1
|
Tendo ON, Galiwango R, Kinyanda E, Sajatovic M, Kaddumukasa M, Kaddumukasa M, Katabira E, Nabbumba C, Soraya S, Hemmings S, Kalungi A. Genetic Determinants Of Major Depressive Disorder (MDD) Among Adult Persons Living With HIV In Uganda. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.19.25324246. [PMID: 40166531 PMCID: PMC11957178 DOI: 10.1101/2025.03.19.25324246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Major Depressive Disorder (MDD) has a heritable component, with estimates of heritability ranging from 30% to 40%. Depression is a significant comorbidity in people living with HIV (PLWHIV), increasing the risk of suicide-related behaviors. This study investigated the genetic risk loci associated with MDD among adults living with HIV in Uganda, where limited data exist on this relationship. Methods The case-control study analyzed 282 samples (139 MDD cases and 143 controls), assessed for MDD at baseline, six months, and one year using the Mini International Neuropsychiatric Interview. Blood samples were collected at these intervals, with DNA genotyping conducted in South Africa using the H3Africa array. Data were analyzed using PLINK2 and GEMMA for quality control and genome-wide association analysis respectively, followed by functional mapping with FUMA. Results While no significant single nucleotide polymorphisms (SNPs) were identified at the genome-wide threshold, six SNPs were found to be suggestively associated with MDD. These SNPs, which have been associated with other psychiatric conditions like Alzheimer's, alcohol use disorder, and bipolar disorder and have not previously been linked to MDD. Conclusion The study suggests the potential for novel MDD genetic risk loci discovery in PLWHIV and people of African ancestry, especially with larger sample sizes.
Collapse
Affiliation(s)
- Olga Nsangi Tendo
- Department of Immunology and Molecular Biology, Makerere University, P.O.Box 7072, Kampala, Uganda
- African Centers of Excellence in Bioinformatics and Data Intensive Sciences, Kampala, Uganda
| | - Ronald Galiwango
- Department of Immunology and Molecular Biology, Makerere University, P.O.Box 7072, Kampala, Uganda
- African Centers of Excellence in Bioinformatics and Data Intensive Sciences, Kampala, Uganda
- Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418, Kampala, Uganda
| | - Eugene Kinyanda
- Medical Research Council/Uganda Virus Research Institute (MRC/UVRI) and London School of Hygiene & Tropical Medicine(LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Martha Sajatovic
- School of Medicine, Case Western Reserve University School, Cleveland, Ohio, United States of America
| | - Mark Kaddumukasa
- Department of Medicine, Makerere University, P.O.Box 7072, Kampala, Uganda
| | - Martin Kaddumukasa
- Department of Medicine, Makerere University, P.O.Box 7072, Kampala, Uganda
| | - Elly Katabira
- Department of Medicine, Makerere University, P.O.Box 7072, Kampala, Uganda
| | - Catherine Nabbumba
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Seedat Soraya
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Sian Hemmings
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Allan Kalungi
- Medical Research Council/Uganda Virus Research Institute (MRC/UVRI) and London School of Hygiene & Tropical Medicine(LSHTM) Uganda Research Unit, Entebbe, Uganda
| |
Collapse
|
2
|
Willett JDS, Waqas M, Choi Y, Ngai T, Mullin K, Tanzi RE, Prokopenko D. Identification of 16 novel Alzheimer's disease loci using multi-ancestry meta-analyses. Alzheimers Dement 2025; 21:e14592. [PMID: 39998322 PMCID: PMC11852348 DOI: 10.1002/alz.14592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most prevalent form of dementia. While many AD-associated genetic determinants have been identified, few studies have analyzed individuals of non-European ancestry. METHODS We conducted a multi-ancestry genome-wide association study (GWAS) of clinically diagnosed AD and AD-by-proxy using whole genome sequencing data from the National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site (NIAGADS), National Institute of Mental Health, UK Biobank (UKB), and All of Us (AoU) consisting of 49,149 cases (12,074 clinically diagnosed and 37,075 AD-by-proxy) and 383,225 controls. Nearly half of NIAGADS and AoU participants were of non-European ancestry. RESULTS For clinically diagnosed AD, we identified 14 new loci-five common (FBN2/SCL27A6, AC090115.1, DYM, KCNG1/AL121785.1, TIAM1) and nine rare (VWA5B1, RNU6-755P/LMX1A, MOB1A, MORC1-AS1, LINC00989, PDE4D, RNU2-49P/CDO1, NEO1, and SLC35G3/AC022916.1). Meta-analysis of UKB and AoU AD-by-proxy cases yielded two new rare loci (RPL23/LASP1 and CEBPA/AC008738.6), also nominally significant in NIAGADS. DISCUSSION In summary, we provide evidence for 16 novel AD loci and advocate for more studies using whole genome sequencing-based GWAS of diverse cohorts. HIGHLIGHTS We used whole-genome sequencing data from large and diverse cohorts. We found novel genome-wide association study findings based on whole-genome data. We performed a multiancestry meta-analysis and incorporated results from underrepresented groups.
Collapse
Affiliation(s)
- Julian Daniel Sunday Willett
- Genetics and Aging Research Unit and the McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Mohammad Waqas
- Genetics and Aging Research Unit and the McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Younjung Choi
- Genetics and Aging Research Unit and the McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Tiffany Ngai
- Genetics and Aging Research Unit and the McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Department of Systems Design EngineeringUniversity of WaterlooWaterlooOntarioCanada
| | - Kristina Mullin
- Genetics and Aging Research Unit and the McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit and the McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit and the McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
3
|
Willett JDS, Mullin K, Tanzi RE, Prokopenko D. Matching Heterogeneous Cohorts by Projected Principal Components Reveals Two Novel Alzheimer's Disease-Associated Genes in the Hispanic Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.18.25320774. [PMID: 39867396 PMCID: PMC11759617 DOI: 10.1101/2025.01.18.25320774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in elderly, affecting 6.9 million individuals in the United States. Some studies have suggested the prevalence of AD is greater in individuals who self-identify as Hispanic. Focused results are relevant for personalized and equitable clinical interventions. Ethnicity as a stratifying tool in genetic studies is often accompanied by genomic inflation due to heterogeneity. In this study, we report GWAS and meta-analyses conducted among NIAGADS subjects who self-identified as Hispanic and All of Us (AoU) sub-cohorts matched to that cohort, using projected genetically-derived principal components, with and without age and sex. In Hispanic NIAGADS subjects, we identified a common variant in PIEZO2 that was protective for AD with a p-value just beyond genome-wide significance (p = 5.4 * 10-8). Meta-analyses with genetically-matched AoU participants yielded three (two novel) genome-wide significant AD-associated loci based on rare lead variants: rs374043832 (RGS6/PSEN1), rs192423465 (ASPSCR1), and rs935208076 (GDAP2), which were also nominally significant in AoU sub-cohorts. We also show how genomic inflation can be mitigated in heterogeneous populations while increasing sample size and result generalizability.
Collapse
Affiliation(s)
- Julian Daniel Sunday Willett
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Kristina Mullin
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Rudolph E. Tanzi
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Dmitry Prokopenko
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
4
|
Steffens DC, Garrett ME, Soldano KL, McQuoid DR, Ashley-Koch AE, Potter GG. Genome-wide screen to identify genetic loci associated with cognitive decline in late-life depression. Int Psychogeriatr 2024; 36:1021-1029. [PMID: 32641180 PMCID: PMC7794099 DOI: 10.1017/s1041610220001143] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/30/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE This study sought to conduct a comprehensive search for genetic risk of cognitive decline in the context of geriatric depression. DESIGN A genome-wide association study (GWAS) analysis in the Neurocognitive Outcomes of Depression in the Elderly (NCODE) study. SETTING Longitudinal, naturalistic follow-up study. PARTICIPANTS Older depressed adults, both outpatients and inpatients, receiving care at an academic medical center. MEASUREMENTS The Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropsychological battery was administered to the study participants at baseline and a minimum of twice within a subsequent 3-year period in order to measure cognitive decline. A GWAS analysis was conducted to identify genetic variation that is associated with baseline and change in the CERAD Total Score (CERAD-TS) in NCODE. RESULTS The GWAS of baseline CERAD-TS revealed a significant association with an intergenic single-nucleotide polymorphism (SNP) on chromosome 6, rs17662598, that surpassed adjustment for multiple testing (p = 3.7 × 10-7; false discovery rate q = 0.0371). For each additional G allele, average baseline CERAD-TS decreased by 8.656 points. The most significant SNP that lies within a gene was rs11666579 in SLC27A1 (p = 1.1 × 10-5). Each additional copy of the G allele was associated with an average decrease of baseline CERAD-TS of 4.829 points. SLC27A1 is involved with processing docosahexaenoic acid (DHA), an endogenous neuroprotective compound in the brain. Decreased levels of DHA have been associated with the development of Alzheimer's disease. The most significant SNP associated with CERAD-TS decline over time was rs73240021 in GRXCR1 (p = 1.1 × 10-6), a gene previously linked with deafness. However, none of the associations within genes survived adjustment for multiple testing. CONCLUSIONS Our GWAS of cognitive function and decline among individuals with late-life depression (LLD) has identified promising candidate genes that, upon replication in other cohorts of LLD, may be potential biomarkers for cognitive decline and suggests DHA supplementation as a possible therapy of interest.
