapse
Affiliation(s)
- Peter Swann
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - Anastasia Mirza-Davies
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| | - John O'Brien
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
19
|
Oguma T, Jino K, Nakahara K, Asada H, Fuchino K, Nagatani K, Kouki K, Okamoto R, Takai N, Koda K, Fujita S, Sekiguchi Y, Yasuo K, Mayumi K, Abe A, Imono M, Horiguchi N, Iwata S, Kusakabe KI. Dual 5-HT 2A and 5-HT 2C Receptor Inverse Agonist That Affords In Vivo Antipsychotic Efficacy with Minimal hERG Inhibition for the Treatment of Dementia-Related Psychosis. J Med Chem 2024; 67:14478-14492. [PMID: 39137033 DOI: 10.1021/acs.jmedchem.4c01244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Psychosis is a distressing symptom commonly occurring in people with dementia. To treat Parkinson's disease psychosis, pimavanserin (1), a 5-HT2A receptor inverse agonist having minimal 5-HT2C receptor affinity and no dopamine D2 receptor affinity, was approved in the United States, but not for dementia-related psychosis due to limited efficacy issues. Herein, we report on the identification of a potent and dual 5-HT2A and 5-HT2C receptor inverse agonist 8 having minimal hERG inhibition, after having demonstrated the involvement of both 5-HT2A and 5-HT2C receptors to deliver antipsychotic efficacy in an MK-801-induced locomotor model and having conducted 5-HT2A and 5-HT2C occupancy studies including a surrogate method. The introduction of a spirocyclopropyl group boosting 5-HT2C affinity in 1 followed by further optimization to control lipophilicity resulted in balanced dual potency and metabolic stability, and mitigating hERG inhibition led to 8 that showed significant antipsychotic efficacy due to the involvement of both receptors.
Collapse
Affiliation(s)
- Takuya Oguma
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kohei Jino
- Laboratory for Drug Discovery & Disease Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kenji Nakahara
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Hidetsugu Asada
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-yu, Kyoto 606-8501, Japan
| | - Kouki Fuchino
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kotaro Nagatani
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kensuke Kouki
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Ryuji Okamoto
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Nozomi Takai
- Laboratory for Drug Discovery & Disease Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Ken Koda
- Laboratory for Drug Discovery & Disease Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Sayaka Fujita
- Laboratory for Drug Discovery & Disease Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Yusuke Sekiguchi
- Laboratory for Bio-Modality Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kazuya Yasuo
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Kei Mayumi
- Laboratory for Drug Discovery & Development, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Ayane Abe
- Laboratory for Drug Discovery & Development, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Masaaki Imono
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - Naotaka Horiguchi
- Laboratory for Drug Discovery & Disease Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| | - So Iwata
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-yu, Kyoto 606-8501, Japan
| | - Ken-Ichi Kusakabe
- Laboratory for Medicinal Chemistry Research, Shionogi Pharmaceutical Research Center, 1-1 Futaba-cho 3-chome, Toyonaka, Osaka 561-0825, Japan
| |
Collapse
|
20
|
Argyriou S, Fullard JF, Krivinko JM, Lee D, Wingo TS, Wingo AP, Sweet RA, Roussos P. Beyond memory impairment: the complex phenotypic landscape of Alzheimer's disease. Trends Mol Med 2024; 30:713-722. [PMID: 38821772 PMCID: PMC11329360 DOI: 10.1016/j.molmed.2024.04.016] [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: 02/12/2024] [Revised: 04/15/2024] [Accepted: 04/26/2024] [Indexed: 06/02/2024]
Abstract
Neuropsychiatric symptoms (NPSs) in Alzheimer's disease (AD) constitute multifaceted behavioral manifestations that reflect processes of emotional regulation, thinking, and social behavior. They are as prevalent in AD as cognitive impairment and develop independently during the progression of neurodegeneration. As studying NPSs in AD is clinically challenging, most AD research to date has focused on cognitive decline. In this opinion article we summarize emerging literature on the prevalence, time course, and the underlying genetic, molecular, and pathological mechanisms related to NPSs in AD. Overall, we propose that NPSs constitute a cluster of core symptoms in AD, and understanding their neurobiology can lead to a more holistic approach to AD research, paving the way for more accurate diagnostic tests and personalized treatments embracing the goals of precision medicine.
Collapse
Affiliation(s)
- Stathis Argyriou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John F Fullard
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Josh M Krivinko
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donghoon Lee
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thomas S Wingo
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Aliza P Wingo
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA; Veterans Affairs Atlanta Health Care System, Decatur, GA, USA
| | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Street, Bronx, NY, USA; Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Street, Bronx, NY, USA.
| |
Collapse
|
21
|
Ray NR, Kumar A, Zaman A, del Rosario P, Mena PR, Manoochehri M, Stein C, De Vito AN, Sweet RA, Hohman TJ, Cuccaro ML, Beecham GW, Huey ED, Reitz C. Disentangling the genetic underpinnings of neuropsychiatric symptoms in Alzheimer's disease in the Alzheimer's Disease Sequencing Project: Study design and methodology. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70000. [PMID: 39183746 PMCID: PMC11342352 DOI: 10.1002/dad2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Neuropsychiatric symptoms (NPS) are highly prevalent in Alzheimer's disease (AD). There are no effective treatments targeting these symptoms. METHODS To facilitate identification of causative mechanistic pathways, we initiated an effort (NIH: U01AG079850) to collate, harmonize, and analyze all available NPS data (≈ 100,000 samples) of diverse ancestries with whole-genome sequencing data from the Alzheimer's Disease Sequencing Project (ADSP). RESULTS This study will generate a genomic resource for Alzheimer's disease with both harmonized whole-genome sequencing and NPS phenotype data that will be publicly available through NIAGADS. Primary analyses will (1) identify novel genetic risk factors associated with NPS in AD, (2) characterize the shared genetic architecture of NPS in AD and primary psychiatric disorders, and (3) assess the role of ancestry effects in the etiology of NPS in AD. DISCUSSION Expansion of the ADSP to harmonize and refine NPS phenotypes coupled with the proposed core analyses will lay the foundation to disentangle the molecular mechanisms underlying these detrimental symptoms in AD in diverse populations. Highlights Neuropsychiatric symptoms (NPS) are highly prevalent in Alzheimer's disease (AD).There are no effective treatments targeting NPS in AD.The current effort aims to collate, harmonize, and analyze all NPS data from the Alzheimer's Disease Sequencing Project.Core analyses will identify underlying genetic factors and mechanistic pathways.The harmonized genomic and phenotypic data from this initiative will be available through National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site.
Collapse
Affiliation(s)
- Nicholas R. Ray
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Ajneesh Kumar
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Andrew Zaman
- The John P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFloridaUSA
- Dr. John T. MacDonald Foundation Department of Human GeneticsUniversity of MiamiMiamiFloridaUSA
| | - Pamela del Rosario
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Pedro R. Mena
- The John P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFloridaUSA
- Dr. John T. MacDonald Foundation Department of Human GeneticsUniversity of MiamiMiamiFloridaUSA
| | - Masood Manoochehri
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Colin Stein
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Alyssa N. De Vito
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Robert A. Sweet
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Michael L. Cuccaro
- The John P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFloridaUSA
- Dr. John T. MacDonald Foundation Department of Human GeneticsUniversity of MiamiMiamiFloridaUSA
| | - Gary W. Beecham
- The John P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFloridaUSA
- Dr. John T. MacDonald Foundation Department of Human GeneticsUniversity of MiamiMiamiFloridaUSA
| | - Edward D. Huey
- Department of Psychiatry and Human BehaviorAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Christiane Reitz
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
- Department of EpidemiologyColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
22
|
Almeida FC, Jesus T, Coelho A, Quintas-Neves M, Gauthreaux K, Teylan MA, Mock CN, Kukull WA, Crary JF, Oliveira TG. Psychosis in Alzheimer's disease is associated with specific changes in brain MRI volume, cognition and neuropathology. Neurobiol Aging 2024; 138:10-18. [PMID: 38471417 DOI: 10.1016/j.neurobiolaging.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024]
Abstract
Psychosis in Alzheimer's Disease (AD) is prevalent and indicates poor prognosis. However, the neuropathological, cognitive and brain atrophy patterns underlying these symptoms have not been fully elucidated. In this study, we evaluated 178 patients with AD neuropathological change (ADNC) and ante-mortem volumetric brain magnetic resonance imaging (MRI). Presence of psychosis was determined using the Neuropsychiatric Inventory Questionnaire. Clinical Dementia Rating Sum-of-boxes (CDR-SB) was longitudinally compared between groups with a follow-up of 3000 days using mixed-effects multiple linear regression. Neuropsychological tests closest to the time of MRI and brain regional volumes were cross-sectionally compared. Psychosis was associated with lower age of death, higher longitudinal CDR-SB scores, multi-domain cognitive deficits, higher neuritic plaque severity, Braak stage, Lewy Body pathology (LB) and right temporal lobe regional atrophy. Division according to the presence of LB showed differential patterns of AD-typical pathology, cognitive deficits and regional atrophy. In conclusion, psychosis in ADNC with and without LB has clinical value and associates with subgroup patterns of neuropathology, cognition and regional atrophy.
