1
|
Aumont-Rodrigue G, Picard C, Labonté A, Poirier J. Apolipoprotein B gene expression and regulation in relation to Alzheimer's disease pathophysiology. J Lipid Res 2024:100667. [PMID: 39395793 DOI: 10.1016/j.jlr.2024.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Apolipoprotein B (APOB), a receptor-binding protein present in cholesterol-rich lipoproteins, has been implicated in Alzheimer's disease (AD). High levels of APOB-containing low-density lipoproteins (LDL) are linked to the pathogenesis of both early-onset familial and late onset sporadic AD. Rare coding mutations in the APOB gene are associated with familial AD, suggesting a role for APOB-bound lipoproteins in the central nervous system. This research explores APOB gene regulation across the AD spectrum using four cohorts: BRAINEAC (elderly control brains), DBCBB (controls, AD brains), ROSMAP (controls, MCI, AD brains), and ADNI (control, MCI, AD clinical subjects). APOB protein levels, measured via mass spectrometry and ELISA, positively correlated with AD pathology indices and cognition, while APOB mRNA levels showed negative correlations. Brain APOB protein levels also correlated with cortical Aβ levels. A common coding variant in the APOB gene locus affected its expression but didn't impact AD risk or brain cholesterol concentrations, except for 24-S-hydroxycholesterol. Polymorphisms in the CYP27A1 gene, notably rs4674344, were associated with APOB protein levels. A negative correlation was observed between brain APOB gene expression and AD biomarker levels. CSF APOB correlated with Tau pathology in presymptomatic subjects, while cortical APOB was strongly associated with cortical Aβ deposition in late-stage AD. The study discusses the potential link between blood-brain barrier dysfunction and AD symptoms in relation to APOB neurobiology. Overall, APOB's involvement in lipoprotein metabolism appears to influence AD pathology across different stages of the disease.
Collapse
Affiliation(s)
- Gabriel Aumont-Rodrigue
- Douglas Mental Health University Institute, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3; McGill University, Montréal, Québec, Canada, H3A 0G4
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3; McGill University, Montréal, Québec, Canada, H3A 0G4
| |
Collapse
|
2
|
Friligkou E, Løkhammer S, Cabrera-Mendoza B, Shen J, He J, Deiana G, Zanoaga MD, Asgel Z, Pilcher A, Di Lascio L, Makharashvili A, Koller D, Tylee DS, Pathak GA, Polimanti R. Gene discovery and biological insights into anxiety disorders from a large-scale multi-ancestry genome-wide association study. Nat Genet 2024; 56:2036-2045. [PMID: 39294497 DOI: 10.1038/s41588-024-01908-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/13/2024] [Indexed: 09/20/2024]
Abstract
We leveraged information from more than 1.2 million participants, including 97,383 cases, to investigate the genetics of anxiety disorders across five continental groups. Through ancestry-specific and cross-ancestry genome-wide association studies, we identified 51 anxiety-associated loci, 39 of which were novel. In addition, polygenic risk scores derived from individuals of European descent were associated with anxiety in African, admixed American and East Asian groups. The heritability of anxiety was enriched for genes expressed in the limbic system, cerebral cortex, cerebellum, metencephalon, entorhinal cortex and brain stem. Transcriptome-wide and proteome-wide analyses highlighted 115 genes associated with anxiety through brain-specific and cross-tissue regulation. Anxiety also showed global and local genetic correlations with depression, schizophrenia and bipolar disorder and widespread pleiotropy with several physical health domains. Overall, this study expands our knowledge regarding the genetic risk and pathogenesis of anxiety disorders, highlighting the importance of investigating diverse populations and integrating multi-omics information.
Collapse
Affiliation(s)
- Eleni Friligkou
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Solveig Løkhammer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Brenda Cabrera-Mendoza
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Jie Shen
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Jun He
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Giovanni Deiana
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience, Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Mihaela Diana Zanoaga
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Zeynep Asgel
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Child and Adolescent Psychiatry, NYU Langone Health, New York Metropolitan Area, New York, NY, USA
| | - Abigail Pilcher
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Luciana Di Lascio
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- IRCCS Istituto Clinico Humanitas, Rozzano, Milan, Italy; Humanitas University, Pieve Emanuele, Milan, Italy
| | - Ana Makharashvili
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Dora Koller
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Daniel S Tylee
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Gita A Pathak
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA
| | - Renato Polimanti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Cooperative Studies Program Clinical Epidemiology Research Center (CSP-CERC), VA Connecticut Healthcare System, West Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA.
- Department of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Choi A, Zhang N, Adler CH, Beach TG, Shill HA, Driver-Dunckley E, Mehta S, Belden C, Atri A, Sabbagh MN, Caviness JN. Resting-state EEG predicts cognitive decline in a neuropathologically diagnosed longitudinal community autopsied cohort. Parkinsonism Relat Disord 2024; 128:107120. [PMID: 39236511 DOI: 10.1016/j.parkreldis.2024.107120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/02/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVE To assess correlative strengths of quantitative electroencephalography (qEEG) and visual rating scale EEG features on cognitive outcomes in only autopsied cases from the Arizona Study of Neurodegenerative Disorders (AZSAND). We hypothesized that autopsy proven Parkinson Disease will show distinct EEG features from Alzheimer's Disease prior to dementia (mild cognitive impairment). BACKGROUND Cognitive decline is debilitating across neurodegenerative diseases. Resting-state EEG analysis, including spectral power across frequency bins (qEEG), has shown significant associations with neurodegenerative disease classification and cognitive status, with autopsy confirmed diagnosis relatively lacking. METHODS Biannual EEG was analyzed from autopsied cases in AZSAND who had at least one rsEEG (>1 min eyes closed±eyes open). Analysis included global relative spectral power and a previously described visual rating scale (VRS). Linear mixed regression was performed for neuropsychological assessment and testing within 2 years of death (n = 236, 594 EEG exams) in a mixed linear regression model. RESULTS The cohort included cases with final clinicopathologic diagnoses of Parkinson's disease (n = 73), Alzheimer disease (n = 65), and tauopathy not otherwise specified (n = 56). A VRS score of 3 diffuse or frequent generalized slowing) over the study duration was associated with an increase in consensus diagnosis cognitive worsening at 4.9 (3.1) years (HR 2.02, CI 1.05-3.87). Increases in global theta power% and VRS were the most consistently associated with large regression coefficients inversely with cognitive performance measures. CONCLUSION Resting-state EEG analysis was meaningfully related to cognitive performance measures in a community-based autopsy cohort. EEG deserves further study and use as a cognitive biomarker.
Collapse
Affiliation(s)
- Alexander Choi
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr, Sun City, AZ, 85351, USA.
| | - Nan Zhang
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr, Sun City, AZ, 85351, USA
| | - Holly A Shill
- Barrow Neurological Institute, 240 W. Thomas Suite 301, Phoenix, AZ, 85013, USA
| | - Erika Driver-Dunckley
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Shyamal Mehta
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Christine Belden
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr, Sun City, AZ, 85351, USA
| | - Alireza Atri
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr, Sun City, AZ, 85351, USA
| | - Marwan N Sabbagh
- Barrow Neurological Institute, 240 W. Thomas Suite 301, Phoenix, AZ, 85013, USA
| | - John N Caviness
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| |
Collapse
|
4
|
Adler CH, Halverson M, Zhang N, Shill HA, Driver-Dunckley E, Mehta SH, Atri A, Caviness JN, Serrano GE, Shprecher DR, Belden CM, Sabbagh MN, Long K, Beach TG. Conjugal Synucleinopathies: A Clinicopathologic Study. Mov Disord 2024; 39:1212-1217. [PMID: 38597193 PMCID: PMC11260251 DOI: 10.1002/mds.29783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND While preclinical studies have shown that alpha-synuclein can spread through cell-to-cell transmission whether it can be transmitted between humans is unknown. OBJECTIVES The aim was to assess the presence of a synucleinopathy in autopsied conjugal couples. METHODS Neuropathological findings in conjugal couples were categorized as Parkinson's disease (PD), dementia with Lewy bodies (DLB), Alzheimer's disease with Lewy bodies (ADLB), incidental Lewy body disease (ILBD), or no Lewy bodies. RESULTS Ninety conjugal couples were included; the mean age of death was 88.3 years; 32 couples had no Lewy bodies; 42 couples had 1 spouse with a synucleinopathy: 10 PD, 3 DLB, 13 ADLB, and 16 ILBD; 16 couples had both spouses with a synucleinopathy: in 4 couples both spouses had PD, 1 couple had PD and DLB, 4 couples had PD and ADLB, 2 couples had PD and ILBD, 1 couple had DLB and ADLB, in 3 couples both had ADLB, and 1 couple had ADLB and ILBD. No couples had both spouses with ILBD. CONCLUSIONS This large series of 90 autopsied conjugal couples found 16 conjugal couples with synucleinopathies, suggesting transmission of synucleinopathy between spouses is unlikely. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Charles H. Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ
| | | | - Nan Zhang
- Department of Biostatistics, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ
| | | | | | | | - Alireza Atri
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | | | - Geidy E. Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ
| | | | | | | | - Kathy Long
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ
| |
Collapse
|
5
|
Poirier A, Picard C, Labonté A, Aubry I, Auld D, Zetterberg H, Blennow K, Tremblay ML, Poirier J. PTPRS is a novel marker for early Tau pathology and synaptic integrity in Alzheimer's disease. Sci Rep 2024; 14:14718. [PMID: 38926456 PMCID: PMC11208446 DOI: 10.1038/s41598-024-65104-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
We examined the role of protein tyrosine phosphatase receptor sigma (PTPRS) in the context of Alzheimer's disease and synaptic integrity. Publicly available datasets (BRAINEAC, ROSMAP, ADC1) and a cohort of asymptomatic but "at risk" individuals (PREVENT-AD) were used to explore the relationship between PTPRS and various Alzheimer's disease biomarkers. We identified that PTPRS rs10415488 variant C shows features of neuroprotection against early Tau pathology and synaptic degeneration in Alzheimer's disease. This single nucleotide polymorphism correlated with higher PTPRS transcript abundance and lower p(181)Tau and GAP-43 levels in the CSF. In the brain, PTPRS protein abundance was significantly correlated with the quantity of two markers of synaptic integrity: SNAP25 and SYT-1. We also found the presence of sexual dimorphism for PTPRS, with higher CSF concentrations in males than females. Male carriers for variant C were found to have a 10-month delay in the onset of AD. We thus conclude that PTPRS acts as a neuroprotective receptor in Alzheimer's disease. Its protective effect is most important in males, in whom it postpones the age of onset of the disease.