Collapse
Affiliation(s)
- D C Steffens
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - M E Garrett
- Department of Medicine, Duke University Medicine Center, Durham, NC, USA
| | - K L Soldano
- Department of Medicine, Duke University Medicine Center, Durham, NC, USA
| | - D R McQuoid
- Department of Psychiatry, Duke University Medicine Center, Durham, NC, USA
| | - A E Ashley-Koch
- Department of Medicine, Duke University Medicine Center, Durham, NC, USA
| | - G G Potter
- Department of Psychiatry, Duke University Medicine Center, Durham, NC, USA
| |
Collapse
|
5
|
Willett JDS, Waqas M, Choi Y, Ngai T, Mullin K, Tanzi RE, Prokopenko D. Identification of 16 novel Alzheimer's disease susceptibility loci using multi-ancestry meta-analyses of clinical Alzheimer's disease and AD-by-proxy cases from four whole genome sequencing datasets. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.11.24313439. [PMID: 39314934 PMCID: PMC11419201 DOI: 10.1101/2024.09.11.24313439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. While many AD-associated genetic determinants have been previously identified, few studies have analyzed individuals of non-European ancestry. Here, we describe a multi-ancestry genome-wide association study of clinically-diagnosed AD and AD-by-proxy using whole genome sequencing data from NIAGADS, NIMH, UKB, and All of Us (AoU) consisting of 49,149 cases (12,074 clinically-diagnosed and 37,075 AD-by-proxy) and 383,225 controls. Nearly half of NIAGADS and AoU participants are of non-European ancestry. For clinically-diagnosed AD, we identified 14 new loci - five common (FBN2,/SCL27A6, AC090115.1, DYM, KCNG1/AL121785.1, TIAM1) and nine rare (VWA5B1, RNU6-755P/LMX1A, MOB1A, MORC1-AS1, LINC00989, PDE4D, RNU2-49P/CDO1, NEO1, and SLC35G3/AC022916.1). Meta-analysis of UKB and AoU AD-by-proxy cases yielded two new rare loci (RPL23/LASP1 and CEBPA/ AC008738.6) which were also nominally significant in NIAGADS. In summary, we provide evidence for 16 novel AD loci and advocate for more studies using WGS-based GWAS of diverse cohorts.
Collapse
Affiliation(s)
- Julian Daniel Sunday Willett
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Mohammad Waqas
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Younjung Choi
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Tiffany Ngai
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
- University of Waterloo, Department of Systems Design Engineering, Waterloo, Ontario, Canada
| | - Kristina Mullin
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit and the McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
6
|
Huang YY, Gan YH, Yang L, Cheng W, Yu JT. Depression in Alzheimer's Disease: Epidemiology, Mechanisms, and Treatment. Biol Psychiatry 2024; 95:992-1005. [PMID: 37866486 DOI: 10.1016/j.biopsych.2023.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/13/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023]
Abstract
Depression and Alzheimer's disease (AD) are substantial public health concerns. In the past decades, a link between the 2 disease entities has received extensive acknowledgment, yet the complex nature of this relationship demands further clarification. Some evidence indicates that midlife depression may be an AD risk factor, while a chronic course of depression in late life may be a precursor to or symptom of dementia. Recently, multiple pathophysiological mechanisms have been proposed to underlie the bidirectional relationship between depression and AD, including genetic predisposition, immune dysregulation, accumulation of AD-related biomarkers (e.g., amyloid-β and tau), and alterations in brain structure. Accordingly, numerous therapeutic approaches, such as pharmacology treatments, psychotherapy, and lifestyle interventions, have been suggested as potential means of interfering with these pathways. However, the current literature on this topic remains fragmented and lacks a comprehensive review characterizing the association between depression and AD. In this review, we aim to address these gaps by providing an overview of the co-occurrence and temporal relationship between depression and AD, as well as exploring their underlying mechanisms. We also examine the current therapeutic regimens for depression and their implications for AD management and outline key challenges facing the field.
Collapse
Affiliation(s)
- Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Han Gan
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Fisher DW, Dunn JT, Dong H. Distinguishing features of depression in dementia from primary psychiatric disease. DISCOVER MENTAL HEALTH 2024; 4:3. [PMID: 38175420 PMCID: PMC10767128 DOI: 10.1007/s44192-023-00057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
Depression is a common and devastating neuropsychiatric symptom in the elderly and in patients with dementia. In particular, nearly 80% of patients with Alzheimer's Disease dementia experience depression during disease development and progression. However, it is unknown whether the depression in patients with dementia shares the same molecular mechanisms as depression presenting as primary psychiatric disease or occurs and persists through alternative mechanisms. In this review, we discuss how the clinical presentation and treatment differ between depression in dementia and as a primary psychiatric disease, with a focus on major depressive disorder. Then, we hypothesize several molecular mechanisms that may be unique to depression in dementia such as neuropathological changes, inflammation, and vascular events. Finally, we discuss existing issues and future directions for investigation and treatment of depression in dementia.
Collapse
Affiliation(s)
- Daniel W Fisher
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, 1959 NE Pacific Street, Box 356560, Seattle, WA, 98195, USA
| | - Jeffrey T Dunn
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA.
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA.
| |
Collapse
|
8
|
Reynolds RH, Wagen AZ, Lona-Durazo F, Scholz SW, Shoai M, Hardy J, Gagliano Taliun SA, Ryten M. Local genetic correlations exist among neurodegenerative and neuropsychiatric diseases. NPJ Parkinsons Dis 2023; 9:70. [PMID: 37117178 PMCID: PMC10147945 DOI: 10.1038/s41531-023-00504-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 03/27/2023] [Indexed: 04/30/2023] Open
Abstract
Genetic correlation ([Formula: see text]) between traits can offer valuable insight into underlying shared biological mechanisms. Neurodegenerative diseases overlap neuropathologically and often manifest comorbid neuropsychiatric symptoms. However, global [Formula: see text] analyses show minimal [Formula: see text] among neurodegenerative and neuropsychiatric diseases. Importantly, local [Formula: see text] s can exist in the absence of global relationships. To investigate this possibility, we applied LAVA, a tool for local [Formula: see text] analysis, to genome-wide association studies of 3 neurodegenerative diseases (Alzheimer's disease, Lewy body dementia and Parkinson's disease) and 3 neuropsychiatric disorders (bipolar disorder, major depressive disorder and schizophrenia). We identified several local [Formula: see text] s missed in global analyses, including between (i) all 3 neurodegenerative diseases and schizophrenia and (ii) Alzheimer's and Parkinson's disease. For those local [Formula: see text] s identified in genomic regions containing disease-implicated genes, such as SNCA, CLU and APOE, incorporation of expression quantitative trait loci identified genes that may drive genetic overlaps between diseases. Collectively, we demonstrate that complex genetic relationships exist among neurodegenerative and neuropsychiatric diseases, highlighting putative pleiotropic genomic regions and genes. These findings imply sharing of pathogenic processes and the potential existence of common therapeutic targets.
Collapse
Affiliation(s)
- Regina H Reynolds
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Aaron Z Wagen
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, London, UK
- Neurodegeneration Biology Laboratory, The Francis Crick Institute, London, UK
| | - Frida Lona-Durazo
- Montréal Heart Institute, Montréal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Maryam Shoai
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Neurodegenerative Diseases, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - John Hardy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Neurodegenerative Diseases, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Sarah A Gagliano Taliun
- Montréal Heart Institute, Montréal, QC, Canada
- Department of Medicine & Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Mina Ryten
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK.
| |
Collapse
|
9
|
Pan AL, Audrain M, Sakakibara E, Joshi R, Zhu X, Wang Q, Wang M, Beckmann ND, Schadt EE, Gandy S, Zhang B, Ehrlich ME, Salton SR. Dual-Specificity Protein Phosphatase 4 (DUSP4) Overexpression Improves Learning Behavior Selectively in Female 5xFAD Mice, and Reduces β-Amyloid Load in Males and Females. Cells 2022; 11:3880. [PMID: 36497141 PMCID: PMC9737364 DOI: 10.3390/cells11233880] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Recent multiscale network analyses of banked brains from subjects who died of late-onset sporadic Alzheimer's disease converged on VGF (non-acronymic) as a key hub or driver. Within this computational VGF network, we identified the dual-specificity protein phosphatase 4 (DUSP4) [also known as mitogen-activated protein kinase (MAPK) phosphatase 2] as an important node. Importantly, DUSP4 gene expression, like that of VGF, is downregulated in postmortem Alzheimer's disease (AD) brains. We investigated the roles that this VGF/DUSP4 network plays in the development of learning behavior impairment and neuropathology in the 5xFAD amyloidopathy mouse model. We found reductions in DUSP4 expression in the hippocampi of male AD subjects, correlating with increased CDR scores, and in 4-month-old female and 12-18-month-old male 5xFAD hippocampi. Adeno-associated virus (AAV5)-mediated overexpression of DUSP4 in 5xFAD mouse dorsal hippocampi (dHc) rescued impaired Barnes maze performance in females but not in males, while amyloid loads were reduced in both females and males. Bulk RNA sequencing of the dHc from 5-month-old mice overexpressing DUSP4, and Ingenuity Pathway and Enrichr analyses of differentially expressed genes (DEGs), revealed that DUSP4 reduced gene expression in female 5xFAD mice in neuroinflammatory, interferon-gamma (IFNγ), programmed cell death protein-ligand 1/programmed cell death protein 1 (PD-L1/PD-1), and extracellular signal-regulated kinase (ERK)/MAPK pathways, via which DUSP4 may modulate AD phenotype with gender-specificity.