Collapse
Affiliation(s)
- Francisco C Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; Department of Neuroradiology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Tiago Jesus
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; Center Algoritmi, LASI, University of Minho, Braga 4710-057, Portugal
| | - Ana Coelho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Miguel Quintas-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; Department of Neuroradiology, Hospital de Braga, Braga, Portugal
| | - Kathryn Gauthreaux
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, USA
| | - Merilee A Teylan
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, USA
| | - Charles N Mock
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, USA
| | - Walter A Kukull
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, USA
| | - John F Crary
- Neuropathology Brain Bank & Research Core, Department of Pathology, Nash Family Department of Neuroscience, Department of Artificial Intelligence & Human Health, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; Department of Neuroradiology, Hospital de Braga, Braga, Portugal.
| |
Collapse
|
23
|
Ballard C, Tariot P, Soto-Martin M, Pathak S, Liu IY. Challenges and proposed solutions to conducting Alzheimer's disease psychosis trials. Front Psychiatry 2024; 15:1384176. [PMID: 38812491 PMCID: PMC11135469 DOI: 10.3389/fpsyt.2024.1384176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/05/2024] [Indexed: 05/31/2024] Open
Abstract
Alzheimer's disease psychosis (ADP) produces a significant burden for patients and their care partners, but at present there are no approved treatments for ADP. The lack of approved treatments may be due to the challenges of conducting clinical trials for this disease. This perspective article discusses distinct challenges and proposed solutions of conducting ADP trials involving seven key areas: (1) methods to reduce the variable and sometimes high rates of placebo response that occur for treatments of neuropsychiatric symptoms; (2) the use of combined or updated criteria that provide a precise, consensus definition of ADP; (3) the use of eligibility criteria to help recruit individuals representative of the larger ADP population and overcome the difficulty of recruiting patients with moderate-to-severe ADP; (4) consideration of multiple perspectives and implementation of technology to reduce the variability in the administration and scoring of neuropsychiatric symptom assessments; (5) the use of clinically appropriate, a priori-defined severity thresholds and responder cutoffs; (6) the use of statistical approaches that address absolute effect sizes and a three-tier approach to address the fluctuation of neuropsychiatric symptoms; and (7) the implementation of feasible diagnostic and target-engagement biomarkers as they become available. The goal of these proposed solutions is to improve the evaluation of potential ADP therapies, within the context of randomized, placebo-controlled trials with clinically meaningful endpoints and sustained treatment responses.
Collapse
Affiliation(s)
- Clive Ballard
- Institute of Health Research, University of Exeter Medical School, Exeter, United Kingdom
| | - Pierre Tariot
- Banner Alzheimer’s Institute and University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Maria Soto-Martin
- Alzheimer Clinical and Research Centre, Gérontopôle, Toulouse University Hospital, University Hospital Institutes (IHU) HealthAge, Toulouse, France
| | - Sanjeev Pathak
- Department of Medical Affairs, Acadia Pharmaceuticals Inc., San Diego, CA, United States
| | - I-Yuan Liu
- Department of Medical Affairs, Acadia Pharmaceuticals Inc., San Diego, CA, United States
| |
Collapse
|
24
|
Singh AK, Malviya R, Prakash A, Verma S. Neuropsychiatric Manifestations in Alzheimer's Disease Patients: Genetics and Treatment Options. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:39-54. [PMID: 36856177 DOI: 10.2174/1871527322666230301111216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/03/2022] [Accepted: 12/27/2022] [Indexed: 03/02/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by neuropsychiatric symptoms (NPS), which cause great misery to those with dementia and those who care for them and may lead to early institutionalization. OBJECTIVE The present systematic review aims to discuss the various aspects of Alzheimer's, including treatment options. METHODS The databases Embase, PubMed, and Web of Science were searched to collect data. RESULTS Incipient cognitive deterioration is commonly accompanied by these early warning signals of neurocognitive diseases. The neurobiology of NPSs in Alzheimer's disease, as well as particular symptoms, including psychosis, agitation, apathy, sadness, and sleep disorders, will be examined in this review. For NPSs in Alzheimer's disease, clinical trial designs, as well as regulatory issues, were also addressed. A fresh wave of research, however, is helping to push the discipline ahead. For medication development and repurposing, we highlight the most recent results in genetics, neuroimaging, and neurobiology. Even though identifying and treating psychosis in adults with dementia is still a challenging endeavor, new options are coming up that give the field fresh focus and hope. Conclsuion: It can be concluded from the complete literature survey that Alzheimer's-related psychosis as well as other symptoms that are not psychotic, have made significant progress in the last decade. These milestones in the development of safer, more effective treatments have been achieved as a consequence of great focus on non-pharmacological interventions like DICE or WHELD; the investigation into ways to improve existing drugs like aripiprazole, risperidone, amisulpride, and Escitalopram for safer precision-based treatment; and the development of a clinical trial program for pimavanserin.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Anuj Prakash
- Reference Standard Division, Indian Pharmacopoeia Commission, Sec-23, Raj Nagar, Ghaziabad, Uttar Pradesh, India
| | - Swati Verma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| |
Collapse
|
25
|
Zhang J, Wang Y, Zhang Y, Yao J. Genome-wide association study in Alzheimer's disease: a bibliometric and visualization analysis. Front Aging Neurosci 2023; 15:1290657. [PMID: 38094504 PMCID: PMC10716290 DOI: 10.3389/fnagi.2023.1290657] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Thousands of research studies concerning genome-wide association studies (GWAS) in Alzheimer's disease (AD) have been published in the last decades. However, a comprehensive understanding of the current research status and future development trends of GWAS in AD have not been clearly shown. In this study, we tried to gain a systematic overview of GWAS in AD by bibliometric and visualization analysis. METHODS The literature search terms are: ("genome-wide analysis" or "genome-wide association study" or "whole-genome analysis") AND ("Alzheimer's Disease" or "Alzheimer Disease"). Relevant publications were extracted from the Web of Science Core Collection (WoSCC) database. Collected data were further analyzed using VOSviewer, CiteSpace and R package Bibliometrix. The countries, institutions, authors and scholar collaborations were investigated. The co-citation analysis of publications was visualized. In addition, research hotspots and fronts were examined. RESULTS A total of 1,350 publications with 59,818 citations were identified. The number of publications and citations presented a significant rising trend since 2013. The United States was the leading country with an overwhelming number of publications (775) and citations (42,237). The University of Washington and Harvard University were the most prolific institutions with 101 publications each. Bennett DA was the most influential researcher with the highest local H-index. Neurobiology of Aging was the journal with the highest number of publications. Aβ, tau, immunity, microglia and DNA methylation were research hotspots. Disease and causal variants were research fronts. CONCLUSION The most frequently studied AD pathogenesis and research hotspots are (1) Aβ and tau, (2) immunity and microglia, with TREM2 as a potential immunotherapy target, and (3) DNA methylation. The research fronts are (1) looking for genetic similarities between AD and other neurological diseases and syndromes, and (2) searching for causal variants of AD. These hotspots suggest noteworthy directions for future studies on AD pathogenesis and genetics, in which basic research regarding immunity is promising for clinical conversion. The current under-researched directions are (1) GWAS in AD biomarkers based on large sample sizes, (2) studies of causal variants of AD, and (3) GWAS in AD based on non-European populations, which need to be strengthened in the future.