Collapse
Affiliation(s)
- Alexandre Poirier
- Division of Experimental Medicine, Faculty of Medicine and Health Science, McGill University, Montréal, QC, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Isabelle Aubry
- Goodman Cancer Institute, McGill University, Montréal, Canada
- McGill University, Montréal, QC, Canada
| | - Daniel Auld
- McGill University, Montréal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, QC, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, SAR, People's Republic of China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- University of Science and Technology of China, Hefei, Anhui, People's Republic of China
- Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
| | - Michel L Tremblay
- Division of Experimental Medicine, Faculty of Medicine and Health Science, McGill University, Montréal, QC, Canada.
- Goodman Cancer Institute, McGill University, Montréal, Canada.
- McGill University, Montréal, QC, Canada.
- Department of Biochemistry, McGill University, Montréal, Canada.
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, QC, Canada.
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada.
- McGill University, Montréal, QC, Canada.
| |
Collapse
|
6
|
Poirier A, Picard C, Labonté A, Aubry I, Auld D, Zetterberg H, Blennow K, Tremblay ML, Poirier J. PTPRS is a novel marker for early tau pathology and synaptic integrity in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593733. [PMID: 38766183 PMCID: PMC11100782 DOI: 10.1101/2024.05.12.593733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
We examined the role of protein tyrosine phosphatase receptor sigma (PTPRS) in the context of Alzheimer's disease and synaptic integrity. Publicly available datasets (BRAINEAC, ROSMAP, ADC1) and a cohort of asymptomatic but "at risk" individuals (PREVENT-AD) were used to explore the relationship between PTPRS and various Alzheimer's disease biomarkers. We identified that PTPRS rs10415488 variant C shows features of neuroprotection against early tau pathology and synaptic degeneration in Alzheimer's disease. This single nucleotide polymorphism correlated with higher PTPRS transcript abundance and lower P-tau181 and GAP-43 levels in the CSF. In the brain, PTPRS protein abundance was significantly correlated with the quantity of two markers of synaptic integrity: SNAP25 and SYT-1. We also found the presence of sexual dimorphism for PTPRS, with higher CSF concentrations in males than females. Male carriers for variant C were found to have a 10-month delay in the onset of AD. We thus conclude that PTPRS acts as a neuroprotective receptor in Alzheimer's disease. Its protective effect is most important in males, in whom it postpones the age of onset of the disease.
Collapse
|
7
|
Serrano GE, Walker J, Nelson C, Glass M, Arce R, Intorcia A, Cline MP, Nabaty N, Acuña A, Huppert Steed A, Sue LI, Belden C, Choudhury P, Reiman E, Atri A, Beach TG. Correlation of Presynaptic and Postsynaptic Proteins with Pathology in Alzheimer's Disease. Int J Mol Sci 2024; 25:3130. [PMID: 38542104 PMCID: PMC10970005 DOI: 10.3390/ijms25063130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 04/28/2024] Open
Abstract
Synaptic transmission is essential for nervous system function and the loss of synapses is a known major contributor to dementia. Alzheimer's disease dementia (ADD) is characterized by synaptic loss in the mesial temporal lobe and cerebral neocortex, both of which are brain areas associated with memory and cognition. The association of synaptic loss and ADD was established in the late 1980s, and it has been estimated that 30-50% of neocortical synaptic protein is lost in ADD, but there has not yet been a quantitative profiling of different synaptic proteins in different brain regions in ADD from the same individuals. Very recently, positron emission tomography (PET) imaging of synapses is being developed, accelerating the focus on the role of synaptic loss in ADD and other conditions. In this study, we quantified the densities of two synaptic proteins, the presynaptic protein Synaptosome Associated Protein 25 (SNAP25) and the postsynaptic protein postsynaptic density protein 95 (PSD95) in the human brain, using enzyme-linked immunosorbent assays (ELISA). Protein was extracted from the cingulate gyrus, hippocampus, frontal, primary visual, and entorhinal cortex from cognitively unimpaired controls, subjects with mild cognitive impairment (MCI), and subjects with dementia that have different levels of Alzheimer's pathology. SNAP25 is significantly reduced in ADD when compared to controls in the frontal cortex, visual cortex, and cingulate, while the hippocampus showed a smaller, non-significant reduction, and entorhinal cortex concentrations were not different. In contrast, all brain areas showed lower PSD95 concentrations in ADD when compared to controls without dementia, although in the hippocampus, this failed to reach significance. Interestingly, cognitively unimpaired cases with high levels of AD pathology had higher levels of both synaptic proteins in all brain regions. SNAP25 and PSD95 concentrations significantly correlated with densities of neurofibrillary tangles, amyloid plaques, and Mini Mental State Examination (MMSE) scores. Our results suggest that synaptic transmission is affected by ADD in multiple brain regions. The differences were less marked in the entorhinal cortex and the hippocampus, most likely due to a ceiling effect imposed by the very early development of neurofibrillary tangles in older people in these brain regions.
Collapse
Affiliation(s)
- Geidy E. Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Jessica Walker
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Courtney Nelson
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Michael Glass
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Richard Arce
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Anthony Intorcia
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Madison P. Cline
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Natalie Nabaty
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Amanda Acuña
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Ashton Huppert Steed
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Lucia I. Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Christine Belden
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA (P.C.)
| | - Parichita Choudhury
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA (P.C.)
| | - Eric Reiman
- The Banner Alzheimer’s Institute, Phoenix, AZ 85006, USA
| | - Alireza Atri
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA (P.C.)
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| |
Collapse
|
8
|
Friligkou E, Løkhammer S, Cabrera-Mendoza B, Shen J, He J, Deiana G, Zanoaga MD, Asgel Z, Pilcher A, Di Lascio L, Makharashvili A, Koller D, Tylee DS, Pathak GA, Polimanti R. Gene Discovery and Biological Insights into Anxiety Disorders from a Multi-Ancestry Genome-wide Association Study of >1.2 Million Participants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.14.24302836. [PMID: 38405718 PMCID: PMC10889004 DOI: 10.1101/2024.02.14.24302836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
We leveraged information from more than 1.2 million participants to investigate the genetics of anxiety disorders across five continental ancestral groups. Ancestry-specific and cross-ancestry genome-wide association studies identified 51 anxiety-associated loci, 39 of which are novel. Additionally, polygenic risk scores derived from individuals of European descent were associated with anxiety in African, Admixed-American, and East Asian groups. The heritability of anxiety was enriched for genes expressed in the limbic system, the cerebral cortex, the cerebellum, the metencephalon, the entorhinal cortex, and the brain stem. Transcriptome- and proteome-wide analyses highlighted 115 genes associated with anxiety through brain-specific and cross-tissue regulation. We also observed global and local genetic correlations with depression, schizophrenia, and bipolar disorder and putative causal relationships with several physical health conditions. Overall, this study expands the knowledge regarding the genetic risk and pathogenesis of anxiety disorders, highlighting the importance of investigating diverse populations and integrating multi-omics information.
Collapse
|
9
|
Walker JE, Oliver JC, Stewart AM, Beh ST, Arce RA, Glass MJ, Vargas DE, Qiji SH, Intorcia AJ, Borja CI, Cline MP, Hemmingsen SJ, Krupp AN, McHattie RD, Mariner MR, Lorenzini I, Aslam S, Tremblay C, Beach TG, Serrano GE. Measuring Up: A Comparison of TapeStation 4200 and Bioanalyzer 2100 as Measurement Tools for RNA Quality in Postmortem Human Brain Samples. Int J Mol Sci 2023; 24:13795. [PMID: 37762097 PMCID: PMC10531353 DOI: 10.3390/ijms241813795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
The determination of RNA integrity is a critical quality assessment tool for gene expression studies where the experiment's success is highly dependent on the sample quality. Since its introduction in 1999, the gold standard in the scientific community has been the Agilent 2100 Bioanalyzer's RNA integrity number (RIN), which uses a 1-10 value system, from 1 being the most degraded, to 10 being the most intact. In 2015, Agilent launched 4200 TapeStation's RIN equivalent, and reported a strong correlation of r2 of 0.936 and a median error < ±0.4 RIN units. To evaluate this claim, we compared the Agilent 4200 TapeStation's RIN equivalent (RINe) and DV200 to the Agilent 2100 Bioanalyzer's RIN for 183 parallel RNA samples. In our study, using RNA from a total of 183 human postmortem brain samples, we found that the RIN and RINe values only weakly correlate, with an r2 of 0.393 and an average difference of 3.2 RIN units. DV200 also only weakly correlated with RIN (r2 of 0.182) and RINe (r2 of 0.347). Finally, when applying a cut-off value of 6.5 for both metrics, we found that 95.6% of samples passed with RIN, while only 23.5% passed with RINe. Our results suggest that even though RIN (Bioanalyzer) and RINe (TapeStation) use the same 1-10 value system, they should not be used interchangeably, and cut-off values should be calculated independently.