Collapse
Affiliation(s)
- Allen L. Pan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Mickael Audrain
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Emmy Sakakibara
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Rajeev Joshi
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Xiaodong Zhu
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Qian Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Noam D. Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Eric E. Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Psychiatry and Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Stephen R. Salton
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
10
|
Gillespie NA, Gentry AE, Kirkpatrick RM, Reynolds CA, Mathur R, Kendler KS, Maes HH, Webb BT, Peterson RE. Determining the stability of genome-wide factors in BMI between ages 40 to 69 years. PLoS Genet 2022; 18:e1010303. [PMID: 35951648 PMCID: PMC9398001 DOI: 10.1371/journal.pgen.1010303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 08/23/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Genome-wide association studies (GWAS) have successfully identified common variants associated with BMI. However, the stability of aggregate genetic variation influencing BMI from midlife and beyond is unknown. By analysing 165,717 men and 193,073 women from the UKBiobank, we performed BMI GWAS on six independent five-year age intervals between 40 and 72 years. We then applied genomic structural equation modeling to test competing hypotheses regarding the stability of genetic effects for BMI. LDSR genetic correlations between BMI assessed between ages 40 to 73 were all very high and ranged 0.89 to 1.00. Genomic structural equation modeling revealed that molecular genetic variance in BMI at each age interval could not be explained by the accumulation of any age-specific genetic influences or autoregressive processes. Instead, a common set of stable genetic influences appears to underpin genome-wide variation in BMI from middle to early old age in men and women alike.
Collapse
Affiliation(s)
- Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Amanda Elswick Gentry
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Robert M. Kirkpatrick
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Chandra A. Reynolds
- Department of Psychology, University of California, Riverside, California, United States of America
| | - Ravi Mathur
- GenOmics, Bioinformatics, and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, North Carolina, United States of America
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Hermine H. Maes
- Virginia Institute for Psychiatric and Behavior Genetics, Departments of Human and Molecular Genetics, Psychiatry, & Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Bradley T. Webb
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- GenOmics, Bioinformatics, and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, North Carolina, United States of America
| | - Roseann E. Peterson
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
11
|
Joshi R, Salton SRJ. Neurotrophin Crosstalk in the Etiology and Treatment of Neuropsychiatric and Neurodegenerative Disease. Front Mol Neurosci 2022; 15:932497. [PMID: 35909451 PMCID: PMC9335126 DOI: 10.3389/fnmol.2022.932497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/23/2022] [Indexed: 12/27/2022] Open
Abstract
This article reviews the current progress in our understanding of the mechanisms by which growth factors, including brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF), and select neurotrophin-regulated gene products, such as VGF (non-acronymic) and VGF-derived neuropeptides, function in the central nervous system (CNS) to modulate neuropsychiatric and neurodegenerative disorders, with a discussion of the possible therapeutic applications of these growth factors to major depressive disorder (MDD) and Alzheimer’s disease (AD). BDNF and VEGF levels are generally decreased regionally in the brains of MDD subjects and in preclinical animal models of depression, changes that are associated with neuronal atrophy and reduced neurogenesis, and are reversed by conventional monoaminergic and novel ketamine-like antidepressants. Downstream of neurotrophins and their receptors, VGF was identified as a nerve growth factor (NGF)- and BDNF-inducible secreted protein and neuropeptide precursor that is produced and trafficked throughout the CNS, where its expression is greatly influenced by neuronal activity and exercise, and where several VGF-derived peptides modulate neuronal activity, function, proliferation, differentiation, and survival. Moreover, levels of VGF are reduced in the CSF of AD subjects, where it has been repetitively identified as a disease biomarker, and in the hippocampi of subjects with MDD, suggesting possible shared mechanisms by which reduced levels of VGF and other proteins that are similarly regulated by neurotrophin signaling pathways contribute to and potentially drive the pathogenesis and progression of co-morbid neuropsychiatric and neurodegenerative disorders, particularly MDD and AD, opening possible therapeutic windows.
Collapse
Affiliation(s)
- Rajeev Joshi
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Stephen R. J. Salton
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, United States
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Stephen R. J. Salton,
| |
Collapse
|
12
|
Harerimana NV, Liu Y, Gerasimov ES, Duong D, Beach TG, Reiman EM, Schneider JA, Boyle P, Lori A, Bennett DA, Lah JJ, Levey AI, Seyfried NT, Wingo TS, Wingo AP. Genetic Evidence Supporting a Causal Role of Depression in Alzheimer's Disease. Biol Psychiatry 2022; 92:25-33. [PMID: 35177243 PMCID: PMC9200901 DOI: 10.1016/j.biopsych.2021.11.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 11/04/2021] [Accepted: 11/26/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Depression has been associated with a higher risk of Alzheimer's disease (AD) in several prospective studies; however, mechanisms underlying this association remain unclear. METHODS We examined genetic correlation between depression and AD using linkage disequilibrium score regression. We then tested for evidence of causality between depression and AD using Mendelian randomization and genome-wide association study results. Subsequently, cis and trans quantitative trait locus analyses for the depression genome-wide association study signals were performed to resolve the genetic signals to specific DNA methylation sites, brain transcripts, and proteins. These transcripts and proteins were then examined for associations with AD and its endophenotypes. Finally, the associations between depression polygenic risk score and AD endophenotypes were examined. RESULTS We detected a significant genetic correlation between depression and AD, suggesting that they have a shared genetic basis. Furthermore, we found that depression had a causal role in AD through Mendelian randomization but did not find evidence for a causal role of AD on depression. Moreover, we identified 75 brain transcripts and 28 brain proteins regulated by the depression genome-wide association study signals through quantitative trait locus analyses. Of these, 46 transcripts and seven proteins were associated with rates of cognitive decline over time, AD pathologies, and AD diagnosis in two separate cohorts, thus implicating them in AD. In addition, we found that a higher depression polygenic risk score was associated with a faster decline of episodic memory over time. CONCLUSIONS Depression appears to have a causal role in AD, and this causal relationship is likely driven, in part, by the 53 brain transcripts and proteins identified in this study.
Collapse
Affiliation(s)
- Nadia V Harerimana
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Yue Liu
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | | | - Duc Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia
| | | | - Eric M Reiman
- Banner Alzheimer's Institute, Arizona State University and University of Arizona, Phoenix, Arizona
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Patricia Boyle
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Adriana Lori
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia
| | - Thomas S Wingo
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia; Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia.
| | - Aliza P Wingo
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia; Division of Mental Health, Atlanta VA Medical Center, Decatur, Georgia.
| |
Collapse
|
13
|
Faborode OS, Dalle E, Mabandla MV. Inescapable footshocks induce molecular changes in the prefrontal cortex of rats in an amyloid-beta-42 model of Alzheimer's disease. Behav Brain Res 2022; 419:113679. [PMID: 34826515 DOI: 10.1016/j.bbr.2021.113679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) affects several brain areas, including the prefrontal cortex (PFC) involved in execution, working memory, and fear extinction. Despite these critical roles, the PFC is understudied in AD pathology. People with post-traumatic stress disorder (PTSD) have twice the risk of developing AD, and the underlying mechanisms linking these two diseases are less understood. Here, we investigated the effect of footshock stress on behavioural vis-a-vis molecular changes in the PFC of an amyloid-beta (Aβ)-42 lesion rat model of AD. Trauma-like conditions were induced by exposing the animals to several footshocks. AD-like condition was induced via intra-hippocampal injection of Aβ-42 peptide. Following Aβ-42 injections, animals were tested for behavioural changes using the Open Field Test (OFT) and Y-maze test. The PFC was later harvested for neurochemical analyses. Our results showed an interactive effect of footshocks and Aβ-42 lesion on: reduced percentage alternation in the Y-maze test, suggesting memory impairment; reduced number of line crosses and time spent in the centre square of the OFT, indicating anxiogenic responses. Similarly, there was an interactive effect of footshocks and Aβ-42 lesion on: increased FK506 binding protein 51 (FKBP5) expression, which can be associated with stress-induced anxiogenic behaviours; and increased neuronal apoptosis in the PFC of the animals. In addition, footshocks, as well as Aβ-42 lesion, reduced superoxide dismutase levels and Bridging Integrator-1 (BIN1) expression in the PFC of the animals, which can be linked to the observed memory impairment. In conclusion, our findings indicate that footshocks exaggerate PFC-associated behavioural and molecular changes induced by an AD-like pathology.