Collapse
Affiliation(s)
- Junyao Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinuo Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junyan Yao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Boyanova ST, Lloyd-Morris E, Corpe C, Rahman KM, Farag DB, Page LK, Wang H, Fleckney AL, Gatt A, Troakes C, Vizcay-Barrena G, Fleck R, Reeves SJ, Thomas SA. Interaction of amisulpride with GLUT1 at the blood-brain barrier. Relevance to Alzheimer's disease. PLoS One 2023; 18:e0286278. [PMID: 37874822 PMCID: PMC10597500 DOI: 10.1371/journal.pone.0286278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/27/2023] [Indexed: 10/26/2023] Open
Abstract
Blood-brain barrier (BBB) dysfunction may be involved in the increased sensitivity of Alzheimer's disease (AD) patients to antipsychotics, including amisulpride. Studies indicate that antipsychotics interact with facilitated glucose transporters (GLUT), including GLUT1, and that GLUT1 BBB expression decreases in AD. We tested the hypotheses that amisulpride (charge: +1) interacts with GLUT1, and that BBB transport of amisulpride is compromised in AD. GLUT1 substrates, GLUT1 inhibitors and GLUT-interacting antipsychotics were identified by literature review and their physicochemical characteristics summarised. Interactions between amisulpride and GLUT1 were studied using in silico approaches and the human cerebral endothelial cell line, hCMEC/D3. Brain distribution of [3H]amisulpride was determined using in situ perfusion in wild type (WT) and 5xFamilial AD (5xFAD) mice. With transmission electron microscopy (TEM) we investigated brain capillary degeneration in WT mice, 5xFAD mice and human samples. Western blots determined BBB transporter expression in mouse and human. Literature review revealed that, although D-glucose has no charge, charged molecules can interact with GLUT1. GLUT1 substrates are smaller (184.95±6.45g/mol) than inhibitors (325.50±14.40g/mol) and GLUT-interacting antipsychotics (369.38±16.04). Molecular docking showed beta-D-glucose (free energy binding: -15.39kcal/mol) and amisulpride (-29.04kcal/mol) interact with GLUT1. Amisulpride did not affect [14C]D-glucose hCMEC/D3 accumulation. [3H]amisulpride uptake into the brain (except supernatant) of 5xFAD mice compared to WT remained unchanged. TEM revealed brain capillary degeneration in human AD. There was no difference in GLUT1 or P-glycoprotein BBB expression between WT and 5xFAD mice. In contrast, caudate P-glycoprotein, but not GLUT1, expression was decreased in human AD capillaries versus controls. This study provides new details about the BBB transport of amisulpride, evidence that amisulpride interacts with GLUT1 and that BBB transporter expression is altered in AD. This suggests that antipsychotics could potentially exacerbate the cerebral hypometabolism in AD. Further research into the mechanism of amisulpride transport by GLUT1 is important for improving antipsychotics safety.
Collapse
Affiliation(s)
- Sevda T. Boyanova
- King’s College London, Institute of Pharmaceutical Science, London, United Kingdom
| | - Ethlyn Lloyd-Morris
- King’s College London, Institute of Pharmaceutical Science, London, United Kingdom
| | - Christopher Corpe
- King’s College London, Department of Nutritional Sciences, School of Life Course Sciences, Faculty of Life Sciences and Medicine, London, United Kingdom
| | | | - Doaa B. Farag
- Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Lee K. Page
- King’s College London, Institute of Pharmaceutical Science, London, United Kingdom
| | - Hao Wang
- King’s College London, Institute of Pharmaceutical Science, London, United Kingdom
| | - Alice L. Fleckney
- King’s College London, Institute of Pharmaceutical Science, London, United Kingdom
| | - Ariana Gatt
- King’s College London, Wolfson Centre for Age Related Disease, London, United Kingdom
| | - Claire Troakes
- King’s College London, London Neurodegenerative Diseases Brain Bank, IoPPN, London, United Kingdom
| | - Gema Vizcay-Barrena
- King’s College London, Centre for Ultrastructural Imaging, London, United Kingdom
| | - Roland Fleck
- King’s College London, Centre for Ultrastructural Imaging, London, United Kingdom
| | - Suzanne J. Reeves
- Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Sarah A. Thomas
- King’s College London, Department of Physiology, London, United Kingdom
| |
Collapse
|
27
|
Pless A, Ware D, Saggu S, Rehman H, Morgan J, Wang Q. Understanding neuropsychiatric symptoms in Alzheimer's disease: challenges and advances in diagnosis and treatment. Front Neurosci 2023; 17:1263771. [PMID: 37732300 PMCID: PMC10508352 DOI: 10.3389/fnins.2023.1263771] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Neuropsychiatric symptoms (NPS) in Alzheimer's disease (AD) affect up to 97% of AD patients, with an estimated 80% of current AD patients experiencing these symptoms. Common AD-associated NPS include depression, anxiety, agitation, aggression, and apathy. The severity of NPS in AD is typically linked to the disease's progression and the extent of cognitive decline. Additionally, these symptoms are responsible for a significant increase in morbidity, mortality, caregiver burden, earlier nursing home placement, and greater healthcare expenditure. Despite their high prevalence and significant impact, there is a notable lack of clinical research on NPS in AD. In this article, we explore and analyze the prevalence, symptom manifestations, challenges in diagnosis, and treatment options of NPS associated with AD. Our literature review reveals that distinguishing and accurately diagnosing the NPS associated with AD remains a challenging task in clinical settings. It is often difficult to discern whether NPS are secondary to pathophysiological changes from AD or are comorbid psychiatric conditions. Furthermore, the availability of effective pharmaceutical interventions, as well as non-pharmacotherapies for NPS in AD, remains limited. By highlighting the advance and challenges in diagnosis and treatment of AD-associated NPS, we aspire to offer new insights into the complexity of identifying and treating these symptoms within the context of AD, and contribute to a deeper understanding of the multifaceted nature of NPS in AD.
Collapse
Affiliation(s)
- Andrew Pless
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Destany Ware
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Hasibur Rehman
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - John Morgan
- Department of Neurology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
28
|
Zamboni G, Mattioli I, Arya Z, Tondelli M, Vinceti G, Chiari A, Jenkinson M, Huey ED, Grafman J. Multimodal nonlinear correlates of behavioural symptoms in frontotemporal dementia. RESEARCH SQUARE 2023:rs.3.rs-3271530. [PMID: 37674710 PMCID: PMC10479452 DOI: 10.21203/rs.3.rs-3271530/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Background Studies exploring the brain correlates of behavioural symptoms in the frontotemporal dementia spectrum (FTD) have mainly searched for linear correlations with single modality neuroimaging data, either structural magnetic resonance imaging (MRI) or fluoro-deoxy-D-glucose positron emission tomography (FDG-PET). We aimed at studying the two imaging modalities in combination to identify nonlinear co-occurring patterns of atrophy and hypometabolism related to behavioural symptoms. Methods We analysed data from 93 FTD patients who underwent T1-weighted MRI, FDG-PET imaging, and neuropsychological assessment including the Neuropsychiatric Inventory, Frontal Systems Behaviour Scale, and Neurobehavioral Rating Scale. We used a data-driven approach to identify the principal components underlying behavioural variability, then related the identified components to brain variability using a newly developed method fusing maps of grey matter volume and FDG metabolism. Results A component representing apathy, executive dysfunction, and emotional withdrawal was associated with atrophy in bilateral anterior insula and putamen, and with hypometabolism in the right prefrontal cortex. Another component representing the disinhibition versus depression/mutism continuum was associated with atrophy in the right striatum and ventromedial prefrontal cortex for disinhibition, and hypometabolism in the left fronto-opercular region and sensorimotor cortices for depression/mutism. A component representing psychosis was associated with hypometabolism in the prefrontal cortex and hypermetabolism in auditory and visual cortices. Discussion Behavioural symptoms in FTD are associated with atrophy and altered metabolism of specific brain regions, especially located in the frontal lobes, in a hierarchical way: apathy and disinhibition are mostly associated with grey matter atrophy, whereas psychotic symptoms are mostly associated with hyper-/hypo-metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jordan Grafman
- Shirley Ryan AbilityLab & Northwestern University Feinberg School of Medicine
| |
Collapse
|
29
|
Fan P, Zeng L, Ding Y, Kofler J, Silverstein J, Krivinko J, Sweet RA, Wang L. Combination of antidepressants and antipsychotics as a novel treatment option for psychosis in Alzheimer's disease. CPT Pharmacometrics Syst Pharmacol 2023; 12:1119-1131. [PMID: 37128639 PMCID: PMC10431054 DOI: 10.1002/psp4.12979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023] Open
Abstract
Psychotic symptoms are reported as one of the most common complications of Alzheimer's disease (AD), in whom they are associated with more rapid deterioration and increased mortality. Empiric treatments, namely first and second-generation antipsychotics, confer modest efficacy in patients with AD and with psychosis (AD+P) and themselves increase mortality. Recent studies suggested the use and beneficial effects of antidepressants among patients with AD+P. This motivates our rationale for exploring their potential as a novel combination therapy option among these patients. We included electronic medical records of 10,260 patients with AD in our study. Survival analysis was performed to assess the effects of the combination of antipsychotics and antidepressants on the mortality of these patients. A protein-protein interaction network representing AD+P was built, and network analysis methods were used to quantify the efficacy of these drugs on AD+P. A combined score was developed to measure the potential synergetic effect against AD+P. Our survival analyses showed that the co-administration of antidepressants with antipsychotics have a significant beneficial effect in reducing mortality. Our network analysis showed that the targets of antipsychotics and antidepressants are well-separated, and antipsychotics and antidepressants have similar Signed Jaccard Index (SJI) scores to AD+P. Eight drug pairs, including some popular recommendations like aripiprazole/sertraline, showed higher than average scores which suggest their potential in treating AD+P via strong synergetic effects. Our proposed combinations of antipsychotic and antidepressant therapy showed a strong superiority over current antipsychotics treatment for AD+P. The observed beneficial effects can be further strengthened by optimizing drug-pair selection based on our systems pharmacology analysis.