Collapse
|
10
|
Hamsafar Y, Chen Q, Borowsky AD, Beach TG, Serrano GE, Sue LI, Adler CH, Walker DG, Dugger BN. Biochemical analyses of tau and other neuronal markers in the submandibular gland and frontal cortex across stages of Alzheimer disease. Neurosci Lett 2023; 810:137330. [PMID: 37330193 PMCID: PMC11006283 DOI: 10.1016/j.neulet.2023.137330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 02/14/2023] [Accepted: 06/05/2023] [Indexed: 06/19/2023]
Abstract
Hyperphosphorylation of the microtubule-associated protein tau is hypothesized to lead to the development of neurofibrillary tangles in select brain regions during normal aging and in Alzheimer disease (AD). The distribution of neurofibrillary tangles is staged by its involvement starting in the transentorhinal regions of the brain and in final stages progress to neocortices. However, it has also been determined neurofibrillary tangles can extend into the spinal cord and select tau species are found in peripheral tissues and this may be depended on AD disease stage. To further understand the relationships of peripheral tissues to AD, we utilized biochemical methods to evaluate protein levels of total tau and phosphorylated tau (p-tau) as well as other neuronal proteins (i.e., tyrosine hydroxylase (TH), neurofilament heavy chain (NF-H), and microtubule-associated protein 2 (MAP2)) in the submandibular gland and frontal cortex of human cases across different clinicopathological stages of AD (n = 3 criteria not met or low, n = 6 intermediate, and n = 9 high likelihood that dementia is due to AD based on National Institute on Aging-Reagan criteria). We report differential protein levels based on the stage of AD, anatomic specific tau species, as well as differences in TH and NF-H. In addition, exploratory findings were made of the high molecular weight tau species big tau that is unique to peripheral tissues. Although sample sizes were small, these findings are, to our knowledge, the first comparison of these specific protein changes in these tissues.
Collapse
Affiliation(s)
- Yamah Hamsafar
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA
| | - Qian Chen
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr., Sun City, AZ 95351, USA
| | - Geidy E Serrano
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr., Sun City, AZ 95351, USA
| | - Lucia I Sue
- Banner Sun Health Research Institute, 10515 W Santa Fe Dr., Sun City, AZ 95351, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E. Shea Blvd., Scottsdale, AZ 85259, USA
| | - Douglas G Walker
- School of Life Sciences and Biodesign Institute, Arizona State University, 1151 S. Forest Ave., Tempe, AZ 85281, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 4400 V Street, Sacramento, CA 95817, USA.
| |
Collapse
|
11
|
Walker JE, Oliver JC, Stewart AM, Beh ST, Arce RA, Glass MJ, Vargas DE, Qiji SH, Intorcia AJ, Borja CI, Cline MP, Hemmingsen SJ, Krupp AN, McHattie RD, Mariner MR, Lorenzini I, Aslam S, Tremblay C, Beach TG, Serrano GE. Measuring up: A Comparison of Tapestation 4200 and Bioanalyzer 2100 as Measurement Tools for RNA Quality in Postmortem Human Brain Samples. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.03.23291969. [PMID: 37461632 PMCID: PMC10350153 DOI: 10.1101/2023.07.03.23291969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Determining RNA integrity is a critical quality assessment tool for gene expression studies where the experiment's success is highly dependent on sample quality. Since its introduction in 1999, the gold standard in the scientific community has been the Agilent 2100 Bioanalyzer's RNA Integrity Number (RIN) which uses a 1-10 value system with 1 being the most degraded to 10 being the most intact. In 2015, Agilent launched the 4200 Tapestation's RIN equivalent and reported a strong correlation of r 2 of 0.936 and median error < ± 0.4 RIN units. To evaluate this claim, we compared the Agilent 4200 Tapestation's RIN equivalent (RINe) and DV200 to the Agilent 2100 Bioanalyzer's RIN for 183 parallel RNA samples. In our study, using RNA from a total of 183 human postmortem brain samples, we found that the RIN and RINe values only weakly correlate with an r 2 of 0.393 and an average difference of 3.2 RIN units. DV200 also only weakly correlated with RIN (r 2 of 0.182) and RINe (r 2 of 0.347). Finally, when applying a cut-off value of 6.5 for both metrics, we found that 95.6% of samples passed with RIN, while only 23.5% passed with RINe. Our results suggest that even though RIN (Bioanalyzer) and RINe (Tapestation) use the same 1-10 value system, they should not be used interchangeably, and cut-off values should be calculated independently.
Collapse
|
12
|
Tremblay C, Choudhury P, Belden CM, Goldfarb D, Lorenzini I, Beach TG, Serrano GE. The role of sex differences in depression in pathologically defined Alzheimer's disease. Front Aging Neurosci 2023; 15:1156764. [PMID: 37234269 PMCID: PMC10206015 DOI: 10.3389/fnagi.2023.1156764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Sex differences in Alzheimer's disease (AD) may contribute to disease heterogeneity and affect prevalence, risk factors, disease trajectories and outcomes. Depression impacts a large number of patients with AD and has been reported to be more prevalent in women. We aimed to better understand the interaction between sex, depression and AD neuropathology, which could have implications for detection of symptoms, earlier diagnosis, therapeutic management, and enhanced quality of life. Methods We compared 338 cases with clinicopathologically confirmed AD (46% women) to 258 control cases (50% women), without dementia, parkinsonism or a significant pathological diagnosis. Depression was assessed both, using the Hamilton Depression Scale (HAM-D), and as being reported in their medical history combined with treatment with antidepressant medication. Results In the control group, women showed a higher depression severity, and a higher proportion of women were found to meet the cut-off score for depression on the HAM-D (32 vs. 16%) and having an history of depression (33 vs. 21%), while these sex differences were not observed in AD. Further, in both groups, female sex independently predicted the presence of depression, with covariates for age and cognitive status. AD subjects had higher mean HAM-D scores, were more likely to meet cutoff scores for depression (41 vs. 24%) and have a history of depression than controls (47 vs. 27%). When comparing the increase in frequency of depression in controls versus AD, the difference was significantly greater in men (AD men - control men: 24%) than in women (AD women - control women: 9%). Although subjects with depression were more likely to have higher levels of AD neuropathology, these differences were not observed when investigating the control or AD group separately. Discussion Control women had a higher likelihood and severity of depression than control men, but this sex difference was not noted when considering only those with pathologically defined AD, emphasizing the importance of considering sex in aging studies. AD was associated with higher rates of depression and men may be more likely to report or be diagnosed with depression once they develop AD indicating the importance of more frequent depression screenings in men.
Collapse
Affiliation(s)
- Cécilia Tremblay
- Department of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, United States
| | | | | | | | | | | | | |
Collapse
|
13
|
Driver-Dunckley ED, Zhang N, Serrano GE, Dunckley NA, Sue LI, Shill HA, Mehta SH, Belden C, Tremblay C, Atri A, Adler CH, Beach TG. Low clinical sensitivity and unexpectedly high incidence for neuropathologically diagnosed progressive supranuclear palsy. J Neuropathol Exp Neurol 2023; 82:438-451. [PMID: 37040756 PMCID: PMC10117158 DOI: 10.1093/jnen/nlad025] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023] Open
Abstract
The objective of this study was to determine the prevalence, incidence, and clinical diagnostic accuracy for neuropathologically diagnosed progressive supranuclear palsy (PSP) with data from a longitudinal clinicopathological study using Rainwater criteria to define neuropathological PSP. Of 954 autopsy cases, 101 met Rainwater criteria for the neuropathologic diagnosis of PSP. Of these, 87 were termed clinicopathological PSP as they also had either dementia or parkinsonism or both. The prevalence of clinicopathologically defined PSP subjects in the entire autopsy dataset was 9.1%, while the incidence rate was estimated at 780 per 100 000 persons per year, roughly 50-fold greater than most previous clinically determined PSP incidence estimates. A clinical diagnosis of PSP was 99.6% specific but only 9.2% sensitive based on first examination, and 99.3% specific and 20.7% sensitive based on the final clinical exam. Of the clinicopathologically defined PSP cases, 35/87 (∼40%) had no form of parkinsonism at first assessment, while this decreased to 18/83 (21.7%) at final assessment. Our study confirms a high specificity but low sensitivity for the clinical diagnosis of PSP. The low clinical sensitivity for PSP is likely primarily responsible for previous underestimates of the PSP population incidence rate.