Collapse
MESH Headings
- Alzheimer Disease/chemically induced
- Alzheimer Disease/etiology
- Alzheimer Disease/metabolism
- Alzheimer Disease/physiopathology
- Amyloid beta-Peptides/pharmacology
- Animals
- Anxiety/chemically induced
- Anxiety/etiology
- Anxiety/metabolism
- Anxiety/physiopathology
- Apoptosis/drug effects
- Apoptosis/physiology
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Disease Models, Animal
- Electroshock
- Male
- Memory Disorders/chemically induced
- Memory Disorders/etiology
- Memory Disorders/metabolism
- Memory Disorders/physiopathology
- Memory, Short-Term/drug effects
- Memory, Short-Term/physiology
- Peptide Fragments/pharmacology
- Prefrontal Cortex/metabolism
- Prefrontal Cortex/physiopathology
- Rats
- Rats, Sprague-Dawley
- Stress Disorders, Post-Traumatic/chemically induced
- Stress Disorders, Post-Traumatic/etiology
- Stress Disorders, Post-Traumatic/metabolism
- Stress Disorders, Post-Traumatic/physiopathology
- Tacrolimus Binding Proteins/metabolism
Collapse
Affiliation(s)
- Oluwaseun Samuel Faborode
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa.
| | - Ernest Dalle
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa.
| | - Musa Vuyisile Mabandla
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa.
| |
Collapse
|
14
|
Wang H, Alda M, Trappenberg T, Nunes A. A scoping review and comparison of approaches for measuring genetic heterogeneity in psychiatric disorders. Psychiatr Genet 2022; 32:1-8. [PMID: 34694248 DOI: 10.1097/ypg.0000000000000304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
An improved understanding of genetic etiological heterogeneity in a psychiatric condition may help us (a) isolate a neurophysiological 'final common pathway' by identifying its upstream genetic origins and (b) facilitate characterization of the condition's phenotypic variation. This review aims to identify existing genetic heterogeneity measurements in the psychiatric literature and provides a conceptual review of their mechanisms, limitations, and assumptions. The Scopus database was searched for studies that quantified genetic heterogeneity or correlation of psychiatric phenotypes with human genetic data. Ninety studies were included. Eighty-seven reports quantified genetic correlation, five applied genomic structural equation modelling, three evaluated departure from the Hardy-Weinberg equilibrium at one or more loci, and two applied a novel approach known as MiXeR. We found no study that rigorously measured genetic etiological heterogeneity across a large number of markers. Developing such approaches may help better characterize the biological diversity of psychopathology.
Collapse
Affiliation(s)
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Abraham Nunes
- Faculty of Computer Science
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
15
|
Joshi P, Perni M, Limbocker R, Mannini B, Casford S, Chia S, Habchi J, Labbadia J, Dobson CM, Vendruscolo M. Two human metabolites rescue a C. elegans model of Alzheimer's disease via a cytosolic unfolded protein response. Commun Biol 2021; 4:843. [PMID: 34234268 PMCID: PMC8263720 DOI: 10.1038/s42003-021-02218-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Age-related changes in cellular metabolism can affect brain homeostasis, creating conditions that are permissive to the onset and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Although the roles of metabolites have been extensively studied with regard to cellular signaling pathways, their effects on protein aggregation remain relatively unexplored. By computationally analysing the Human Metabolome Database, we identified two endogenous metabolites, carnosine and kynurenic acid, that inhibit the aggregation of the amyloid beta peptide (Aβ) and rescue a C. elegans model of Alzheimer's disease. We found that these metabolites act by triggering a cytosolic unfolded protein response through the transcription factor HSF-1 and downstream chaperones HSP40/J-proteins DNJ-12 and DNJ-19. These results help rationalise previous observations regarding the possible anti-ageing benefits of these metabolites by providing a mechanism for their action. Taken together, our findings provide a link between metabolite homeostasis and protein homeostasis, which could inspire preventative interventions against neurodegenerative disorders.
Collapse
Affiliation(s)
- Priyanka Joshi
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK ,grid.47840.3f0000 0001 2181 7878Present Address: The California Institute for Quantitative Biosciences (QB3-Berkeley), University of California, Berkeley, CA USA
| | - Michele Perni
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Ryan Limbocker
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK ,grid.419884.80000 0001 2287 2270Present Address: Department of Chemistry and Life Science, United States Military Academy, West Point, NY USA
| | - Benedetta Mannini
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Sam Casford
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Sean Chia
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Johnny Habchi
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Johnathan Labbadia
- grid.83440.3b0000000121901201Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Christopher M. Dobson
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Michele Vendruscolo
- grid.5335.00000000121885934Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Monereo-Sánchez J, Schram MT, Frei O, O’Connell K, Shadrin AA, Smeland OB, Westlye LT, Andreassen OA, Kaufmann T, Linden DEJ, van der Meer D. Genetic Overlap Between Alzheimer's Disease and Depression Mapped Onto the Brain. Front Neurosci 2021; 15:653130. [PMID: 34290577 PMCID: PMC8288283 DOI: 10.3389/fnins.2021.653130] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Alzheimer's disease (AD) and depression are debilitating brain disorders that are often comorbid. Shared brain mechanisms have been implicated, yet findings are inconsistent, reflecting the complexity of the underlying pathophysiology. As both disorders are (partly) heritable, characterising their genetic overlap may provide aetiological clues. While previous studies have indicated negligible genetic correlations, this study aims to expose the genetic overlap that may remain hidden due to mixed directions of effects. Methods: We applied Gaussian mixture modelling, through MiXeR, and conjunctional false discovery rate (cFDR) analysis, through pleioFDR, to genome-wide association study (GWAS) summary statistics of AD (n = 79,145) and depression (n = 450,619). The effects of identified overlapping loci on AD and depression were tested in 403,029 participants of the UK Biobank (UKB) (mean age 57.21, 52.0% female), and mapped onto brain morphology in 30,699 individuals with brain MRI data. Results: MiXer estimated 98 causal genetic variants overlapping between the 2 disorders, with 0.44 concordant directions of effects. Through pleioFDR, we identified a SNP in the TMEM106B gene, which was significantly associated with AD (B = -0.002, p = 9.1 × 10-4) and depression (B = 0.007, p = 3.2 × 10-9) in the UKB. This SNP was also associated with several regions of the corpus callosum volume anterior (B > 0.024, p < 8.6 × 10-4), third ventricle volume ventricle (B = -0.025, p = 5.0 × 10-6), and inferior temporal gyrus surface area (B = 0.017, p = 5.3 × 10-4). Discussion: Our results indicate there is substantial genetic overlap, with mixed directions of effects, between AD and depression. These findings illustrate the value of biostatistical tools that capture such overlap, providing insight into the genetic architectures of these disorders.
Collapse
Affiliation(s)
- Jennifer Monereo-Sánchez
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Miranda T. Schram
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Internal Medicine, School for Cardiovascular Disease (CARIM), Maastricht University, Maastricht, Netherlands
- Heart and Vascular Centre, Maastricht University Medical Center, Maastricht, Netherlands
| | - Oleksandr Frei
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Informatics, Centre for Bioinformatics, University of Oslo, Oslo, Norway
| | - Kevin O’Connell
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A. Shadrin
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav B. Smeland
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars T. Westlye
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Tobias Kaufmann
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - David E. J. Linden
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Dennis van der Meer
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Pomara N, Imbimbo BP. Brain Amyloid Deposition in Late-Life Depression. Biol Psychiatry 2021; 89:e41-e42. [PMID: 33189332 DOI: 10.1016/j.biopsych.2020.07.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Nunzio Pomara
- Division of Geriatric Psychiatry, Nathan Kline Institute, Orangeburg, New York; Department of Psychiatry and Pathology, NYU Grossman School of Medicine, New York, New York.
| | | |
Collapse
|
18
|
Pirolli NH, Bentley WE, Jay SM. Bacterial Extracellular Vesicles and the Gut-Microbiota Brain Axis: Emerging Roles in Communication and Potential as Therapeutics. Adv Biol (Weinh) 2021; 5:e2000540. [PMID: 33857347 DOI: 10.1002/adbi.202000540] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/24/2021] [Indexed: 12/20/2022]
Abstract
Bacterial extracellular vesicles (BEVs) have emerged as candidate signaling vectors for long-distance interkingdom communication within the gut-microbiota brain axis. Most bacteria release these nanosized vesicles, capable of signaling to the brain via their abundant protein and small RNA cargo, possibly directly via crossing the blood-brain barrier. BEVs have been shown to regulate brain gene expression and induce pathology at most stages of neuroinflammation and neurodegeneration, and thus they may play a causal role in diseases such as Alzheimer's, Parkinson's, and depression/anxiety. On the other hand, BEVs have intrinsic therapeutic properties that may be relevant to probiotic therapy and can also be engineered to function as drug delivery vehicles and vaccines. Thus, BEVs may be both a cause of and solution to neuropathological conditions. In this review, current knowledge of the physiological roles of BEVs as well as state of the art pertaining to the development of therapeutic BEVs in the context of the microbiome-gut-brain axis are summarized.