Collapse
Affiliation(s)
- Peihao Fan
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences/School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Lang Zeng
- Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ying Ding
- Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Julia Kofler
- Division of Neuropathology, Department of PathologyUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Jonathan Silverstein
- Department of Biomedical Informatics, School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Joshua Krivinko
- Department of Psychiatry, School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Robert A. Sweet
- Department of Psychiatry, School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Alzheimer Disease Research CenterUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Lirong Wang
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences/School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
30
|
Eassa NE, Perez SM, Boley AM, Elam HB, Sharmin D, Cook JM, Lodge DJ. α5-GABAA Receptor Modulation Reverses Behavioral and Neurophysiological Correlates of Psychosis in Rats with Ventral Hippocampal Alzheimer's Disease-like Pathology. Int J Mol Sci 2023; 24:11788. [PMID: 37511546 PMCID: PMC10380527 DOI: 10.3390/ijms241411788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Of the 35 million people in the world suffering from Alzheimer's Disease (AD), up to half experience comorbid psychosis. Antipsychotics, used to treat psychosis, are contraindicated in elderly patients because they increase the risk of premature death. Reports indicate that the hippocampus is hyperactive in patients with psychosis and those with AD. Preclinical studies have demonstrated that the ventral hippocampus (vHipp) can regulate dopamine system function, which is thought to underlie symptoms of psychosis. A viral-mediated approach was used to express mutated human genes known to contribute to AD pathology: the Swedish (K670N, M671L), Florida (I716V), and London (V717I) mutations of amyloid precursor protein and two mutations (M146L and L286V) of presenilin 1 specifically in the vHipp, to investigate the selective contribution of AD-like pathology in this region. We observed a significant increase in dopamine neuron population activity and behavioral deficits in this AD-AAV model that mimics observations in rodent models with psychosis-like symptomatologies. Further, systemic administration of MP-III-022 (α5-GABAA receptor selective positive allosteric modulator) was able to reverse aberrant dopamine system function in AD-AAV rats. This study provides evidence for the development of drugs that target α5-GABAA receptors for patients with AD and comorbid psychosis.
Collapse
Affiliation(s)
- Nicole E. Eassa
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (N.E.E.); (A.M.B.); (H.B.E.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Stephanie M. Perez
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (N.E.E.); (A.M.B.); (H.B.E.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Angela M. Boley
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (N.E.E.); (A.M.B.); (H.B.E.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Hannah B. Elam
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (N.E.E.); (A.M.B.); (H.B.E.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA; (D.S.); (J.M.C.)
| | - James M. Cook
- Department of Chemistry and Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA; (D.S.); (J.M.C.)
| | - Daniel J. Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA; (N.E.E.); (A.M.B.); (H.B.E.); (D.J.L.)
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| |
Collapse
|
31
|
Fan P, Miranda O, Qi X, Kofler J, Sweet RA, Wang L. Unveiling the Enigma: Exploring Risk Factors and Mechanisms for Psychotic Symptoms in Alzheimer's Disease through Electronic Medical Records with Deep Learning Models. Pharmaceuticals (Basel) 2023; 16:911. [PMID: 37513822 PMCID: PMC10385983 DOI: 10.3390/ph16070911] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Around 50% of patients with Alzheimer's disease (AD) may experience psychotic symptoms after onset, resulting in a subtype of AD known as psychosis in AD (AD + P). This subtype is characterized by more rapid cognitive decline compared to AD patients without psychosis. Therefore, there is a great need to identify risk factors for the development of AD + P and explore potential treatment options. In this study, we enhanced our deep learning model, DeepBiomarker, to predict the onset of psychosis in AD utilizing data from electronic medical records (EMRs). The model demonstrated superior predictive capacity with an AUC (area under curve) of 0.907, significantly surpassing conventional risk prediction models. Utilizing a perturbation-based method, we identified key features from multiple medications, comorbidities, and abnormal laboratory tests, which notably influenced the prediction outcomes. Our findings demonstrated substantial agreement with existing studies, underscoring the vital role of metabolic syndrome, inflammation, and liver function pathways in AD + P. Importantly, the DeepBiomarker model not only offers a precise prediction of AD + P onset but also provides mechanistic understanding, potentially informing the development of innovative treatments. With additional validation, this approach could significantly contribute to early detection and prevention strategies for AD + P, thereby improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Peihao Fan
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Oshin Miranda
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiguang Qi
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Julia Kofler
- Department of Pathology, Division of Neuropathology, UPMC Presbyterian Hospital, Pittsburgh, PA 15213, USA
| | - Robert A Sweet
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lirong Wang
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
32
|
Krivinko JM, DeChellis-Marks MR, Zeng L, Fan P, Lopez OL, Ding Y, Wang L, Kofler J, MacDonald ML, Sweet RA. Targeting the post-synaptic proteome has therapeutic potential for psychosis in Alzheimer Disease. Commun Biol 2023; 6:598. [PMID: 37268664 PMCID: PMC10238472 DOI: 10.1038/s42003-023-04961-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/20/2023] [Indexed: 06/04/2023] Open
Abstract
Individuals with Alzheimer Disease who develop psychotic symptoms (AD + P) experience more rapid cognitive decline and have reduced indices of synaptic integrity relative to those without psychosis (AD-P). We sought to determine whether the postsynaptic density (PSD) proteome is altered in AD + P relative to AD-P, analyzing PSDs from dorsolateral prefrontal cortex of AD + P, AD-P, and a reference group of cognitively normal elderly subjects. The PSD proteome of AD + P showed a global shift towards lower levels of all proteins relative to AD-P, enriched for kinases, proteins regulating Rho GTPases, and other regulators of the actin cytoskeleton. We computationally identified potential novel therapies predicted to reverse the PSD protein signature of AD + P. Five days of administration of one of these drugs, the C-C Motif Chemokine Receptor 5 inhibitor, maraviroc, led to a net reversal of the PSD protein signature in adult mice, nominating it as a novel potential treatment for AD + P.
Collapse
Affiliation(s)
- J M Krivinko
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - M R DeChellis-Marks
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - L Zeng
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - P Fan
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - O L Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Y Ding
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - L Wang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - J Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - M L MacDonald
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - R A Sweet
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
33
|
Monge-García S, García-Ayllón MS, Sánchez-Payá J, Gasparini-Berenguer R, Cortés-Gómez MÁ, Sáez-Valero J, Monge-Argilés JA. Validity of CSF alpha-synuclein to predict psychosis in prodromal Alzheimer's disease. Front Neurol 2023; 14:1124145. [PMID: 37292130 PMCID: PMC10244520 DOI: 10.3389/fneur.2023.1124145] [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: 12/14/2022] [Accepted: 04/18/2023] [Indexed: 06/10/2023] Open
Abstract
Background Alzheimer's disease (AD) accompanied by psychotic symptoms (PS) has a poor prognosis and may be associated with imbalances in key neural proteins such as alpha-synuclein (AS). Aim The aim of the study was to evaluate the diagnostic validity of AS levels in the cerebrospinal fluid (CSF) as a predictor of the emergence of PS in patients with prodromal AD. Materials and methods Patients with mild cognitive impairment were recruited between 2010 and 2018. Core AD biomarkers and AS levels were measured in CSF obtained during the prodromal phase of the illness. All patients who met the NIA-AA 2018 criteria for AD biomarkers received treatment with anticholinesterasic drugs. Follow-up evaluations were conducted to assess patients for the presence of psychosis using current criteria; the use of neuroleptic drugs was required for inclusion in the psychosis group. Several comparisons were made, taking into account the timing of the emergence of PS. Results A total of 130 patients with prodromal AD were included in this study. Of these, 50 (38.4%) met the criteria for PS within an 8-year follow-up period. AS was found to be a valuable CSF biomarker to differentiate between the psychotic and non-psychotic groups in every comparison made, depending on the onset of PS. Using an AS level of 1,257 pg/mL as the cutoff, this predictor achieved at least 80% sensitivity. Conclusion To our knowledge, this study represents the first time that a CSF biomarker has shown diagnostic validity for prediction of the emergence of PS in patients with prodromal AD.