Collapse
Affiliation(s)
- Erika D Driver-Dunckley
- Department of Neurology, Parkinson’s Disease and Movement Disorders Center, Mayo Clinic, Scottsdale, Arizona, USA
| | - Nan Zhang
- Department of Quantitative Health Sciences, Section of Biostatistics, Mayo Clinic, Scottsdale, Arizona, USA
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Banner Health, Sun City, Arizona, USA
| | | | - Lucia I Sue
- Banner Sun Health Research Institute, Banner Health, Sun City, Arizona, USA
| | - Holly A Shill
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Shyamal H Mehta
- Department of Neurology, Parkinson’s Disease and Movement Disorders Center, Mayo Clinic, Scottsdale, Arizona, USA
| | - Christine Belden
- Banner Sun Health Research Institute, Banner Health, Sun City, Arizona, USA
| | - Cecilia Tremblay
- Banner Sun Health Research Institute, Banner Health, Sun City, Arizona, USA
| | - Alireza Atri
- Banner Sun Health Research Institute, Banner Health, Sun City, Arizona, USA
- Department of Neurology, Center for Mind/Brain Medicine, Brigham & Women’s Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Charles H Adler
- Department of Neurology, Parkinson’s Disease and Movement Disorders Center, Mayo Clinic, Scottsdale, Arizona, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, Banner Health, Sun City, Arizona, USA
| |
Collapse
|
14
|
Salvadó G, Ossenkoppele R, Ashton NJ, Beach TG, Serrano GE, Reiman EM, Zetterberg H, Mattsson-Carlgren N, Janelidze S, Blennow K, Hansson O. Specific associations between plasma biomarkers and postmortem amyloid plaque and tau tangle loads. EMBO Mol Med 2023; 15:e17123. [PMID: 36912178 PMCID: PMC10165361 DOI: 10.15252/emmm.202217123] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Several promising plasma biomarkers for Alzheimer's disease have been recently developed, but their neuropathological correlates have not yet been fully determined. To investigate and compare independent associations between multiple plasma biomarkers (p-tau181, p-tau217, p-tau231, Aβ42/40, GFAP, and NfL) and neuropathologic measures of amyloid and tau, we included 105 participants from the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND) with antemortem plasma samples and a postmortem neuropathological exam, 48 of whom had longitudinal p-tau217 and p-tau181. When simultaneously including plaque and tangle loads, the Aβ42/40 ratio and p-tau231 were only associated with plaques (ρAβ42/40 [95%CI] = -0.53[-0.65, -0.35], ρp-tau231 [95%CI] = 0.28[0.10, 0.43]), GFAP was only associated with tangles (ρGFAP [95%CI] = 0.39[0.17, 0.57]), and p-tau217 and p-tau181 were associated with both plaques (ρp-tau217 [95%CI] = 0.40[0.21, 0.56], ρp-tau181 [95%CI] = 0.36[0.15, 0.50]) and tangles (ρp-tau217 [95%CI] = 0.52[0.34, 0.66]; ρp-tau181 [95%CI] = 0.36[0.17, 0.52]). A model combining p-tau217 and the Aβ42/40 ratio showed the highest accuracy for predicting the presence of Alzheimer's disease neuropathological change (ADNC, AUC[95%CI] = 0.89[0.82, 0.96]) and plaque load (R2 = 0.55), while p-tau217 alone was optimal for predicting tangle load (R2 = 0.45). Our results suggest that high-performing assays of plasma p-tau217 and Aβ42/40 might be an optimal combination to assess Alzheimer's-related pathology in vivo.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley, NHS Foundation, London, UK
| | | | | | - Eric M Reiman
- Banner Alzheimer's Institute, Arizona State University and University of Arizona, Phoenix, AZ, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
15
|
Lue LF, Walker DG, Beh ST, Beach TG. Isolation of Human Microglia from Neuropathologically Diagnosed Cases in the Single-Cell Era. Methods Mol Biol 2023; 2561:43-62. [PMID: 36399264 DOI: 10.1007/978-1-0716-2655-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
This chapter describes the core procedures that we have developed over the last two decades to isolate routinely the microglia from postmortem human brains. The method is suitable for brain slices consisting of both gray and white matter.The ability to concomitantly isolate vascular cells with glial cells provides the opportunity to investigate multiple cell types originating from the same donor. This represents a novel approach for -omics research, with the potential for discovering the shared or distinct molecular features among the glia and vascular cells from the same individual.
Collapse
Affiliation(s)
- Lih-Fen Lue
- Human Cell Core for Translational Research, Banner Sun Health Research Institute, Sun City, AZ, USA.
| | - Douglas G Walker
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga, Otsu, Japan
| | - Suet Theng Beh
- Human Cell Core for Translational Research, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas G Beach
- Civin Neuropathology Laboratory, Banner Sun Health Research Institute, Sun City, AZ, USA
| |
Collapse
|
16
|
Krassner MM, Kauffman J, Sowa A, Cialowicz K, Walsh S, Farrell K, Crary JF, McKenzie AT. Postmortem changes in brain cell structure: a review. FREE NEUROPATHOLOGY 2023; 4:4-10. [PMID: 37384330 PMCID: PMC10294569 DOI: 10.17879/freeneuropathology-2023-4790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/15/2023] [Indexed: 06/30/2023]
Abstract
Brain cell structure is a key determinant of neural function that is frequently altered in neurobiological disorders. Following the global loss of blood flow to the brain that initiates the postmortem interval (PMI), cells rapidly become depleted of energy and begin to decompose. To ensure that our methods for studying the brain using autopsy tissue are robust and reproducible, there is a critical need to delineate the expected changes in brain cell morphometry during the PMI. We searched multiple databases to identify studies measuring the effects of PMI on the morphometry (i.e. external dimensions) of brain cells. We screened 2119 abstracts, 361 full texts, and included 172 studies. Mechanistically, fluid shifts causing cell volume alterations and vacuolization are an early event in the PMI, while the loss of the ability to visualize cell membranes altogether is a later event. Decomposition rates are highly heterogenous and depend on the methods for visualization, the structural feature of interest, and modifying variables such as the storage temperature or the species. Geometrically, deformations of cell membranes are common early events that initiate within minutes. On the other hand, topological relationships between cellular features appear to remain intact for more extended periods. Taken together, there is an uncertain period of time, usually ranging from several hours to several days, over which cell membrane structure is progressively lost. This review may be helpful for investigators studying human postmortem brain tissue, wherein the PMI is an unavoidable aspect of the research.
Collapse
Affiliation(s)
- Margaret M. Krassner
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Departments of Pathology, Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Justin Kauffman
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Departments of Pathology, Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Allison Sowa
- Microscopy and Advanced Bioimaging Core, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Katarzyna Cialowicz
- Microscopy and Advanced Bioimaging Core, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Samantha Walsh
- Hunter College Libraries, CUNY Hunter College, New York, NY
| | - Kurt Farrell
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Departments of Pathology, Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John F. Crary
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Departments of Pathology, Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew T. McKenzie
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Departments of Pathology, Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research Core and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
17
|
Targeted Methylation Profiling of Single Laser-Capture Microdissected Post-Mortem Brain Cells by Adapted Limiting Dilution Bisulfite Pyrosequencing (LDBSP). Int J Mol Sci 2022; 23:ijms232415571. [PMID: 36555213 PMCID: PMC9779089 DOI: 10.3390/ijms232415571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
A reoccurring issue in neuroepigenomic studies, especially in the context of neurodegenerative disease, is the use of (heterogeneous) bulk tissue, which generates noise during epigenetic profiling. A workable solution to this issue is to quantify epigenetic patterns in individually isolated neuronal cells using laser capture microdissection (LCM). For this purpose, we established a novel approach for targeted DNA methylation profiling of individual genes that relies on a combination of LCM and limiting dilution bisulfite pyrosequencing (LDBSP). Using this approach, we determined cytosine-phosphate-guanine (CpG) methylation rates of single alleles derived from 50 neurons that were isolated from unfixed post-mortem brain tissue. In the present manuscript, we describe the general workflow and, as a showcase, demonstrate how targeted methylation analysis of various genes, in this case, RHBDF2, OXT, TNXB, DNAJB13, PGLYRP1, C3, and LMX1B, can be performed simultaneously. By doing so, we describe an adapted data analysis pipeline for LDBSP, allowing one to include and correct CpG methylation rates derived from multi-allele reactions. In addition, we show that the efficiency of LDBSP on DNA derived from LCM neurons is similar to the efficiency obtained in previously published studies using this technique on other cell types. Overall, the method described here provides the user with a more accurate estimation of the DNA methylation status of each target gene in the analyzed cell pools, thereby adding further validity to this approach.
Collapse
|
18
|
Hasan S, Adler CH, Zhang N, Serrano GE, Sue LI, Shill HA, Mehta SH, Beach TG, Driver-Dunckley ED. Olfactory Dysfunction in Incidental Lewy Body Disease and Parkinson's Disease: An Update. INNOVATIONS IN CLINICAL NEUROSCIENCE 2022; 19:19-23. [PMID: 36591548 PMCID: PMC9776774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Objective Measuring olfactory dysfunction shows promise as one of a number of nonmotor biomarkers that can be used to detect clinically manifest and prodromal Parkinson's disease (PD) and dementia with Lewy bodies (DLB) and to differentiate these from nonsynucleinopathies. Using a larger sample size than in our previous study, we evaluated the relationship between olfactory dysfunction based on the University of Pennsylvania Smell Identification Test (UPSIT) to the clinicopathological findings in patients with PD (n=41), patients with incidental Lewy body disease (ILBD) (n=47), and controls with no neurodegenerative disease (n=137). Design This study was conducted through the Arizona Study of Aging and Neurodegenerative Disease (AZSAND). We selected individuals who had an UPSIT score completed antemortem and were clinicopathologically diagnosed with PD, ILBD, or control. Various measures included density of Lewy type synucleinopathy (aSyn) in the olfactory bulb and tract, as well as connected mesial temporal lobe structures. Cases and controls were analyzed using one-way analysis of variance (ANOVA) with pairwise contrasts. Results Compared to controls (mean: 27.8, standard deviation [SD]: 6.0), the mean UPSIT scores were lower for PD (15.8, SD: 6.0, p<0.001) and ILBD (24.1, SD: 8.6, p<0.001). The sensitivity for detecting ILBD from controls, based on a cutoff score of less than 23 (23/47), was 48.9 percent. The specificity for detecting a control was 79.6 percent with a cutoff greater than 23 (109/137). Conclusion These findings replicate, with a larger sample size, our previously published findings that individuals with autopsy-confirmed PD and ILBD have significantly lower UPSIT scores compared to controls. These data add to the growing body of evidence supporting early olfactory dysfunction as a prodromal biomarker for the risk of developing PD and ILBD as a prodromal Lewy body disorder.