Collapse
Affiliation(s)
- Nicholas H Pirolli
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD, 20742, USA
| | - William E Bentley
- Fischell Department of Bioengineering, Robert E. Fischell Institute, and Institute for Bioscience and Biotechnology Research, University of Maryland, 5120A A. James Clark Hall, College Park, MD, 20742, USA
| | - Steven M Jay
- Fischell Department of Bioengineering and Program in Molecular and Cell Biology, University of Maryland, 3116 A. James Clark Hall, College Park, MD, 20742, USA
| |
Collapse
|
19
|
Santiago JA, Potashkin JA. The Impact of Disease Comorbidities in Alzheimer's Disease. Front Aging Neurosci 2021; 13:631770. [PMID: 33643025 PMCID: PMC7906983 DOI: 10.3389/fnagi.2021.631770] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
A wide range of comorbid diseases is associated with Alzheimer's disease (AD), the most common neurodegenerative disease worldwide. Evidence from clinical and molecular studies suggest that chronic diseases, including diabetes, cardiovascular disease, depression, and inflammatory bowel disease, may be associated with an increased risk of AD in different populations. Disruption in several shared biological pathways has been proposed as the underlying mechanism for the association between AD and these comorbidities. Notably, inflammation is a common dysregulated pathway shared by most of the comorbidities associated with AD. Some drugs commonly prescribed to patients with diabetes and cardiovascular disease have shown promising results in AD patients. Systems-based biology studies have identified common genetic factors and dysregulated pathways that may explain the relationship of comorbid disorders in AD. Nonetheless, the precise mechanisms for the occurrence of disease comorbidities in AD are not entirely understood. Here, we discuss the impact of the most common comorbidities in the clinical management of AD patients.
Collapse
Affiliation(s)
| | - Judith A Potashkin
- Cellular and Molecular Pharmacology Department, Center for Neurodegenerative Diseases and Therapeutics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
20
|
Zhou X, Li YYT, Fu AKY, Ip NY. Polygenic Score Models for Alzheimer's Disease: From Research to Clinical Applications. Front Neurosci 2021; 15:650220. [PMID: 33854414 PMCID: PMC8039467 DOI: 10.3389/fnins.2021.650220] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
The high prevalence of Alzheimer's disease (AD) among the elderly population and its lack of effective treatments make this disease a critical threat to human health. Recent epidemiological and genetics studies have revealed the polygenic nature of the disease, which is possibly explainable by a polygenic score model that considers multiple genetic risks. Here, we systemically review the rationale and methods used to construct polygenic score models for studying AD. We also discuss the associations of polygenic risk scores (PRSs) with clinical outcomes, brain imaging findings, and biochemical biomarkers from both the brain and peripheral system. Finally, we discuss the possibility of incorporating polygenic score models into research and clinical practice along with potential challenges.
Collapse
Affiliation(s)
- Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, China
| | - Yolanda Y. T. Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Amy K. Y. Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, China
| | - Nancy Y. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, China
- *Correspondence: Nancy Y. Ip,
| |
Collapse
|
21
|
Huang J, Zuber V, Matthews PM, Elliott P, Tzoulaki J, Dehghan A. Sleep, major depressive disorder, and Alzheimer disease: A Mendelian randomization study. Neurology 2020; 95:e1963-e1970. [PMID: 32817390 PMCID: PMC7682841 DOI: 10.1212/wnl.0000000000010463] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 04/23/2020] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE To explore the causal relationships between sleep, major depressive disorder (MDD), and Alzheimer disease (AD). METHODS We conducted bidirectional 2-sample Mendelian randomization analyses. Genetic associations were obtained from the largest genome-wide association studies currently available in UK Biobank (n = 446,118), Psychiatric Genomics Consortium (n = 18,759), and International Genomics of Alzheimer's Project (n = 63,926). We used the inverse variance-weighted Mendelian randomization method to estimate causal effects and weighted median and Mendelian randomization-Egger for sensitivity analyses to test for pleiotropic effects. RESULTS We found that higher risk of AD was significantly associated with being a "morning person" (odds ratio [OR] 1.01, p = 0.001), shorter sleep duration (self-reported: β = -0.006, p = 1.9 × 10-4; accelerometer based: β = -0.015, p = 6.9 × 10-5), less likely to report long sleep (β = -0.003, p = 7.3 × 10-7), earlier timing of the least active 5 hours (β = -0.024, p = 1.7 × 10-13), and a smaller number of sleep episodes (β = -0.025, p = 5.7 × 10-14) after adjustment for multiple comparisons. We also found that higher risk of AD was associated with lower risk of insomnia (OR 0.99, p = 7 × 10-13). However, we did not find evidence that these abnormal sleep patterns were causally related to AD or for a significant causal relationship between MDD and risk of AD. CONCLUSION We found that AD may causally influence sleep patterns. However, we did not find evidence supporting a causal role of disturbed sleep patterns for AD or evidence for a causal relationship between MDD and AD.
Collapse
Affiliation(s)
- Jian Huang
- From the MRC Centre for Environment and Health (J.H., V.Z., P.E., J.T., A.D.), Department of Epidemiology and Biostatistics, School of Public Health, St. Mary's Campus, Imperial College London, Norfolk Place; UK Dementia Research Institute at Imperial College London (J.H., P.M.M., J.T., A.D.); Imperial College NIHR Biomedical Research Centre (J.H., P.E.); Department of Brain Sciences (P.M.M., P.E.), Faculty of Medicine, Imperial College London; Health Data Research UK-London; and Department of Hygiene and Epidemiology (P.E., J.T.), University of Ioannina Medical School, Greece
| | - Verena Zuber
- From the MRC Centre for Environment and Health (J.H., V.Z., P.E., J.T., A.D.), Department of Epidemiology and Biostatistics, School of Public Health, St. Mary's Campus, Imperial College London, Norfolk Place; UK Dementia Research Institute at Imperial College London (J.H., P.M.M., J.T., A.D.); Imperial College NIHR Biomedical Research Centre (J.H., P.E.); Department of Brain Sciences (P.M.M., P.E.), Faculty of Medicine, Imperial College London; Health Data Research UK-London; and Department of Hygiene and Epidemiology (P.E., J.T.), University of Ioannina Medical School, Greece
| | - Paul M Matthews
- From the MRC Centre for Environment and Health (J.H., V.Z., P.E., J.T., A.D.), Department of Epidemiology and Biostatistics, School of Public Health, St. Mary's Campus, Imperial College London, Norfolk Place; UK Dementia Research Institute at Imperial College London (J.H., P.M.M., J.T., A.D.); Imperial College NIHR Biomedical Research Centre (J.H., P.E.); Department of Brain Sciences (P.M.M., P.E.), Faculty of Medicine, Imperial College London; Health Data Research UK-London; and Department of Hygiene and Epidemiology (P.E., J.T.), University of Ioannina Medical School, Greece
| | - Paul Elliott
- From the MRC Centre for Environment and Health (J.H., V.Z., P.E., J.T., A.D.), Department of Epidemiology and Biostatistics, School of Public Health, St. Mary's Campus, Imperial College London, Norfolk Place; UK Dementia Research Institute at Imperial College London (J.H., P.M.M., J.T., A.D.); Imperial College NIHR Biomedical Research Centre (J.H., P.E.); Department of Brain Sciences (P.M.M., P.E.), Faculty of Medicine, Imperial College London; Health Data Research UK-London; and Department of Hygiene and Epidemiology (P.E., J.T.), University of Ioannina Medical School, Greece
| | - Joanna Tzoulaki
- From the MRC Centre for Environment and Health (J.H., V.Z., P.E., J.T., A.D.), Department of Epidemiology and Biostatistics, School of Public Health, St. Mary's Campus, Imperial College London, Norfolk Place; UK Dementia Research Institute at Imperial College London (J.H., P.M.M., J.T., A.D.); Imperial College NIHR Biomedical Research Centre (J.H., P.E.); Department of Brain Sciences (P.M.M., P.E.), Faculty of Medicine, Imperial College London; Health Data Research UK-London; and Department of Hygiene and Epidemiology (P.E., J.T.), University of Ioannina Medical School, Greece
| | - Abbas Dehghan
- From the MRC Centre for Environment and Health (J.H., V.Z., P.E., J.T., A.D.), Department of Epidemiology and Biostatistics, School of Public Health, St. Mary's Campus, Imperial College London, Norfolk Place; UK Dementia Research Institute at Imperial College London (J.H., P.M.M., J.T., A.D.); Imperial College NIHR Biomedical Research Centre (J.H., P.E.); Department of Brain Sciences (P.M.M., P.E.), Faculty of Medicine, Imperial College London; Health Data Research UK-London; and Department of Hygiene and Epidemiology (P.E., J.T.), University of Ioannina Medical School, Greece.