Collapse
Affiliation(s)
- Sonia Monge-García
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - María-Salud García-Ayllón
- Hospital General Universitario de Elche, FISABIO,Unidad de Investigación, Valencia, Spain
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
- Unidad de Investigación, Hospital General Universitario de Elche, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Elche, Spain
| | - José Sánchez-Payá
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Servicio de Medicina Preventiva, Hospital General Universitario Dr. Balmis, Alicante, Spain
| | | | - María-Ángeles Cortés-Gómez
- Hospital General Universitario de Elche, FISABIO,Unidad de Investigación, Valencia, Spain
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
- Unidad de Investigación, Hospital General Universitario de Elche, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Elche, Spain
| | - Javier Sáez-Valero
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
- Unidad de Investigación, Hospital General Universitario de Elche, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Elche, Spain
| | - José-Antonio Monge-Argilés
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Servicio de Neurología, Hospital General Universitario Dr. Balmis, Alicante, Spain
| |
Collapse
|
34
|
Ehtezazi T, Rahman K, Davies R, Leach AG. The Pathological Effects of Circulating Hydrophobic Bile Acids in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:173-211. [PMID: 36994114 PMCID: PMC10041467 DOI: 10.3233/adr-220071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Recent clinical studies have revealed that the serum levels of toxic hydrophobic bile acids (deoxy cholic acid, lithocholic acid [LCA], and glycoursodeoxycholic acid) are significantly higher in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) when compared to control subjects. The elevated serum bile acids may be the result of hepatic peroxisomal dysfunction. Circulating hydrophobic bile acids are able to disrupt the blood-brain barrier and promote the formation of amyloid-β plaques through enhancing the oxidation of docosahexaenoic acid. Hydrophobic bile acid may find their ways into the neurons via the apical sodium-dependent bile acid transporter. It has been shown that hydrophobic bile acids impose their pathological effects by activating farnesoid X receptor and suppressing bile acid synthesis in the brain, blocking NMDA receptors, lowering brain oxysterol levels, and interfering with 17β-estradiol actions such as LCA by binding to E2 receptors (molecular modelling data exclusive to this paper). Hydrophobic bile acids may interfere with the sonic hedgehog signaling through alteration of cell membrane rafts and reducing brain 24(S)-hydroxycholesterol. This article will 1) analyze the pathological roles of circulating hydrophobic bile acids in the brain, 2) propose therapeutic approaches, and 3) conclude that consideration be given to reducing/monitoring toxic bile acid levels in patients with AD or aMCI, prior/in combination with other treatments.
Collapse
Affiliation(s)
- Touraj Ehtezazi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rhys Davies
- The Walton Centre, NHS Foundation Trust, Liverpool, UK
| | - Andrew G Leach
- School of Pharmacy, University of Manchester, Manchester, UK
| |
Collapse
|
35
|
Kawakami I, Arai T, Ikeda K, Niizato K, Oshima K, Akiyama H. Possible association of limbic tau pathology with psychosis or behavioral disturbances: Studies of two autopsied psychiatric patients. Neuropathology 2023; 43:44-50. [PMID: 36341554 DOI: 10.1111/neup.12870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/09/2022]
Abstract
Tauopathies, including Alzheimer's disease and primary age-related tauopathy (PART), present heterogeneous clinico-pathological phenotypes that include dementia, aphasia, motor neuron diseases, and psychiatric symptoms. PART is neuropathologically characterized by the presence of neurofibrillary tangles in limbic regions without significant Aβ deposition, but its clinical features have not yet been fully established. Here, we present two patients with distinct psychosis and behavioral symptoms. At autopsy, these patients showed tau pathologies that could not be classified as typical PART, although PART-like neurofibrillary tangles were present in limbic regions. Clinically, both patients were admitted to mental hospitals due to severe delusions or other neuropsychiatric/behavioral symptoms. The first case presented with hallucination, delusion, and apathy at age 70, and died of pancreatic cancer at age 75. He had neuronal cytoplasmic inclusions with selective accumulation of 3Rtau in the striatum and thorn-shaped astrocytes in the amygdala. The second case, who presented with abnormal behaviors such as wandering, agitation and disinhibition, exhibited limbic neurodegeneration with massive 4R tau-positive oligodendroglial inclusions in the medial temporal white matter. His age at onset was 73, and the duration of disease was 15 years. These findings support the notion that distinct limbic tau pathology with concomitant degeneration of the related neural circuits might induce specific psychosis and behavioral symptoms. This underlines the importance of neuropathological evaluation for both clinical education and practice in the fields of neuropathology and neuropsychiatry.
Collapse
Affiliation(s)
- Ito Kawakami
- Department of Psychiatry, Juntendo University School of Medicine, Tokyo, Japan.,Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Tokyo Metropolitan Matsuzawa Hospital, Tokyo, Japan
| | - Tetsuaki Arai
- Department of Psychiatry, University of Tsukuba, Tsukuba, Japan
| | - Kenji Ikeda
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | | - Haruhiko Akiyama
- Department of Clinical Research, Yokohama Brain and Spine Center, Yokohama, Japan
| |
Collapse
|
36
|
Fan P, Zeng L, Ding Y, Kofler J, Silverstein J, Krivinko J, Sweet RA, Wang L. Combination of Antidepressants and Antipsychotics as A Novel Treatment Option for Psychosis in Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.24.23284970. [PMID: 36747620 PMCID: PMC9901071 DOI: 10.1101/2023.01.24.23284970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Psychotic symptoms are reported as one of the most common complications of Alzheimer's disease (AD), affecting approximately half of AD patients, in whom they are associated with more rapid deterioration and increased mortality. Empiric treatments, namely first and second-generation antipsychotics, confer modest efficacy in AD patients with psychosis (AD+P) and themselves increase mortality. A recent genome-wide meta-analysis and early clinical trials suggest the use and beneficial effects of antidepressants among AD+P patients. This motivates our rationale for exploring their potential as a novel combination therapy option amongst these patients. Methods We included University of Pittsburgh Medical Center (UPMC) electronic medical records (EMRs) of 10,260 AD patients from January 2004 and October 2019 in our study. Survival analysis was performed to assess the effects of the combination of antipsychotics and antidepressants on the mortality of these patients. To provide more valuable insights on the hidden mechanisms of the combinatorial therapy, a protein-protein interaction (PPI) network representing AD+P was built, and network analysis methods were used to quantify the efficacy of these drugs on AD+P. An indicator score combining the measurements on the separation between drugs and the proximity between the drugs and AD+P was used to measure the effect of an antipsychotic-antidepressant drug pair against AD+P. Results Our survival analyses replicated that antipsychotic usage is strongly associated with increased mortality in AD patients while the co-administration of antidepressants with antipsychotics showed a significant beneficial effect in reducing mortality. Our network analysis showed that the targets of antipsychotics and antidepressants are well-separated, and antipsychotics and antidepressants have similar proximity scores to AD+P. Eight drug pairs, including some popular recommendations like Aripiprazole/Sertraline and other pairs not reported previously like Iloperidone/Maprotiline showed higher than average indicator scores which suggest their potential in treating AD+P via strong synergetic effects as seen in our study. Conclusion Our proposed combinations of antipsychotics and antidepressants therapy showed a strong superiority over current antipsychotics treatment for AD+P. The observed beneficial effects can be further strengthened by optimizing drug-pair selection based on our systems pharmacology analysis.
Collapse
|
37
|
Fisher DW, Dunn JT, Keszycki R, Rodriguez G, Bennett DA, Wilson RS, Dong H. Unique Transcriptional Signatures Correlate with Behavioral and Psychological Symptom Domains in Alzheimer's Disease. RESEARCH SQUARE 2023:rs.3.rs-2444391. [PMID: 36711772 PMCID: PMC9882691 DOI: 10.21203/rs.3.rs-2444391/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Despite the significant burden, cost, and worse prognosis of Alzheimer's disease (AD) with behavioral and psychological symptoms of dementia (BPSD), little is known about the molecular causes of these symptoms. Using antemortem assessments of BPSD in AD, we demonstrate that individual BPSD can be grouped into 4 domain factors in our sample: affective, apathy, agitation, and psychosis. Then, we performed a transcriptome-wide analysis for each domain utilizing bulk RNA-seq of post-mortem anterior cingulate cortex (ACC) tissue. Though all 4 domains are associated with a predominantly downregulated pattern of hundreds of differentially expressed genes (DEGs), most DEGs are unique to each domain, with only 22 DEGs being common to all BPSD domains, including TIMP1. Weighted gene co-expression network analysis (WGCNA) yielded multiple transcriptional modules that were shared between BPSD domains or unique to each domain, and NetDecoder was used to analyze context-dependent information flow through the biological network. For the agitation domain, we found that all DEGs and a highly correlated transcriptional module were functionally enriched for ECM-related genes including TIMP1, TAGLN, and FLNA. Another unique transcriptional module also associated with the agitation domain was enriched with genes involved in post-synaptic signaling, including DRD1, PDE1B, CAMK4, and GABRA4. By comparing context-dependent changes in DEGs between cases and control networks, ESR1 and PARK2 were implicated as two high impact genes associated with agitation that mediated significant information flow through the biological network. Overall, our work establishes unique targets for future study of the biological mechanisms of BPSD and resultant drug development.