Collapse
Affiliation(s)
- Shemonti Hasan
- Drs. Hasan, Adler, Mehta, and Driver-Dunckley are with the Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic in Scottsdale, Arizona
| | - Charles H Adler
- Drs. Hasan, Adler, Mehta, and Driver-Dunckley are with the Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic in Scottsdale, Arizona
| | - Nan Zhang
- Ms. Zhang is with the Department of Health Science Research, Section of Biostatistics, Mayo Clinic in Scottsdale, Arizona
| | - Geidy E Serrano
- Drs. Serrano and Beach and Ms. Sue are with Civin Laboratory for Neuropathology, Banner Sun Health Research Institute in Sun City, Arizona
| | - Lucia I Sue
- Drs. Serrano and Beach and Ms. Sue are with Civin Laboratory for Neuropathology, Banner Sun Health Research Institute in Sun City, Arizona
| | - Holly A Shill
- Dr. Shill is with Barrow Neurological Institute in Phoenix, Arizona
| | - Shyamal H Mehta
- Drs. Hasan, Adler, Mehta, and Driver-Dunckley are with the Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic in Scottsdale, Arizona
| | - Thomas G Beach
- Drs. Serrano and Beach and Ms. Sue are with Civin Laboratory for Neuropathology, Banner Sun Health Research Institute in Sun City, Arizona
| | - Erika D Driver-Dunckley
- Drs. Hasan, Adler, Mehta, and Driver-Dunckley are with the Parkinson's Disease and Movement Disorders Center, Department of Neurology, Mayo Clinic in Scottsdale, Arizona
| |
Collapse
|
19
|
Hall S, Orrù CD, Serrano GE, Galasko D, Hughson AG, Groveman BR, Adler CH, Beach TG, Caughey B, Hansson O. Performance of αSynuclein RT-QuIC in relation to neuropathological staging of Lewy body disease. Acta Neuropathol Commun 2022; 10:90. [PMID: 35733234 PMCID: PMC9219141 DOI: 10.1186/s40478-022-01388-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/21/2022] Open
Abstract
Currently, there is a need for diagnostic markers in Lewy body disorders (LBD). α-synuclein (αSyn) RT-QuIC has emerged as a promising assay to detect misfolded αSyn in clinically or neuropathologically established patients with various synucleinopathies. In this study, αSyn RT-QuIC was used to analyze lumbar CSF in a clinical cohort from the Swedish BioFINDER study and postmortem ventricular CSF in a neuropathological cohort from the Arizona Study of Aging and Neurodegenerative Disorders/Brain and Body Donation Program (AZSAND/BBDP). The BioFINDER cohort included 64 PD/PDD, 15 MSA, 15 PSP, 47 controls and two controls who later converted to PD/DLB. The neuropathological cohort included 101 cases with different brain disorders, including LBD and controls. In the BioFINDER cohort αSyn RT-QuIC identified LBD (i.e. PD, PDD and converters) vs. controls with a sensitivity of 95% and a specificity of 83%. The two controls that converted to LBD were αSyn RT-QuIC positive. Within the AZSAND/BBDP cohort, αSyn RT-QuIC identified neuropathologically verified "standard LBD" (i.e. PD, PD with AD and DLB; n = 25) vs. no LB pathology (n = 53) with high sensitivity (100%) and specificity (94%). Only 57% were αSyn RT-QuIC positive in the subgroup with "non-standard" LBD (i.e., AD with Lewy Bodies not meeting criteria for DLB or PD, and incidental LBD, n = 23). Furthermore, αSyn RT-QuIC reliably identified cases with LB pathology in the cortex (97% sensitivity) vs. cases with no LBs or LBs present only in the olfactory bulb (93% specificity). However, the sensitivity was low, only 50%, for cases with LB pathology restricted to the brainstem or amygdala, not affecting the allocortex or neocortex. In conclusion, αSyn RT-QuIC of CSF samples is highly sensitive and specific for identifying cases with clinicopathologically-defined Lewy body disorders and shows a lower sensitivity for non-standard LBD or asymptomatic LBD or in cases with modest LB pathology not affecting the cortex.
Collapse
Affiliation(s)
- Sara Hall
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, 20502, Malmö, Sweden.
| | - Christina D Orrù
- LPVD, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, USA
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Andrew G Hughson
- LPVD, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, USA
| | | | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Byron Caughey
- LPVD, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, 20502, Malmö, Sweden.
| |
Collapse
|
20
|
Brady CB, Robey I, Stein TD, Huber BR, Riley J, Abdul Rauf N, Spencer KR, Walt G, Adams L, Averill JG, Walker S, McKee AC, Thomson SP, Kowall NW. The Department of Veterans Affairs Gulf War Veterans' Illnesses Biorepository: Supporting Research on Gulf War Veterans' Illnesses. Brain Sci 2021; 11:1349. [PMID: 34679413 PMCID: PMC8533803 DOI: 10.3390/brainsci11101349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 12/27/2022] Open
Abstract
AIMS To introduce a resource supporting research on Gulf War illness (GWI) and related disorders, the Gulf War Veterans' Illnesses Biorepository (GWVIB). METHODS Gulf War era veterans (GWVs) are recruited nationally and enrolled via telephone and email/postal mail. Enrolled veterans receive annual telephone and mail follow-up to collect health data until their passing. A postmortem neuropathological examination is performed, and fixed and frozen brain and spinal cord samples are banked to support research. Investigators studying GWI and related disorders may request tissue and data from the GWVIB. RESULTS As of September 2021, 127 GWVs from 39 states were enrolled; 60 met the criteria for GWI, and 14 met the criteria for chronic multisymptom illness (CMI). Enrollees have been followed up to six years. Postmortem tissue recoveries were performed on 14 GWVs. The most commonly found neuropathologies included amyotrophic lateral sclerosis, chronic traumatic encephalopathy, and Lewy body disease. Tissue was of good quality with an average RNA integrity number of 5.8 (SD = 1.0) and ≥4.8 in all of the cases. DISCUSSION The availability of health data and high-quality CNS tissue from this well-characterized GWV cohort will support research on GWI and related disorders affecting GWVs. Enrollment is ongoing.
Collapse
Affiliation(s)
- Christopher B. Brady
- Research and Development Service, VA Boston Healthcare System, Boston, MA 02130, USA; (J.R.); (N.A.R.); (K.R.S.); (G.W.); (L.A.)
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA; (B.R.H.); (N.W.K.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Ian Robey
- Southern Arizona VA Healthcare System, Tucson, AZ 85723, USA; (I.R.); (J.G.A.); (S.W.); (S.P.T.)
- Department of Endocrinology, University of Arizona, Tucson, AZ 85724, USA
| | - Thor D. Stein
- Pathology Service, VA Boston Healthcare System, Boston, MA 02130, USA; (T.D.S.); (A.C.M.)
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Veterans Affairs Medical Center, Bedford, MA 01730, USA
| | - Bertrand R. Huber
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA; (B.R.H.); (N.W.K.)
- Pathology Service, VA Boston Healthcare System, Boston, MA 02130, USA; (T.D.S.); (A.C.M.)
- National Center for Posttraumatic Stress Disorder, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Jessica Riley
- Research and Development Service, VA Boston Healthcare System, Boston, MA 02130, USA; (J.R.); (N.A.R.); (K.R.S.); (G.W.); (L.A.)
| | - Nazifa Abdul Rauf
- Research and Development Service, VA Boston Healthcare System, Boston, MA 02130, USA; (J.R.); (N.A.R.); (K.R.S.); (G.W.); (L.A.)
| | - Keith R. Spencer
- Research and Development Service, VA Boston Healthcare System, Boston, MA 02130, USA; (J.R.); (N.A.R.); (K.R.S.); (G.W.); (L.A.)
| | - Gabriel Walt
- Research and Development Service, VA Boston Healthcare System, Boston, MA 02130, USA; (J.R.); (N.A.R.); (K.R.S.); (G.W.); (L.A.)
| | - Latease Adams
- Research and Development Service, VA Boston Healthcare System, Boston, MA 02130, USA; (J.R.); (N.A.R.); (K.R.S.); (G.W.); (L.A.)
| | - James G. Averill
- Southern Arizona VA Healthcare System, Tucson, AZ 85723, USA; (I.R.); (J.G.A.); (S.W.); (S.P.T.)
| | - Sean Walker
- Southern Arizona VA Healthcare System, Tucson, AZ 85723, USA; (I.R.); (J.G.A.); (S.W.); (S.P.T.)
| | - Ann C. McKee
- Pathology Service, VA Boston Healthcare System, Boston, MA 02130, USA; (T.D.S.); (A.C.M.)
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Stephen P. Thomson
- Southern Arizona VA Healthcare System, Tucson, AZ 85723, USA; (I.R.); (J.G.A.); (S.W.); (S.P.T.)
- Department of Endocrinology, University of Arizona, Tucson, AZ 85724, USA
| | - Neil W. Kowall
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA; (B.R.H.); (N.W.K.)