| |
Collapse
|
22
|
Thayer TE, Levinson RT, Huang S, Assad T, Farber-Eger E, Wells QS, Mosley JD, Brittain EL. BMI Is Causally Associated With Pulmonary Artery Pressure But Not Hemodynamic Evidence of Pulmonary Vascular Remodeling. Chest 2020; 159:302-310. [PMID: 32712226 DOI: 10.1016/j.chest.2020.07.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND There is an unclear relationship of obesity to the pathogenesis and severity of pulmonary arterial hypertension (PAH) and pulmonary venous hypertension (PVH). RESEARCH QUESTION Is BMI casually associated with pulmonary artery pressure (PAP) and/or markers of pulmonary vascular remodeling? STUDY DESIGN AND METHODS The study design was a two-sample inverse-variance weighted Mendelian randomization. We constructed two BMI genetic risk scores from genome-wide association study summary data and deployed them in nonoverlapping cohorts of subjects referred for right heart catheterization (RHC) or echocardiography. A BMI highly polygenic risk score (hpGRS) optimally powered to detect shared genetic architecture of obesity with other traits was tested for association with RHC parameters including markers of pulmonary vascular remodeling. A BMI strict genetic risk score (sGRS) composed of high-confidence genetic variants was used for Mendelian randomization analyses to assess if higher BMI causes higher PAP. RESULTS Among all subjects, both directly measured BMI and hpGRS were positively associated with pulmonary arterial pressures but not markers of pulmonary vascular remodeling. Categorical analyses revealed BMI and hpGRS were associated with PVH but not PAH. Mendelian randomization of the sGRS supported that higher BMI is causal of higher systolic pulmonary artery pressure (sPAP). Sensitivity analyses showed sPAP-BMI sGRS relationship was preserved when either individuals with PAH or PVH were excluded. In the echocardiographic cohort, BMI and hpGRS were positively associated with estimated PAP and markers of left heart remodeling. INTERPRETATION BMI is a modifier of pulmonary hypertension severity in both PAH and PVH but is only involved in the pathogenesis of PVH.
Collapse
Affiliation(s)
- Timothy E Thayer
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Rebecca T Levinson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Tufik Assad
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Eric Farber-Eger
- Vanderbilt Translational and Clinical Research Center, Vanderbilt University Medical Center, Nashville, TN
| | - Quinn S Wells
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Jonathan D Mosley
- Departments of Medicine and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Evan L Brittain
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
23
|
Lutz MW, Luo S, Williamson DE, Chiba-Falek O. Shared genetic etiology underlying late-onset Alzheimer's disease and posttraumatic stress syndrome. Alzheimers Dement 2020; 16:1280-1292. [PMID: 32588970 DOI: 10.1002/alz.12128] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Late-onset Alzheimer's disease (LOAD) manifests comorbid neuropsychiatric symptoms and posttraumatic stress disorder (PTSD) is associated with an increased risk for dementia in late life, suggesting the two disorders may share genetic etiologies. METHODS We performed genetic pleiotropy analysis using LOAD and PTSD genome-wide association study (GWAS) datasets from white and African-American populations, followed by functional-genomic analyses. RESULTS We found an enrichment for LOAD across increasingly stringent levels of significance with the PTSD GWAS association (LOAD|PTSD) in the discovery and replication cohorts and a modest enrichment for the reverse conditional association (PTSD|LOAD). LOAD|PTSD association analysis identified and replicated the MS4A genes region. These genes showed similar expression pattern in brain regions affected in LOAD, and across-brain-tissue analysis identified a significant association for MS4A6A. The African-American samples showed moderate enrichment; however, no false discovery rate-significant associations. DISCUSSION We demonstrated common genetic signatures for LOAD and PTSD and suggested immune response as a common pathway for these diseases.
Collapse
Affiliation(s)
- Michael W Lutz
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | - Douglas E Williamson
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, USA.,Research Service, Durham VA Medical Center, Durham, North Carolina, USA.,Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA.,Center for Genomic and Computational Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
24
|
Bellou E, Stevenson-Hoare J, Escott-Price V. Polygenic risk and pleiotropy in neurodegenerative diseases. Neurobiol Dis 2020; 142:104953. [PMID: 32445791 PMCID: PMC7378564 DOI: 10.1016/j.nbd.2020.104953] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/12/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
In this paper we explore the phenomenon of pleiotropy in neurodegenerative diseases, focusing on Alzheimer's disease (AD). We summarize the various techniques developed to investigate pleiotropy among traits, elaborating in the polygenic risk scores (PRS) analysis. PRS was designed to assess a cumulative effect of a large number of SNPs for association with a disease and, later for disease risk prediction. Since genetic predictions rely on heritability, we discuss SNP-based heritability from genome-wide association studies and its contribution to the prediction accuracy of PRS. We review work examining pleiotropy in neurodegenerative diseases and related phenotypes and biomarkers. We conclude that the exploitation of pleiotropy may aid in the identification of novel genes and provide further insights in the disease mechanisms, and along with PRS analysis, may be advantageous for precision medicine.
Collapse
|
25
|
Brzezińska A, Bourke J, Rivera-Hernández R, Tsolaki M, Woźniak J, Kaźmierski J. Depression in Dementia or Dementia in Depression? Systematic Review of Studies and Hypotheses. Curr Alzheimer Res 2020; 17:16-28. [DOI: 10.2174/1567205017666200217104114] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/09/2020] [Accepted: 01/18/2020] [Indexed: 01/21/2023]
Abstract
The majority of research works to date suggest that Major Depressive Disorder (MDD) is a
risk factor for dementia and may predispose to cognitive decline in both early and late onset variants.
The presence of depression may not, however, reflect the cause, rather, an effect: it may be a response to
cognitive impairment or alters the threshold at which cognitive impairment might manifest or be detected.
An alternative hypothesis is that depression may be part of a prodrome to Alzheimer’s Disease
(AD), suggesting a neurobiological association rather than one of psychological response alone. Genetic
polymorphisms may explain some of the variances in shared phenomenology between the diagnoses, the
instance, when the conditions arise comorbidly, the order in which they are detected that may depend on
individual cognitive and physical reserves, as well as the medical history and individual vulnerability.
This hypothesis is biologically sound but has not been systematically investigated to date. The current
review highlights how genetic variations are involved in the development of both AD and MDD, and the
risk conferred by these variations on the expression of these two disorders comorbidly is an important
consideration for future studies of pathoaetiological mechanisms and in the stratification of study samples
for randomised controlled trials.
Collapse
Affiliation(s)
- Agnieszka Brzezińska
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - Julius Bourke
- Centre for Psychiatry, Wolfson Institute for Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London E14NS, United Kingdom
| | - Rayito Rivera-Hernández
- Department of Psychiatry, Psychology, Legal Medicine and History of Medicine, University of Salamanca, Salamanca, Spain
| | - Magda Tsolaki
- 3rd Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece, “George Papanicolaou” Hospital, Thessaloniki, Greece
| | - Joanna Woźniak
- Central Clinical Hospital of Medical University of Lodz, Lodz, Poland
| | - Jakub Kaźmierski
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
26
|
Shared genetic etiology underlying Alzheimer's disease and major depressive disorder. Transl Psychiatry 2020; 10:88. [PMID: 32152295 PMCID: PMC7062839 DOI: 10.1038/s41398-020-0769-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/14/2020] [Accepted: 02/25/2020] [Indexed: 01/22/2023] Open
Abstract
Patients with late-onset Alzheimer's disease (LOAD) frequently manifest comorbid neuropsychiatric symptoms with depression and anxiety being most frequent, and individuals with major depressive disorder (MDD) have an increased prevalence of LOAD. This suggests shared etiologies and intersecting pathways between LOAD and MDD. We performed pleiotropy analyses using LOAD and MDD GWAS data sets from the International Genomics of Alzheimer's Project (IGAP) and the Psychiatric Genomics Consortium (PGC), respectively. We found a moderate enrichment for SNPs associated with LOAD across increasingly stringent levels of significance with the MDD GWAS association (LOAD|MDD), of maximum four and eightfolds, including and excluding the APOE-region, respectively. Association analysis excluding the APOE-region identified numerous SNPs corresponding to 40 genes, 9 of which are known LOAD-risk loci primarily in chromosome 11 regions that contain the SPI1 gene and MS4A genes cluster, and others were novel pleiotropic risk-loci for LOAD conditional with MDD. The most significant associated SNPs on chromosome 11 overlapped with eQTLs found in whole-blood and monocytes, suggesting functional roles in gene regulation. The reverse conditional association analysis (MDD|LOAD) showed a moderate level, ~sevenfold, of polygenic overlap, however, no SNP showed significant association. Pathway analyses replicated previously reported LOAD biological pathways related to immune response and regulation of endocytosis. In conclusion, we provide insights into the overlapping genetic signatures underpinning the common phenotypic manifestations and inter-relationship between LOAD and MDD. This knowledge is crucial to the development of actionable targets for novel therapies to treat depression preceding dementia, in an effort to delay or ultimately prevent the onset of LOAD.