Collapse
Affiliation(s)
- Daniel W. Fisher
- Department of Psychiatry and Behavioral Sciences,
Northwestern University Feinberg School of Medicine
- Department of Psychiatry and Behavioral Sciences,
University of Washington School of Medicine
| | - Jeffrey T. Dunn
- Department of Psychiatry and Behavioral Sciences,
Northwestern University Feinberg School of Medicine
| | - Rachel Keszycki
- Department of Psychiatry and Behavioral Sciences,
Northwestern University Feinberg School of Medicine
- Mesulam Center for Cognitive Neurology and
Alzheimer’s Disease, Northwestern University Feinberg School of
Medicine
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences,
Northwestern University Feinberg School of Medicine
| | | | | | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences,
Northwestern University Feinberg School of Medicine
| |
Collapse
|
38
|
Tzeng NS, Her YN, Chang HA, Wan FJ. Lurasidone treatment for delusional infestation in a patient with dementia. JOURNAL OF MEDICAL SCIENCES 2023. [DOI: 10.4103/jmedsci.jmedsci_410_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
|
39
|
Kurhan F, Akın M. A New Hope in Alzheimer's Disease Psychosis: Pimavanserin. Curr Alzheimer Res 2023; 20:403-408. [PMID: 37641988 DOI: 10.2174/1567205020666230825124922] [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: 02/15/2023] [Revised: 06/04/2023] [Accepted: 06/30/2023] [Indexed: 08/31/2023]
Abstract
Alzheimer's disease (AD) ranks first among the causes of dementia worldwide. AD can develop a psychotic manifest at a significant rate. AD prognosis worsens by added psychosis clinic. There is no treatment approved by the United States Food and Drug Administration (FDA) among antipsychotics for Alzheimer's disease Psychosis (ADP). However, pimavanserine, an atypical antipsychotic, has been approved by the FDA for Parkinson's psychosis. It is predicted that pimavanserin, a new antipsychotic, will fill an important gap in this area. In clinical trials, it appears to be effective in the treatment of delusions and hallucinations at psychosis in both Parkinson's and AD. In this systematic review, we evaluated the analysis of current literature data on pimavanserin used in ADP. We searched the existing literature on clinical studies on pimavanserin therapy used in ADP. Data were determined by systematically searching PubMed, MEDLINE, EMBASE, and Google Scholar until December 2022. A total of 35 citations were found and uploaded on the Mendeley program. Abstracts and full texts of literature data were examined. Pimavanserin was observed, and satisfactory results were obtained in treating ADP. Pimavanserin has a unique mechanism of action. Pimavanserin, an atypical antipsychotic drug, has a low affinity for 5-HT2C receptors and has selective 5-HT2A reverse agonist/antagonist action. Pimavanserin has no clinically significant affinity for dopaminergic, histaminergic, muscarinic or adrenergic receptors. This agent may also achieve significant positive results in resistant psychosis treatments.
Collapse
Affiliation(s)
- Faruk Kurhan
- Department of Psychiatry, Faculty of Medicine, Van Yuzuncu Yil University, 65100, Van, Turkey
| | - Mustafa Akın
- Department of Psychiatry, Faculty of Medicine, Van Yuzuncu Yil University, 65100, Van, Turkey
| |
Collapse
|
40
|
Accelerated atrophy in dopaminergic targets and medial temporo-parietal regions precedes the onset of delusions in patients with Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci 2023; 273:229-241. [PMID: 35554669 PMCID: PMC9958148 DOI: 10.1007/s00406-022-01417-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/11/2022] [Indexed: 12/21/2022]
Abstract
People with Alzheimer's disease (AD) and delusions have worse quality of life and prognosis. However, early markers of delusions have not been identified yet. The present study investigated whether there are any detectable differences in grey matter (GM) volume and cognitive changes in the year before symptom onset between patients with AD who did and did not develop delusions. Two matched samples of AD patients, 63 who did (PT-D) and 63 who did not develop delusions (PT-ND) over 1 year, were identified from the Alzheimer's Disease Neuroimaging Initiative database. The Neuropsychiatric Inventory (NPI) was used to assess the presence of delusions. Sixty-three additional matched healthy controls (HC) were selected. Repeated-measures ANCOVA models were used to investigate group-by-time effects on the volume of selected GM regions of interest and on cognitive performance. No neurocognitive differences were observed between patient groups prior to symptom onset. Greater episodic memory decline and GM loss in bilateral caudate nuclei, medio-temporal and midline cingulo-parietal regions were found in the PT-D compared with the PT-ND group. A pattern of faster GM loss in brain areas typically affected by AD and in cortical and subcortical targets of dopaminergic pathways, paralleled by worsening of episodic memory and behavioural symptoms, may explain the emergence of delusions in patients with AD.
Collapse
|
41
|
Wang Y, Chen L. DeepPerVar: a multi-modal deep learning framework for functional interpretation of genetic variants in personal genome. Bioinformatics 2022; 38:5340-5351. [PMID: 36271868 PMCID: PMC9750124 DOI: 10.1093/bioinformatics/btac696] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/04/2022] [Accepted: 10/20/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Understanding the functional consequence of genetic variants, especially the non-coding ones, is important but particularly challenging. Genome-wide association studies (GWAS) or quantitative trait locus analyses may be subject to limited statistical power and linkage disequilibrium, and thus are less optimal to pinpoint the causal variants. Moreover, most existing machine-learning approaches, which exploit the functional annotations to interpret and prioritize putative causal variants, cannot accommodate the heterogeneity of personal genetic variations and traits in a population study, targeting a specific disease. RESULTS By leveraging paired whole-genome sequencing data and epigenetic functional assays in a population study, we propose a multi-modal deep learning framework to predict genome-wide quantitative epigenetic signals by considering both personal genetic variations and traits. The proposed approach can further evaluate the functional consequence of non-coding variants on an individual level by quantifying the allelic difference of predicted epigenetic signals. By applying the approach to the ROSMAP cohort studying Alzheimer's disease (AD), we demonstrate that the proposed approach can accurately predict quantitative genome-wide epigenetic signals and in key genomic regions of AD causal genes, learn canonical motifs reported to regulate gene expression of AD causal genes, improve the partitioning heritability analysis and prioritize putative causal variants in a GWAS risk locus. Finally, we release the proposed deep learning model as a stand-alone Python toolkit and a web server. AVAILABILITY AND IMPLEMENTATION https://github.com/lichen-lab/DeepPerVar. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ye Wang
- Department of Computer Science and Software Engineering, Auburn University, Auburn, AL 36849, USA
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, FL 32603, USA
| |
Collapse
|
42
|
Fan P, Kofler J, Ding Y, Marks M, Sweet RA, Wang L. Efficacy difference of antipsychotics in Alzheimer's disease and schizophrenia: explained with network efficiency and pathway analysis methods. Brief Bioinform 2022; 23:bbac394. [PMID: 36151774 PMCID: PMC9677501 DOI: 10.1093/bib/bbac394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/14/2022] Open
Abstract
Approximately 50% of Alzheimer's disease (AD) patients will develop psychotic symptoms and these patients will experience severe rapid cognitive decline compared with those without psychosis (AD-P). Currently, no medication has been approved by the Food and Drug Administration for AD with psychosis (AD+P) specifically, although atypical antipsychotics are widely used in clinical practice. These drugs have demonstrated modest efficacy in managing psychosis in individuals with AD, with an increased frequency of adverse events, including excess mortality. We compared the differences between the genetic variations/genes associated with AD+P and schizophrenia from existing Genome-Wide Association Study and differentially expressed genes (DEGs). We also constructed disease-specific protein-protein interaction networks for AD+P and schizophrenia. Network efficiency was then calculated to characterize the topological structures of these two networks. The efficiency of antipsychotics in these two networks was calculated. A weight adjustment based on binding affinity to drug targets was later applied to refine our results, and 2013 and 2123 genes were identified as related to AD+P and schizophrenia, respectively, with only 115 genes shared. Antipsychotics showed a significantly lower efficiency in the AD+P network than in the schizophrenia network (P < 0.001) indicating that antipsychotics may have less impact in AD+P than in schizophrenia. AD+P may be caused by mechanisms distinct from those in schizophrenia which result in a decreased efficacy of antipsychotics in AD+P. In addition, the network analysis methods provided quantitative explanations of the lower efficacy of antipsychotics in AD+P.