- Neurology Service, VA Boston Healthcare System, Boston, MA 02130, USA
| |
Collapse
|
21
|
Serrano GE, Walker JE, Intorcia AJ, Glass MJ, Arce RA, Piras IS, Talboom JS, Nelson CM, Cutler BD, Sue LI, Lue LF, Huentelman M, Beach TG. Whole-Cell Dissociated Suspension Analysis in Human Brain Neurodegenerative Disease: A Pilot Study. JOURNAL OF TISSUE SCIENCE & ENGINEERING 2021; 12:242.. [PMID: 39055131 PMCID: PMC11271822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Biochemical analysis of human brain tissue is typically done by homogenizing whole pieces of brain and separately characterizing the proteins, RNA, DNA, and other macromolecules within. While this has been sufficient to identify substantial changes, there is little ability to identify small changes or alterations that may occur in subsets of cells. To effectively investigate the biochemistry of disease in the brain, with its different cell types, we must first separate the cells and study them as phenotypically defined populations or even as individuals. In this project, we developed a new method for the generation of Whole Cell Dissociated Suspensions (WCDS) in fresh human brain tissue that could be shared as a resource with scientists to study single human cells or populations. Characterization of WCDS was done in paraffin-embedded sections stained with H&E, and by phenotyping with antibodies using immunohistochemistry and Fluorescence Activated Cell Sorting (FACS). Additionally, we compared extracted RNA from WCDS with RNA from adjacent intact cortical tissue, using RT-qPCR for cell-type-specific RNA for the same markers as well as whole transcriptome sequencing. More than 11,626 gene transcripts were successfully sequenced and classified using an external database either as being mainly expressed in neurons, astrocytes, microglia, oligodendrocytes, endothelial cells, or mixed (in two or more cell types). This demonstrates that we are currently capable of producing WCDS with a full representation of different brain cell types combined with RNA quality suitable for use in biochemical analysis.
Collapse
Affiliation(s)
- Geidy E Serrano
- Banner Sun Health Research Institute, Arizona, United States
| | | | | | - Michael J Glass
- Banner Sun Health Research Institute, Arizona, United States
| | - Richard A Arce
- Banner Sun Health Research Institute, Arizona, United States
| | - Ignazio S. Piras
- Translational Genomics Research Institute, Phoenix, USA
- Institute of Arizona Alzheimer’s Consortium, Phoenix, USA
| | - Joshua S Talboom
- Translational Genomics Research Institute, Phoenix, USA
- Institute of Arizona Alzheimer’s Consortium, Phoenix, USA
| | | | - Brett D Cutler
- Banner Sun Health Research Institute, Arizona, United States
| | - Lucia I Sue
- Banner Sun Health Research Institute, Arizona, United States
| | - Lih-Fen Lue
- Banner Sun Health Research Institute, Arizona, United States
| | - Matthew Huentelman
- Translational Genomics Research Institute, Phoenix, USA
- Institute of Arizona Alzheimer’s Consortium, Phoenix, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, Arizona, United States
| |
Collapse
|
22
|
Adler CH, Beach TG, Zhang N, Shill HA, Driver-Dunckley E, Mehta SH, Atri A, Caviness JN, Serrano G, Shprecher DR, Sue LI, Belden CM. Clinical Diagnostic Accuracy of Early/Advanced Parkinson Disease: An Updated Clinicopathologic Study. Neurol Clin Pract 2021; 11:e414-e421. [PMID: 34484939 DOI: 10.1212/cpj.0000000000001016] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/01/2020] [Indexed: 11/15/2022]
Abstract
Objective To update data for diagnostic accuracy of a clinical diagnosis of Parkinson disease (PD) using neuropathologic diagnosis as the gold standard. Methods Data from the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND) were used to determine the predictive value of a clinical PD diagnosis. Two clinical diagnostic confidence levels were used, possible PD (PossPD, never treated or not responsive) and probable PD (ProbPD, 2/3 cardinal clinical signs and responsive to dopaminergic medications). Neuropathologic diagnosis was the gold standard. Results Based on the first visit to AZSAND, 15/54 (27.8%) PossPD participants and 138/163 (84.7%) ProbPD participants had confirmed PD. PD was confirmed in 24/34 (70.6%) ProbPD with <5 years and 114/128 (89.1%) with ≥5 years disease duration. Using the consensus final clinical diagnosis following death, 161/187 (86.1%) ProbPD had neuropathologically confirmed PD. Diagnostic accuracy for ProbPD improved if included motor fluctuations, dyskinesias, and hyposmia, and hyposmia for PossPD. Conclusions This updated study confirmed lower clinical diagnostic accuracy for elderly, untreated or poorly responsive PossPD participants and for ProbPD with <5 years of disease duration, even when medication responsive. Caution continues to be needed when interpreting clinical studies of PD, especially studies of early disease, that do not have autopsy confirmation. Classification of Evidence This study provides Class II evidence that a clinical diagnosis of ProbPD at the first visit identifies participants who will have pathologically confirmed PD with a sensitivity of 82.6% and a specificity of 86.0%.
Collapse
Affiliation(s)
- Charles H Adler
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Thomas G Beach
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Nan Zhang
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Holly A Shill
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Erika Driver-Dunckley
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Shyamal H Mehta
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Alireza Atri
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - John N Caviness
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Geidy Serrano
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - David R Shprecher
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lucia I Sue
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Christine M Belden
- Parkinson's Disease and Movement Disorders Center (CHA, EDD, SHM), Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Civin Laboratory for Neuropathology (TGB, GS, LIS), Banner Sun Health Research Institute, Sun City, AZ; Department of Biostatistics (NZ), Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale; Barrow Neurologic Institute (HAS), Phoenix, AZ; Cleo Roberts Center (AA, DRS, CMB), Banner Sun Health Research Institute, Sun City, AZ; and Center for Brain/Mind Medicine (AA), Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
23
|
Lyu Y, Zauhar R, Dana N, Strang CE, Hu J, Wang K, Liu S, Pan N, Gamlin P, Kimble JA, Messinger JD, Curcio CA, Stambolian D, Li M. Implication of specific retinal cell-type involvement and gene expression changes in AMD progression using integrative analysis of single-cell and bulk RNA-seq profiling. Sci Rep 2021; 11:15612. [PMID: 34341398 PMCID: PMC8329233 DOI: 10.1038/s41598-021-95122-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
Age-related macular degeneration (AMD) is a blinding eye disease with no unifying theme for its etiology. We used single-cell RNA sequencing to analyze the transcriptomes of ~ 93,000 cells from the macula and peripheral retina from two adult human donors and bulk RNA sequencing from fifteen adult human donors with and without AMD. Analysis of our single-cell data identified 267 cell-type-specific genes. Comparison of macula and peripheral retinal regions found no cell-type differences but did identify 50 differentially expressed genes (DEGs) with about 1/3 expressed in cones. Integration of our single-cell data with bulk RNA sequencing data from normal and AMD donors showed compositional changes more pronounced in macula in rods, microglia, endothelium, Müller glia, and astrocytes in the transition from normal to advanced AMD. KEGG pathway analysis of our normal vs. advanced AMD eyes identified enrichment in complement and coagulation pathways, antigen presentation, tissue remodeling, and signaling pathways including PI3K-Akt, NOD-like, Toll-like, and Rap1. These results showcase the use of single-cell RNA sequencing to infer cell-type compositional and cell-type-specific gene expression changes in intact bulk tissue and provide a foundation for investigating molecular mechanisms of retinal disease that lead to new therapeutic targets.
Collapse
Affiliation(s)
- Yafei Lyu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Randy Zauhar
- Department of Chemistry and Biochemistry, The University of the Sciences in Philadelphia, Philadelphia, PA, 19104, USA
| | - Nicholas Dana
- Departments of Ophthalmology and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Christianne E Strang
- Department of Psychology, University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - Jian Hu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Kui Wang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Information Theory and Data Science, School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 30071, China
| | - Shanrun Liu
- Department of Biochemistry and Molecular Genetics, University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - Naifei Pan
- Department of Computer and Information Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Paul Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - James A Kimble
- Department of Ophthalmology and Visual Sciences, University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - Jeffrey D Messinger
- Department of Ophthalmology and Visual Sciences, University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - Dwight Stambolian
- Departments of Ophthalmology and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
24
|
Dave N, Vural AS, Piras IS, Winslow W, Surendra L, Winstone JK, Beach TG, Huentelman MJ, Velazquez R. Identification of retinoblastoma binding protein 7 (Rbbp7) as a mediator against tau acetylation and subsequent neuronal loss in Alzheimer's disease and related tauopathies. Acta Neuropathol 2021; 142:279-294. [PMID: 33978814 PMCID: PMC8270842 DOI: 10.1007/s00401-021-02323-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022]
Abstract
Evidence indicates that tau hyper-phosphorylation and subsequent neurofibrillary tangle formation contribute to the extensive neuronal death in Alzheimer's disease (AD) and related tauopathies. Recent work has identified that increased tau acetylation can promote tau phosphorylation. Tau acetylation occurs at lysine 280 resulting from increased expression of the lysine acetyltransferase p300. The exact upstream mechanisms mediating p300 expression remain elusive. Additional work highlights the role of the epigenome in tau pathogenesis, suggesting that dysregulation of epigenetic proteins may contribute to acetylation and hyper-phosphorylation of tau. Here, we identify and focus on the histone-binding subunit of the Nucleosome Remodeling and Deacetylase (NuRD) complex: Retinoblastoma-Binding Protein 7 (Rbbp7). Rbbp7 chaperones chromatin remodeling proteins to their nuclear histone substrates, including histone acetylases and deacetylases. Notably, Rbbp7 binds to p300, suggesting that it may play a role in modulating tau acetylation. We interrogated Rbbp7 in post-mortem brain tissue, cell lines and mouse models of AD. We found reduced Rbbp7 mRNA expression in AD cases, a significant negative correlation with CERAD (neuritic plaque density) and Braak Staging (pathogenic tau inclusions) and a significant positive correlation with post-mortem brain weight. We also found a neuron-specific downregulation of Rbbp7 mRNA in AD patients. Rbbp7 protein levels were significantly decreased in 3xTg-AD and PS19 mice compared to NonTg, but no decreases were found in APP/PS1 mice that lack tau pathology. In vitro, Rbbp7 overexpression rescued TauP301L-induced cytotoxicity in immortalized hippocampal cells and primary cortical neurons. In vivo, hippocampal Rbbp7 overexpression rescued neuronal death in the CA1 of PS19 mice. Mechanistically, we found that increased Rbbp7 reduced p300 levels, tau acetylation at lysine 280 and tau phosphorylation at AT8 and AT100 sites. Collectively, these data identify a novel role of Rbbp7, protecting against tau-related pathologies, and highlight its potential as a therapeutic target in AD and related tauopathies.