Collapse
|
27
|
Fan T, Hu Y, Xin J, Zhao M, Wang J. Analyzing the genes and pathways related to major depressive disorder via a systems biology approach. Brain Behav 2020; 10:e01502. [PMID: 31875662 PMCID: PMC7010578 DOI: 10.1002/brb3.1502] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Major depressive disorder (MDD) is a mental disorder caused by the combination of genetic, environmental, and psychological factors. Over the years, a number of genes potentially associated with MDD have been identified. However, in many cases, the role of these genes and their relationship in the etiology and development of MDD remains unclear. Under such situation, a systems biology approach focusing on the function correlation and interaction of the candidate genes in the context of MDD will provide useful information on exploring the molecular mechanisms underlying the disease. METHODS We collected genes potentially related to MDD by screening the human genetic studies deposited in PubMed (https://www.ncbi.nlm.nih.gov/pubmed). The main biological themes within the genes were explored by function and pathway enrichment analysis. Then, the interaction of genes was analyzed in the context of protein-protein interaction network and a MDD-specific network was built by Steiner minimal tree algorithm. RESULTS We collected 255 candidate genes reported to be associated with MDD from available publications. Functional analysis revealed that biological processes and biochemical pathways related to neuronal development, endocrine, cell growth and/or survivals, and immunology were enriched in these genes. The pathways could be largely grouped into three modules involved in biological procedures related to nervous system, the immune system, and the endocrine system, respectively. From the MDD-specific network, 35 novel genes potentially associated with the disease were identified. CONCLUSION By means of network- and pathway-based methods, we explored the molecular mechanism underlying the pathogenesis of MDD at a systems biology level. Results from our work could provide valuable clues for understanding the molecular features of MDD.
Collapse
Affiliation(s)
- Ting Fan
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ying Hu
- Academy of Psychology and Behavior, Tianjin Normal University, Tianjin, China
| | - Juncai Xin
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Mengwen Zhao
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| |
Collapse
|
28
|
Harrison JR, Mistry S, Muskett N, Escott-Price V. From Polygenic Scores to Precision Medicine in Alzheimer's Disease: A Systematic Review. J Alzheimers Dis 2020; 74:1271-1283. [PMID: 32250305 PMCID: PMC7242840 DOI: 10.3233/jad-191233] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Late-onset Alzheimer's disease (AD) is highly heritable. The effect of many common genetic variants, single nucleotide polymorphisms (SNPs), confer risk. Variants are clustered in areas of biology, notably immunity and inflammation, cholesterol metabolism, endocytosis, and ubiquitination. Polygenic scores (PRS), which weight the sum of an individual's risk alleles, have been used to draw inferences about the pathological processes underpinning AD. OBJECTIVE This paper aims to systematically review how AD PRS are being used to study a range of outcomes and phenotypes related to neurodegeneration. METHODS We searched the literature from July 2008-July 2018 following PRISMA guidelines. RESULTS 57 studies met criteria. The AD PRS can distinguish AD cases from controls. The ability of AD PRS to predict conversion from mild cognitive impairment (MCI) to AD was less clear. There was strong evidence of association between AD PRS and cognitive impairment. AD PRS were correlated with a number of biological phenotypes associated with AD pathology, such as neuroimaging changes and amyloid and tau measures. Pathway-specific polygenic scores were also associated with AD-related biologically relevant phenotypes. CONCLUSION PRS can predict AD effectively and are associated with cognitive impairment. There is also evidence of association between AD PRS and other phenotypes relevant to neurodegeneration. The associations between pathway specific polygenic scores and phenotypic changes may allow us to define the biology of the disease in individuals and indicate who may benefit from specific treatments. Longitudinal cohort studies are required to test the ability of PGS to delineate pathway-specific disease activity.
Collapse
Affiliation(s)
- Judith R. Harrison
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Hadyn Ellis Building, Cardiff University, Cardiff, UK
| | - Sumit Mistry
- MRC Centre for Neuropsychiatric Genetics and Genomics, Hadyn Ellis Building, Cardiff University, Cardiff, UK
| | - Natalie Muskett
- Cardiff University Medical School, University Hospital of Wales, Cardiff, UK
| | - Valentina Escott-Price
- Dementia Research Institute & the MRC Centre for Neuropsychiatric Genetics and Genomics, Hadyn Ellis Building, Cardiff University, Cardiff, UK
| |
Collapse
|
29
|
Ni H, Xu M, Zhan GL, Fan Y, Zhou H, Jiang HY, Lu WH, Tan L, Zhang DF, Yao YG, Zhang C. The GWAS Risk Genes for Depression May Be Actively Involved in Alzheimer's Disease. J Alzheimers Dis 2019; 64:1149-1161. [PMID: 30010129 DOI: 10.3233/jad-180276] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Depression is one of the most frequent psychiatric symptoms observed in people during the development of Alzheimer's disease (AD). We hypothesized that genetic factors conferring risk of depression might affect AD development. In this study, we screened 31 genes, which were located in 19 risk loci for major depressive disorder (MDD) identified by two recent large genome-wide association studies (GWAS), in AD patients at the genomic and transcriptomic levels. Association analysis of common variants was performed by using summary statistics of the International Genomics of Alzheimer's Project (IGAP), and association analysis of rare variants was conducted by sequencing the entire coding region of the 31 MDD risk genes in 107 Han Chinese patients with early-onset and/or familial AD. We also quantified the mRNA expression alterations of these MDD risk genes in brain tissues of AD patients and AD mouse models, followed by protein-protein interaction network prediction to show their potential effects in AD pathways. We found that common and rare variants of L3MBTL2 were significantly associated with AD. mRNA expression levels of 18 MDD risk genes, in particular SORCS3 and OAT, were differentially expressed in AD brain tissues. 13 MDD risk genes were predicted to physically interact with core AD genes. The involvement of HACE1, NEGR1, and SLC6A15 in AD was supported by convergent lines of evidence. Taken together, our results showed that MDD risk genes might play an active role in AD pathology and supported the notion that depression might be the "common cold" of psychiatry.
Collapse
Affiliation(s)
- Hua Ni
- Center for Disease Control and Prevention, Shanghai Xuhui Mental Health Center, Shanghai, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Gui-Lai Zhan
- Center for Disease Control and Prevention, Shanghai Xuhui Mental Health Center, Shanghai, China
| | - Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hejiang Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hong-Yan Jiang
- Department of Psychiatry, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wei-Hong Lu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liwen Tan
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, China
| | - Deng-Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan, China
| | - Chen Zhang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Xu J, Li Q, Qin W, Jun Li M, Zhuo C, Liu H, Liu F, Wang J, Schumann G, Yu C. Neurobiological substrates underlying the effect of genomic risk for depression on the conversion of amnestic mild cognitive impairment. Brain 2019; 141:3457-3471. [PMID: 30445590 DOI: 10.1093/brain/awy277] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 09/12/2018] [Indexed: 12/28/2022] Open
Abstract
Depression increases the conversion risk from amnestic mild cognitive impairment to Alzheimer's disease with unknown mechanisms. We hypothesize that the cumulative genomic risk for major depressive disorder may be a candidate cause for the increased conversion risk. Here, we aimed to investigate the predictive effect of the polygenic risk scores of major depressive disorder-specific genetic variants (PRSsMDD) on the conversion from non-depressed amnestic mild cognitive impairment to Alzheimer's disease, and its underlying neurobiological mechanisms. The PRSsMDD could predict the conversion from amnestic mild cognitive impairment to Alzheimer's disease, and amnestic mild cognitive impairment patients with high risk scores showed 16.25% higher conversion rate than those with low risk. The PRSsMDD was correlated with the left hippocampal volume, which was found to mediate the predictive effect of the PRSsMDD on the conversion of amnestic mild cognitive impairment. The major depressive disorder-specific genetic variants were mapped into genes using different strategies, and then enrichment analyses and protein-protein interaction network analysis revealed that these genes were involved in developmental process and amyloid-beta binding. They showed temporal-specific expression in the hippocampus in middle and late foetal developmental periods. Cell type-specific expression analysis of these genes demonstrated significant over-representation in the pyramidal neurons and interneurons in the hippocampus. These cross-scale neurobiological analyses and functional annotations indicate that major depressive disorder-specific genetic variants may increase the conversion from amnestic mild cognitive impairment to Alzheimer's disease by modulating the early hippocampal development and amyloid-beta binding. The PRSsMDD could be used as a complementary measure to select patients with amnestic mild cognitive impairment with high conversion risk to Alzheimer's disease.