Collapse
Affiliation(s)
- Peihao Fan
- School of Pharmacy, University of Pittsburgh
| | | | - Ying Ding
- Department of Biostatistics at the University of Pittsburgh
| | - Michael Marks
- Center for Neuroscience at the University of Pittsburgh and the Department of Neurobiology
| | - Robert A Sweet
- UPMC Endowed Professor of Psychiatric Neuroscience and Professor of Neurology at the University of Pittsburgh
| | - Lirong Wang
- department of pharmaceutical sciences, school of pharmacy at University of Pittsburgh, USA
| |
Collapse
|
43
|
D'Antonio F, Di Vita A, Zazzaro G, Canevelli M, Trebbastoni A, Campanelli A, Ferracuti S, de Lena C, Guariglia C, Boccia M. Cortical complexity alterations in the medial temporal lobe are associated with Alzheimer's disease psychosis. NEUROPSYCHOLOGY, DEVELOPMENT, AND COGNITION. SECTION B, AGING, NEUROPSYCHOLOGY AND COGNITION 2022; 29:1022-1032. [PMID: 34294005 DOI: 10.1080/13825585.2021.1958139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
Psychosis is frequent in Alzheimer's disease (AD) and it is associated with a worse disease course. AD psychosis may represent a distinct AD phenotype, though its specific neurobiological underpinnings have yet to be identified. This study investigated neural underpinnings of AD psychosis using surface-based-morphometry.Data from 32 AD patients, 17 with psychosis (AD-P) and 15 without were analyzed. Average cortical complexity (fractal dimension, FD) was estimated for each theoretically motivated ROI and patient. First, we compared regional FD in AD-P and AD patients. Then we calculated the correlation coefficients between FD and the severity of misidentification and paranoid psychotic symptoms. AD-P showed decreased FD in ventral-visual-stream compared to AD, suggesting that perceptual processes might be pivotal in psychosis. A negative correlation was found between misidentification severity and FD in the entorhinal cortex suggesting that misidentification may be specifically associated with alterations in regions involved in high-level perceptual and contextualization processes.
Collapse
Affiliation(s)
- Fabrizia D'Antonio
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Antonella Di Vita
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Giulia Zazzaro
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Marco Canevelli
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | | | | | - Stefano Ferracuti
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Carlo de Lena
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Cecilia Guariglia
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maddalena Boccia
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
44
|
Rantala MJ, Luoto S, Borráz-León JI, Krams I. Schizophrenia: the new etiological synthesis. Neurosci Biobehav Rev 2022; 142:104894. [PMID: 36181926 DOI: 10.1016/j.neubiorev.2022.104894] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/25/2022] [Accepted: 09/25/2022] [Indexed: 10/31/2022]
Abstract
Schizophrenia has been an evolutionary paradox: it has high heritability, but it is associated with decreased reproductive success. The causal genetic variants underlying schizophrenia are thought to be under weak negative selection. To unravel this paradox, many evolutionary explanations have been suggested for schizophrenia. We critically discuss the constellation of evolutionary hypotheses for schizophrenia, highlighting the lack of empirical support for most existing evolutionary hypotheses-with the exception of the relatively well supported evolutionary mismatch hypothesis. It posits that evolutionarily novel features of contemporary environments, such as chronic stress, low-grade systemic inflammation, and gut dysbiosis, increase susceptibility to schizophrenia. Environmental factors such as microbial infections (e.g., Toxoplasma gondii) can better predict the onset of schizophrenia than polygenic risk scores. However, researchers have not been able to explain why only a small minority of infected people develop schizophrenia. The new etiological synthesis of schizophrenia indicates that an interaction between host genotype, microbe infection, and chronic stress causes schizophrenia, with neuroinflammation and gut dysbiosis mediating this etiological pathway. Instead of just alleviating symptoms with drugs, the parasite x genotype x stress model emphasizes that schizophrenia treatment should focus on detecting and treating possible underlying microbial infection(s), neuroinflammation, gut dysbiosis, and chronic stress.
Collapse
Affiliation(s)
- Markus J Rantala
- Department of Biology, University of Turku, FIN-20014 Turku, Finland.
| | - Severi Luoto
- School of Population Health, University of Auckland, 1023 Auckland, New Zealand
| | | | - Indrikis Krams
- Institute of Ecology and Earth Sciences, University of Tartu, 51014 Tartu, Estonia; Department of Zoology and Animal Ecology, Faculty of Biology, University of Latvia, 1004, Rīga, Latvia
| |
Collapse
|
45
|
Valcic M, Khoury MA, Kim J, Fornazzari L, Churchill NW, Ismail Z, De Luca V, Tsuang D, Schweizer TA, Munoz DG, Fischer CE. Determining Whether Sex and Zygosity Modulates the Association between APOE4 and Psychosis in a Neuropathologically-Confirmed Alzheimer's Disease Cohort. Brain Sci 2022; 12:1266. [PMID: 36139002 PMCID: PMC9497154 DOI: 10.3390/brainsci12091266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The APOE4 allele is a genetic risk factor for developing late-onset Alzheimer's disease (AD). Previous work by our group revealed that female APOE4 homozygotes with Lewy body (LB) pathology were more likely to experience psychosis compared to female APOE4 non-carriers, whereas in males there was no APOE4 dose-dependent significant effect. The objective of this study was to refine our previous findings by adjusting for covariates and determining the probability of an APOE4 sex-mediated effect on psychosis. METHODS Neuropathologically confirmed AD patients with LB pathology (n = 491) and without LB pathology (n = 716) were extracted from the National Alzheimer's Coordinating Center (NACC). Patients were classified as psychotic if they scored positively for delusions and/or hallucinations on the Neuropsychiatric Inventory. Analysis consisted of a preliminary unadjusted binary logistic regression and a Generalized Additive binary logistic regression Model (GAM) to predict the relationship between APOE4 status and sex on the presence of psychosis in both cohorts, adjusting for age, education and MMSE. RESULTS In the cohort with LB pathology, female APOE4 homozygotes were significantly more likely to experience psychosis compared to female APOE4 non-carriers (OR = 4.15, 95%CI [1.21, 14.2], p = 0.023). Female heterozygotes were also more likely to experience psychosis compared to female APOE4 non-carriers, but to a lesser extent (OR = 2.37, 95%CI [1.01, 5.59], p = 0.048). There was no significant difference in males with LB pathology or in any sex in the cohort without LB pathology. CONCLUSIONS Sex and zygosity influence the effect of APOE4 on psychosis in neuropathologically confirmed AD patients, with the effect being limited to females with LB pathology.
Collapse
Affiliation(s)
- Mila Valcic
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
| | - Marc A. Khoury
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
| | - Julia Kim
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
| | - Luis Fornazzari
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
- Department of Neurology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Nathan W. Churchill
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
| | - Zahinoor Ismail
- Departments of Psychiatry, Clinical Neurosciences, Community Health Sciences, and Pathology, Hotchkiss Brain Institute and O’Brien Institute of Public Health, University of Calgary, Calgary, AB T2N 4Z6, Canada;
| | - Vincenzo De Luca
- Division of Geriatric Psychiatry, Centre for Addiction & Mental Health, Toronto, ON M5T 1R8, Canada;
| | - Debby Tsuang
- GRECC, VA Puget Sound and Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195-6560, USA;
| | - Tom A. Schweizer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Division of Neurosurgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
| | - David G. Munoz
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Pathology, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
| | - Corinne E. Fischer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, ON M5B 1T8, Canada; (M.V.); (M.A.K.); (J.K.); (L.F.); (N.W.C.); (T.A.S.); (D.G.M.)
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| |
Collapse
|
46
|
Krivinko JM, Erickson SL, MacDonald ML, Garver ME, Sweet RA. Fingolimod mitigates synaptic deficits and psychosis-like behavior in APP/PSEN1 mice. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12324. [PMID: 36016832 PMCID: PMC9395154 DOI: 10.1002/trc2.12324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/27/2022] [Accepted: 05/31/2022] [Indexed: 04/18/2023]
Abstract
Introduction Current treatments for psychosis in Alzheimer's disease (AD), a syndrome characterized by more rapid deterioration and reduced synaptic protein abundance relative to non-psychotic AD, are inadequate. Fingolimod, a currently US Food and Drug Administration (FDA)-approved pharmacotherapy for multiple sclerosis, alters synaptic protein expression and warrants preclinical appraisal as a candidate pharmacotherapy for psychosis in AD. Methods Presenilin and amyloid precursor protein transgenic mice (APPswe/PSEN1dE9) and wild-type mice were randomized to fingolimod or saline for 7 days. Psychosis-associated behaviors were quantified by open field testing, pre-pulse inhibition of the acoustic startle response testing, and habituation of the acoustic startle response testing. Synaptic proteins were quantified by liquid chromatography/mass spectrometry in homogenate and postsynaptic density fractions. Results Fingolimod treatment increased the synaptic protein abundance in cortical homogenates and normalized psychosis-associated behaviors in APPswe/PSEN1dE9 mice relative to saline. Mitochondrial-related proteins were preferentially altered by fingolimod treatment and correlated with improvements in psychosis-associated behaviors. Discussion Preclinical studies employing complementary psychosis-associated behavioral assessments and proteomic evaluations across multiple AD-related models are warranted to replicate the current study and further investigate fingolimod as a candidate treatment for psychosis in AD.