Collapse
Affiliation(s)
- Nikhil Dave
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Austin S Vural
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Ignazio S Piras
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Wendy Winslow
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Likith Surendra
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Joanna K Winstone
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Thomas G Beach
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Ramon Velazquez
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA.
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
25
|
Shill HA, Zhang N, Driver-Dunckley E, Mehta S, Adler CH, Beach TG. Olfaction in Neuropathologically Defined Progressive Supranuclear Palsy. Mov Disord 2021; 36:1700-1704. [PMID: 33755262 PMCID: PMC9972484 DOI: 10.1002/mds.28568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Hyposmia is characteristic of idiopathic Parkinson's disease (PD) and dementia with Lewy bodies (DLBs), whereas progressive supranuclear palsy (PSP) typically has normal sense of smell. However, there is a lack of pathologically confirmed data. OBJECTIVE The objective is to study hyposmia in pathologically confirmed PSP patients and compare to PD patients and nondegenerative controls. METHODS We studied autopsied subjects in the Arizona Study of Aging and Neurodegenerative Disorders who had antemortem olfactory testing and a neuropathological diagnosis of either PD, PSP, or control. RESULTS This study included 281 cases. Those with neuropathologically confirmed PSP (N = 24) and controls (N = 174) had significantly better sense of smell than those with PD (N = 76). Although most PSP patients had normal olfaction, there were some with hyposmia, resulting in an overall reduced sense of smell in PSP compared to controls. The sensitivity of having PSP pathologically in those presenting with parkinsonism and normosmia was 93.4% with a specificity of 64.7%. Cases with both PSP and PD pathologically had reduced sense of smell similar to PD alone (N = 7). Hyposmic PSP patients had significantly higher Lewy body burden not meeting criteria for additional PD/DLB diagnosis. CONCLUSIONS Pathologically confirmed PD had reduced olfaction compared with PSP or controls. In the setting of parkinsonism in this sample, the presence of normosmia had high sensitivity for PSP. Hyposmia in PSP suggests the presence of additional Lewy body pathology. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Holly A. Shill
- Barrow Neurological Institute, Phoenix, AZ, USA,University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Nan Zhang
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | | | - Shyamal Mehta
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Charles H. Adler
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | | |
Collapse
|
26
|
Li QS, De Muynck L. Differentially expressed genes in Alzheimer's disease highlighting the roles of microglia genes including OLR1 and astrocyte gene CDK2AP1. Brain Behav Immun Health 2021; 13:100227. [PMID: 34589742 PMCID: PMC8474442 DOI: 10.1016/j.bbih.2021.100227] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with abnormal tau and amyloid-β accumulation in the brain, leading to neurofibrillary tangles, neuropil threads and extracellular amyloid-β plaques. Treatment is limited to symptom management, a disease-modifying therapy is not available. To advance search of therapy approaches, there is a continued need to identify targets for disease intervention both by confirming existing hypotheses and generating new hypotheses. METHODS We conducted a mRNA-seq study to identify genes associated with AD in post-mortem brain samples from the superior temporal gyrus (STG, n = 76), and inferior frontal gyrus (IFG, n = 65) brain regions. Differentially expressed genes (DEGs) were identified correcting for gender and surrogate variables to capture hidden variation not accounted for by pre-planned covariates. The results from this study were compared with the transcriptome studies from the Accelerated Medicine Partnership - Alzheimer's Disease (AMP-AD) initiative. Over-representation and gene set enrichment analysis (GSEA) was used to identify disease-associated pathways. Protein-protein interaction (PPI) and weighted gene co-expression network analysis (WGCNA) analyses were carried out and co-expressed gene modules and their hub genes were identified and associated with additional phenotypic traits of interest. RESULTS Several hundred mRNAs were differentially expressed between AD cases and cognitively normal controls in the STG, while no and few transcripts met the same criteria (adjusted p less than 0.05 and fold change greater than 1.2) in the IFG. The findings were consistent at the gene set level with two out of three cohorts from AMP-AD. PPI analysis suggested that the DEGs were enriched in protein-protein interactions than expected by random chance. Over-representation and GSEA analysis suggested genes playing roles in neuroinflammation, amyloid-β, autophagy and trafficking being important for the AD disease process. At the gene level, 10 genes from the STG that were consistently differentially expressed in this study and in the MSBB study (one of the three cohorts within the AMP-AD initiative) were enriched in microglial genes (TREM2, C3AR1, ITGAX, OLR1, CD74, and HLA-DRA), but also included genes with a broader cell type expression pattern such as CDK2AP1. Among the DEGs with supporting evidence from an independent study, CDK2AP1 (most abundantly expressed in astrocyte) was the transcript with strongest association with antemortem cognitive measure (last Mini-Mental State Examination score) and neurofibril tangle burden but also associated with amyloid plaque burden, while OLR1 was the transcript with strongest association with amyloid plaque burden. GSEA and over-representation analyses revealed gene sets related to immune processes including neutrophil degranulation, interleukin 10 signaling, and interferon gamma signaling, complement and coagulation cascades, phosphatidylinositol signaling system, phagosome and neurotransmitter receptors and postsynaptic signal transmission were enriched from this study and replicated in an independent study. CONCLUSION This study identified differential gene sets, common with two out of three AMP-AD cohorts (ROSMAP and MSBB) and highlights microglia and astrocyte as the key cell-types with DGEs associated with AD clinical diagnosis, and/or antemortem cognitive measure as well as neuropathological indices. Future meta-analysis and causal inferential analysis will be helpful in pinpointing the most relevant pathways and genes to intervene.
Collapse
Affiliation(s)
- Qingqin S. Li
- Neuroscience Department, Janssen Research & Development, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ, 08560, USA
| | - Louis De Muynck
- Neuroscience Department, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, 2340, Beerse, Belgium
| |
Collapse
|
27
|
Li QS, Cai D. Integrated miRNA-Seq and mRNA-Seq Study to Identify miRNAs Associated With Alzheimer's Disease Using Post-mortem Brain Tissue Samples. Front Neurosci 2021; 15:620899. [PMID: 33833661 PMCID: PMC8021900 DOI: 10.3389/fnins.2021.620899] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/23/2021] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD), the leading form of dementia, is associated with abnormal tau and β-amyloid accumulation in the brain. We conducted a miRNA-seq study to identify miRNAs associated with AD in the post-mortem brain from the inferior frontal gyrus (IFG, n = 69) and superior temporal gyrus (STG, n = 81). Four and 64 miRNAs were differentially expressed (adjusted p-value < 0.05) in AD compared to cognitively normal controls in the IFG and STG, respectively. We observed down-regulation of several miRNAs that have previously been implicated in AD, including hsa-miR-212-5p and hsa-miR-132-5p, in AD samples across both brain regions, and up-regulation of hsa-miR-146a-5p, hsa-miR-501-3p, hsa-miR-34a-5p, and hsa-miR-454-3p in the STG. The differentially expressed miRNAs were previously implicated in the formation of amyloid-β plaques, the dysregulation of tau, and inflammation. We have also observed differential expressions for dozens of other miRNAs in the STG, including hsa-miR-4446-3p, that have not been described previously. Putative targets of these miRNAs (adjusted p-value < 0.1) were found to be involved in Wnt signaling pathway, MAPK family signaling cascades, sphingosine 1-phosphate (S1P) pathway, adaptive immune system, innate immune system, and neurogenesis. Our results support the finding of dysregulated miRNAs previously implicated in AD and propose additional miRNAs that appear to be dysregulated in AD for experimental follow-up.
Collapse
Affiliation(s)
- Qingqin S. Li
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, United States
| | | |
Collapse
|
28
|
Giovagnoni C, Ali M, Eijssen LMT, Maes R, Choe K, Mulder M, Kleinjans J, Del Sol A, Glaab E, Mastroeni D, Delvaux E, Coleman P, Losen M, Pishva E, Martinez-Martinez P, van den Hove DLA. Altered sphingolipid function in Alzheimer's disease; a gene regulatory network approach. Neurobiol Aging 2021; 102:178-187. [PMID: 33773368 DOI: 10.1016/j.neurobiolaging.2021.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/04/2020] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
Sphingolipids (SLs) are bioactive lipids involved in various important physiological functions. The SL pathway has been shown to be affected in several brain-related disorders, including Alzheimer's disease (AD). Recent evidence suggests that epigenetic dysregulation plays an important role in the pathogenesis of AD as well. Here, we use an integrative approach to better understand the relationship between epigenetic and transcriptomic processes in regulating SL function in the middle temporal gyrus of AD patients. Transcriptomic analysis of 252 SL-related genes, selected based on GO term annotations, from 46 AD patients and 32 healthy age-matched controls, revealed 103 differentially expressed SL-related genes in AD patients. Additionally, methylomic analysis of the same subjects revealed parallel hydroxymethylation changes in PTGIS, GBA, and ITGB2 in AD. Subsequent gene regulatory network-based analysis identified 3 candidate genes, that is, SELPLG, SPHK1 and CAV1 whose alteration holds the potential to revert the gene expression program from a diseased towards a healthy state. Together, this epigenomic and transcriptomic approach highlights the importance of SL-related genes in AD, and may provide novel biomarkers and therapeutic alternatives to traditionally investigated biological pathways in AD.