Collapse
Affiliation(s)
- Jiayuan Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Qiaojun Li
- College of Information Engineering, Tianjin University of Commerce, Tianjin, P.R. China
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Mulin Jun Li
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Department of Pharmacology, Tianjin Medical University, Tianjin, P.R. China
| | - Chuanjun Zhuo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China.,Department of Psychiatry Functional Neuroimaging Laboratory, Tianjin Mental Health Center, Tianjin Anding Hospital, Tianjin, P.R. China
| | - Huaigui Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Gunter Schumann
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Medical Research Council Social, Genetic and Developmental Psychiatry Centre, London, UK
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, P.R. China
| |
Collapse
|
31
|
Protein misassembly and aggregation as potential convergence points for non-genetic causes of chronic mental illness. Mol Psychiatry 2019; 24:936-951. [PMID: 30089789 DOI: 10.1038/s41380-018-0133-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/10/2018] [Accepted: 06/18/2018] [Indexed: 12/13/2022]
Abstract
Chronic mental illnesses (CMI), such as schizophrenia or recurrent affective disorders, are complex conditions with both genetic and non-genetic elements. In many other chronic brain conditions, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia, sporadic instances of the disease are more common than gene-driven familial cases. Yet, the pathology of these conditions can be characterized by the presence of aberrant protein homeostasis, proteostasis, resulting in misfolded or aggregated proteins in the brains of patients that predominantly do not derive from genetic mutations. While visible deposits of aggregated protein have not yet been detected in CMI patients, we propose the existence of more subtle protein misassembly in these conditions, which form a continuum with the psychiatric phenotypes found in the early stages of many neurodegenerative conditions. Such proteinopathies need not rely on genetic variation. In a similar manner to the established aberrant neurotransmitter homeostasis in CMI, aberrant homeostasis of proteins is a functional statement that can only partially be explained by, but is certainly complementary to, genetic approaches. Here, we review evidence for aberrant proteostasis signatures from post mortem human cases, in vivo animal work, and in vitro analysis of candidate proteins misassembled in CMI. The five best-characterized proteins in this respect are currently DISC1, dysbindin-1, CRMP1, TRIOBP-1, and NPAS3. Misassembly of these proteins with inherently unstructured domains is triggered by extracellular stressors and thus provides a converging point for non-genetic causes of CMI.
Collapse
|
32
|
Chasioti D, Yan J, Nho K, Saykin AJ. Progress in Polygenic Composite Scores in Alzheimer's and Other Complex Diseases. Trends Genet 2019; 35:371-382. [PMID: 30922659 PMCID: PMC6475476 DOI: 10.1016/j.tig.2019.02.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/12/2019] [Accepted: 02/22/2019] [Indexed: 11/25/2022]
Abstract
Advances in high-throughput genotyping and next-generation sequencing (NGS) coupled with larger sample sizes brings the realization of precision medicine closer than ever. Polygenic approaches incorporating the aggregate influence of multiple genetic variants can contribute to a better understanding of the genetic architecture of many complex diseases and facilitate patient stratification. This review addresses polygenic concepts, methodological developments, hypotheses, and key issues in study design. Polygenic risk scores (PRSs) have been applied to many complex diseases and here we focus on Alzheimer's disease (AD) as a primary exemplar. This review was designed to serve as a starting point for investigators wishing to use PRSs in their research and those interested in enhancing clinical study designs through enrichment strategies.
Collapse
Affiliation(s)
- Danai Chasioti
- Department of BioHealth Informatics, Indiana University-Purdue University, Indianapolis, IN 46202, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Jingwen Yan
- Department of BioHealth Informatics, Indiana University-Purdue University, Indianapolis, IN 46202, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kwangsik Nho
- Department of BioHealth Informatics, Indiana University-Purdue University, Indianapolis, IN 46202, USA; Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Andrew J Saykin
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
33
|
Herman FJ, Simkovic S, Pasinetti GM. Neuroimmune nexus of depression and dementia: Shared mechanisms and therapeutic targets. Br J Pharmacol 2019; 176:3558-3584. [PMID: 30632147 DOI: 10.1111/bph.14569] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022] Open
Abstract
Dysfunctional immune activity is a physiological component of both Alzheimer's disease (AD) and major depressive disorder (MDD). The extent to which altered immune activity influences the development of their respective cognitive symptoms and neuropathologies remains under investigation. It is evident, however, that immune activity affects neuronal function and circuit integrity. In both disorders, alterations are present in similar immune networks and neuroendocrine signalling pathways, immune responses persist in overlapping neuroanatomical locations, and morphological and structural irregularities are noted in similar domains. Epidemiological studies have also linked the two disorders, and their genetic and environmental risk factors intersect along immune-activating pathways and can be synonymous with one another. While each of these disorders individually contains a large degree of heterogeneity, their shared immunological components may link distinct phenotypes within each disorder. This review will therefore highlight the shared immune pathways of AD and MDD, their overlapping neuroanatomical features, and previously applied, as well as novel, approaches to pharmacologically manipulate immune pathways, in each neurological condition. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Francis J Herman
- Department of Neurology, Mount Sinai School of Medicine, New York City, New York, USA
| | - Sherry Simkovic
- Department of Neurology, Mount Sinai School of Medicine, New York City, New York, USA
| | - Giulio M Pasinetti
- Department of Neurology, Mount Sinai School of Medicine, New York City, New York, USA.,Geriatrics Research. Education, and Clinical Center, JJ Peters VA Medical Center, Bronx, New York, USA
| |
Collapse
|
34
|
Pan JX, Xia JJ, Deng FL, Liang WW, Wu J, Yin BM, Dong MX, Chen JJ, Ye F, Wang HY, Zheng P, Xie P. Diagnosis of major depressive disorder based on changes in multiple plasma neurotransmitters: a targeted metabolomics study. Transl Psychiatry 2018; 8:130. [PMID: 29991685 PMCID: PMC6039504 DOI: 10.1038/s41398-018-0183-x] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/11/2018] [Accepted: 06/05/2018] [Indexed: 12/13/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating psychiatric illness. However, there is currently no objective laboratory-based diagnostic tests for this disorder. Although, perturbations in multiple neurotransmitter systems have been implicated in MDD, the biochemical changes underlying the disorder remain unclear, and a comprehensive global evaluation of neurotransmitters in MDD has not yet been performed. Here, using a GC-MS coupled with LC-MS/MS-based targeted metabolomics approach, we simultaneously quantified the levels of 19 plasma metabolites involved in GABAergic, catecholaminergic, and serotonergic neurotransmitter systems in 50 first-episode, antidepressant drug-naïve MDD subjects and 50 healthy controls to identify potential metabolite biomarkers for MDD (training set). Moreover, an independent sample cohort comprising 49 MDD patients, 30 bipolar disorder (BD) patients and 40 healthy controls (testing set) was further used to validate diagnostic generalizability and specificity of these candidate biomarkers. Among the 19 plasma neurotransmitter metabolites examined, nine were significantly changed in MDD subjects. These metabolites were mainly involved in GABAergic, catecholaminergic and serotonergic systems. The GABAergic and catecholaminergic had better diagnostic value than serotonergic pathway. A panel of four candidate plasma metabolite biomarkers (GABA, dopamine, tyramine, kynurenine) could distinguish MDD subjects from health controls with an AUC of 0.968 and 0.953 in the training and testing set, respectively. Furthermore, this panel distinguished MDD subjects from BD subjects with high accuracy. This study is the first to globally evaluate multiple neurotransmitters in MDD plasma. The altered plasma neurotransmitter metabolite profile has potential differential diagnostic value for MDD.
Collapse
Affiliation(s)
- Jun-Xi Pan
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016 China
| | - Jin-Jun Xia
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016 China
| | - Feng-Li Deng
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Wei-Wei Liang
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Jing Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Bang-Min Yin
- 0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460 China ,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Mei-Xue Dong
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Jun Chen
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Fei Ye
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China ,grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai-Yang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016 China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016 China
| | - Peng Zheng
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China. .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China. .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402460, China. .,Chongqing Key Laboratory of Neurobiology, Chongqing, 400016, China. .,Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
35
|
Griffin JWD, Liu Y, Bradshaw PC, Wang K. In Silico Preliminary Association of Ammonia Metabolism Genes GLS, CPS1, and GLUL with Risk of Alzheimer's Disease, Major Depressive Disorder, and Type 2 Diabetes. J Mol Neurosci 2018; 64:385-396. [PMID: 29441491 DOI: 10.1007/s12031-018-1035-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/31/2018] [Indexed: 12/28/2022]
Abstract
Ammonia is a toxic by-product of protein catabolism and is involved in changes in glutamate metabolism. Therefore, ammonia metabolism genes may link a range of diseases involving glutamate signaling such as Alzheimer's disease (AD), major depressive disorder (MDD), and type 2 diabetes (T2D). We analyzed data from a National Institute on Aging study with a family-based design to determine if 45 single nucleotide polymorphisms (SNPs) in glutaminase (GLS), carbamoyl phosphate synthetase 1 (CPS1), or glutamate-ammonia ligase (GLUL) genes were associated with AD, MDD, or T2D using PLINK software. HAPLOVIEW software was used to calculate linkage disequilibrium measures for the SNPs. Next, we analyzed the associated variations for potential effects on transcriptional control sites to identify possible functional effects of the SNPs. Of the SNPs that passed the quality control tests, four SNPs in the GLS gene were significantly associated with AD, two SNPs in the GLS gene were associated with T2D, and one SNP in the GLUL gene and three SNPs in the CPS1 gene were associated with MDD before Bonferroni correction. The in silico bioinformatic analysis suggested probable functional roles for six associated SNPs. Glutamate signaling pathways have been implicated in all these diseases, and other studies have detected similar brain pathologies such as cortical thinning in AD, MDD, and T2D. Taken together, these data potentially link GLS with AD, GLS with T2D, and CPS1 and GLUL with MDD and stimulate the generation of testable hypotheses that may help explain the molecular basis of pathologies shared by these disorders.
Collapse
Affiliation(s)
- Jeddidiah W D Griffin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| | - Ying Liu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Patrick C Bradshaw
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Kesheng Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|