Collapse
Affiliation(s)
- Josh M. Krivinko
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Susan L. Erickson
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Matthew L. MacDonald
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Megan E. Garver
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Robert A. Sweet
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
47
|
Gozdas E, Hinkley L, Fingerhut H, Dacorro L, Gu M, Sacchet MD, Hurd R, Hosseini SMH. 1H-MRS neurometabolites and associations with neurite microstructures and cognitive functions in amnestic mild cognitive impairment. Neuroimage Clin 2022; 36:103159. [PMID: 36063758 PMCID: PMC9450331 DOI: 10.1016/j.nicl.2022.103159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/04/2022] [Accepted: 08/18/2022] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) pathogenesis is associated with alterations in neurometabolites and cortical microstructure. However, our understanding of alterations in neurochemicals in the prefrontal cortex and their relationship with changes in cortical microstructure in AD remains unclear. Here, we studied the levels of neurometabolites in the left dorsolateral prefrontal cortex (DLPFC) in healthy older adults and patients with amnestic Mild Cognitive Impairments (aMCI) using single-voxel proton-magnetic resonance spectroscopy (1H-MRS). N-acetyl aspartate (NAA), glutamate+glutamate (Glx), Myo-inositol (mI), and γ-aminobutyric acid (GABA) brain metabolite levels were quantified relative to total creatine (tCr = Cr + PCr). aMCI had significantly decreased NAA/tCr, Glx/tCr, NAA/mI, and increased mI/tCr levels compared with healthy controls. Further, we leveraged advanced diffusion MRI to extract neurite properties in the left DLPFC and found a significant positive correlation between NAA/tCr, related to neuronal intracellular compartment, and neurite density (ICVF, intracellular volume fraction), and a negative correlation between mI/tCr and neurite orientation (ODI) only in healthy older adults. These data suggest a potential decoupling in the relationship between neurite microstructures and NAA and mI concentrations in DLPFC in the early stage of AD. Together, our results confirm altered DLPFC neurometabolites in prodromal phase of AD and provide unique evidence regarding the imbalance in the association between neurometabolites and neurite microstructure in early stage of AD.
Collapse
Affiliation(s)
- Elveda Gozdas
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Lauren Hinkley
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hannah Fingerhut
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Lauren Dacorro
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Meng Gu
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Matthew D Sacchet
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Ralph Hurd
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - S M Hadi Hosseini
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
48
|
Wingo TS, Liu Y, Gerasimov ES, Vattathil SM, Wynne ME, Liu J, Lori A, Faundez V, Bennett DA, Seyfried NT, Levey AI, Wingo AP. Shared mechanisms across the major psychiatric and neurodegenerative diseases. Nat Commun 2022; 13:4314. [PMID: 35882878 PMCID: PMC9325708 DOI: 10.1038/s41467-022-31873-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 07/07/2022] [Indexed: 12/14/2022] Open
Abstract
Several common psychiatric and neurodegenerative diseases share epidemiologic risk; however, whether they share pathophysiology is unclear and is the focus of our investigation. Using 25 GWAS results and LD score regression, we find eight significant genetic correlations between psychiatric and neurodegenerative diseases. We integrate the GWAS results with human brain transcriptomes (n = 888) and proteomes (n = 722) to identify cis- and trans- transcripts and proteins that are consistent with a pleiotropic or causal role in each disease, referred to as causal proteins for brevity. Within each disease group, we find many distinct and shared causal proteins. Remarkably, 30% (13 of 42) of the neurodegenerative disease causal proteins are shared with psychiatric disorders. Furthermore, we find 2.6-fold more protein-protein interactions among the psychiatric and neurodegenerative causal proteins than expected by chance. Together, our findings suggest these psychiatric and neurodegenerative diseases have shared genetic and molecular pathophysiology, which has important ramifications for early treatment and therapeutic development.
Collapse
Affiliation(s)
- Thomas S Wingo
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
| | - Yue Liu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Selina M Vattathil
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Meghan E Wynne
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jiaqi Liu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Adriana Lori
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
| | - Victor Faundez
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Aliza P Wingo
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA.
- Veterans Affairs Atlanta Health Care System, Decatur, GA, USA.
| |
Collapse
|
49
|
Pezzoli S, Manca R, Cagnin A, Venneri A. A Multimodal Neuroimaging and Neuropsychological Study of Visual Hallucinations in Alzheimer’s Disease. J Alzheimers Dis 2022; 89:133-149. [DOI: 10.3233/jad-215107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Hallucinations in Alzheimer’s disease (AD) have been linked to more severe cognitive and functional decline. However, research on visual hallucinations (VH), the most common type of hallucinations in AD, is limited. Objective: To investigate the cognitive and cerebral macrostructural and metabolic features associated with VH in AD. Methods: Twenty-four AD patients with VH, 24 with no VH (NVH), and 24 cognitively normal (CN) matched controls were selected from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. Differences in regional gray matter (GM) volumes and cognitive performance were investigated with whole brain voxel-based morphometry analyses of MRI structural brain scans, and analyses of neuropsychological tests. Glucose metabolic changes were explored in a subsample of patients who had FDG-PET scans available. Results: More severe visuoconstructive and attentional deficits were found in AD VH compared with NVH. GM atrophy and hypometabolism were detected in occipital and temporal areas in VH patients in comparison with CN. On the other hand, NVH patients had atrophy and hypometabolism mainly in temporal areas. No differences in GM volume and glucose metabolism were found in the direct comparison between AD VH and NVH. Conclusion: In addition to the pattern of brain abnormalities typical of AD, occipital alterations were observed in patients with VH compared with CN. More severe visuoconstructive and attentional deficits were found in AD VH when directly compared with NVH, and might contribute to the emergence of VH in AD.
Collapse
Affiliation(s)
- Stefania Pezzoli
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Riccardo Manca
- Department of Life Sciences, Brunel University London, London, UK
| | - Annachiara Cagnin
- Department of Neurosciences, University of Padua, Padua, Italy
- Padua Neuroscience Center (PNC), University of Padua, Padua, Italy
| | - Annalena Venneri
- Department of Life Sciences, Brunel University London, London, UK
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | |
Collapse
|
50
|
Eickhoff S, Franzen L, Korda A, Rogg H, Trulley VN, Borgwardt S, Avram M. The Basal Forebrain Cholinergic Nuclei and Their Relevance to Schizophrenia and Other Psychotic Disorders. Front Psychiatry 2022; 13:909961. [PMID: 35873225 PMCID: PMC9299093 DOI: 10.3389/fpsyt.2022.909961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
The basal forebrain cholinergic nuclei (BFCN) provide the main cholinergic input to prefrontal cortices, the hippocampi, and amygdala. These structures are highly relevant for the regulation and maintenance of many cognitive functions, such as attention and memory. In vivo neuroimaging studies reported alterations of the cholinergic system in psychotic disorders. Particularly, a downregulation of nicotinic and muscarinic acetylcholine receptors has been found. Crucially, such alterations in neurotransmission have been associated with cognitive impairments and positive and negative symptoms. Recent pharmacological studies support these findings, as they demonstrated an association between the manipulation of cholinergic transmission and an attenuation in symptom severity. Targeting acetylcholine receptors has therefore become a focus for the development of novel psychopharmacological drugs. However, many open questions remain. For instance, it remains elusive what causes such alterations in neurotransmission. While evidence supports the idea that BFCN structural integrity is altered in schizophrenia, it remains to be determined whether this is also present in other psychotic disorders. Furthermore, it is unclear when throughout the course of the disorder these alterations make their appearance and whether they reflect changes in the BFCN alone or rather aberrant interactions between the BFCN and other brain areas. In this review, the specific role of the BFCN and their projections are discussed from a neuroimaging perspective and with a focus on psychotic disorders alongside future directions. These directions set the stage for the development of new treatment targets for psychotic disorders.
Collapse
Affiliation(s)
- Sofia Eickhoff
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | | | |
Collapse
|