Collapse
Affiliation(s)
- Caterina Giovagnoni
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands.
| | - Muhammad Ali
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; Computational Biology Group, Luxembourg Centre for System Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg; Biomedical Data Science Group, Luxembourg Centre for System Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Lars M T Eijssen
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, the Netherlands
| | - Richard Maes
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, the Netherlands
| | - Kyonghwan Choe
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Monique Mulder
- Department of Internal Medicine, Division of Pharmacology, Vascular and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jos Kleinjans
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for System Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
| | - Enrico Glaab
- Biomedical Data Science Group, Luxembourg Centre for System Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Diego Mastroeni
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Elaine Delvaux
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Paul Coleman
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Mario Losen
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Ehsan Pishva
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Pilar Martinez-Martinez
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Daniel L A van den Hove
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands; Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
29
|
Age-Related Macular Degeneration: From Epigenetics to Therapeutic Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:221-235. [PMID: 33848004 DOI: 10.1007/978-3-030-66014-7_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Aberrant regulation of epigenetic mechanisms, including the two most common types; DNA methylation and histone modification have been implicated in common chronic progressive conditions, including Alzheimer disease, cardiovascular disease, and age-related macular degeneration (AMD). All these conditions are complex, meaning that environmental factors, genetic factors, and their interactions play a role in disease pathophysiology. Although genome wide association studies (GWAS), and studies on twins demonstrate the genetic/hereditary component to these complex diseases, including AMD, this contribution is much less than 100%. Moreover, the contribution of the hereditary component decreases in the advanced, later onset forms of these chronic diseases including AMD. This underscores the need to elucidate how the genetic and environmental factors function to exert their influence on disease pathophysiology. By teasing out epigenetic mechanisms and how they exert their influence on AMD, therapeutic targets can be tailored to prevent and/or slow down disease progression. Epigenetic studies that incorporate well-characterized patient tissue samples (including affected tissues and peripheral blood), similar to those relevant to gene expression studies, along with genetic and epidemiological information, can be the first step in developing appropriate functional assays to validate findings and identify potential therapies.
Collapse
|
30
|
He P, Schulz P, Sierks MR. A conformation-specific antibody against oligomeric β-amyloid restores neuronal integrity in a mouse model of Alzheimer's disease. J Biol Chem 2021; 296:100241. [PMID: 33376140 PMCID: PMC7948963 DOI: 10.1074/jbc.ra120.015327] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/03/2020] [Accepted: 12/29/2020] [Indexed: 11/06/2022] Open
Abstract
Conformationally distinct aggregates of the amyloid β (Aβ) peptide accumulate in brains of patients with Alzheimer's disease (AD), but the roles of the different aggregates in disease progression are not clear. We previously isolated two single-chain variable domain antibody fragments (scFvs), C6T and A4, that selectively bind different toxic conformational variants of oligomeric Aβ. Here, we utilize these scFvs to localize the presence of these Aβ variants in human AD brain and to demonstrate their potential as therapeutic agents for treating AD. Both A4 and C6T label oligomeric Aβ in extracellular amyloid plaques, whereas C6T also labels intracellular oligomeric Aβ in human AD brain tissue and in an AD mouse model. For therapeutic studies, the A4 and C6T scFvs were expressed in the AD mice by viral infection of liver cells. The scFvs were administered at 2 months of age, and mice sacrificed at 9 months. The scFvs contained a peptide tag to facilitate transport across the blood brain barrier. While treatment with C6T only slightly decreased Aβ deposits and plaque-associated inflammation, it restored neuronal integrity to WT levels, significantly promoted growth of new neurons, and impressively rescued survival rates to WT levels. Treatment with A4 on the other hand significantly decreased Aβ deposits but did not significantly decrease neuroinflammation or promote neuronal integrity, neurogenesis, or survival rate. These results suggest that the specific Aβ conformation targeted in therapeutic applications greatly affects the outcome, and the location of the targeted Aβ variants may also play a critical factor.
Collapse
Affiliation(s)
- Ping He
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA
| | - Philip Schulz
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA
| | - Michael R Sierks
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA.
| |
Collapse
|
31
|
Ortuño-Lizarán I, Sánchez-Sáez X, Lax P, Serrano GE, Beach TG, Adler CH, Cuenca N. Dopaminergic Retinal Cell Loss and Visual Dysfunction in Parkinson Disease. Ann Neurol 2020; 88:893-906. [PMID: 32881029 PMCID: PMC10005860 DOI: 10.1002/ana.25897] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Considering the demonstrated implication of the retina in Parkinson disease (PD) pathology and the importance of dopaminergic cells in this tissue, we aimed to analyze the state of the dopaminergic amacrine cells and some of their main postsynaptic neurons in the retina of PD. METHODS Using immunohistochemistry and confocal microscopy, we evaluated morphology, number, and synaptic connections of dopaminergic cells and their postsynaptic cells, AII amacrine and melanopsin-containing retinal ganglion cells, in control and PD eyes from human donors. RESULTS In PD, dopaminergic amacrine cell number was reduced between 58% and 26% in different retinal regions, involving a decline in the number of synaptic contacts with AII amacrine cells (by 60%) and melanopsin cells (by 35%). Despite losing their main synaptic input, AII cells were not reduced in number, but they showed cellular alterations compromising their adequate function: (1) a loss of mitochondria inside their lobular appendages, which may indicate an energetic failure; and (2) a loss of connexin 36, suggesting alterations in the AII coupling and in visual signal transmission from the rod pathway. INTERPRETATION The dopaminergic system impairment and the affection of the rod pathway through the AII cells may explain and be partially responsible for the reduced contrast sensitivity or electroretinographic response described in PD. Also, dopamine reduction and the loss of synaptic contacts with melanopsin cells may contribute to the melanopsin retinal ganglion cell loss previously described and to the disturbances in circadian rhythm and sleep reported in PD patients. These data support the idea that the retina reproduces brain neurodegeneration and is highly involved in PD pathology. ANN NEUROL 2020;88:893-906.
Collapse
Affiliation(s)
- Isabel Ortuño-Lizarán
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | | | | | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
| |
Collapse
|
32
|
Human Autopsy-Derived Scalp Fibroblast Biobanking for Age-Related Neurodegenerative Disease Research. Cells 2020; 9:cells9112383. [PMID: 33143239 PMCID: PMC7692621 DOI: 10.3390/cells9112383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/28/2020] [Indexed: 11/23/2022] Open
Abstract
The Arizona Study of Aging and Neurodegenerative Disorders/Brain and Body Donation Program at Banner Sun Health Research Institute (BSHRI) is a longitudinal clinicopathological study with a current enrollment of more than 900 living subjects for aging and neurodegenerative disease research. Annual clinical assessments are done by cognitive and movement neurologists and neuropsychologists. Brain and body tissues are collected at a median postmortem interval of 3.0 h for neuropathological diagnosis and banking. Since 2018, the program has undertaken banking of scalp fibroblasts derived from neuropathologically characterized donors with Alzheimer’s disease, Parkinson’s disease, and other neurodegenerative diseases. Here, we describe the procedure development and cell characteristics from 14 male and 15 female donors (mean ± SD of age: 83.6 ± 12.2). Fibroblasts from explant cultures were banked at passage 3. The results of mRNA analysis showed positive expression of fibroblast activation protein, vimentin, fibronectin, and THY1 cell surface antigen. We also demonstrated that the banked fibroblasts from a postmortem elderly donor were successfully reprogramed to human-induced pluripotent stem cells (hiPSCs). Taken together, we have demonstrated the successful establishment of a human autopsy-derived fibroblast banking program. The cryogenically preserved cells are available for request at the program website of the BSHRI.
Collapse
|
33
|
Li QS, Sun Y, Wang T. Epigenome-wide association study of Alzheimer's disease replicates 22 differentially methylated positions and 30 differentially methylated regions. Clin Epigenetics 2020; 12:149. [PMID: 33069246 PMCID: PMC7568396 DOI: 10.1186/s13148-020-00944-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Growing evidence shows that epigenetic modifications play a role in Alzheimer's disease (AD). We performed an epigenome-wide association study (EWAS) to evaluate the DNA methylation differences using postmortem superior temporal gyrus (STG) and inferior frontal gyrus (IFG) samples. RESULTS Samples from 72 AD patients and 62 age-matched cognitively normal controls were assayed using Illumina© Infinium MethylationEPIC BeadChip. Five and 14 differentially methylated positions (DMPs) associated with pathology (i.e., Braak stage) with p value less than Bonferroni correction threshold of 6.79 × 10-8 in the STG and IFG were identified, respectively. These cytosine-phosphate-guanine (CpG) sites included promoter associated cg26263477 annotated to ABCA7 in the STG (p = 1.21 × 10-11), and cg14058329 annotated to the HOXA5/HOXA3/HOXA-AS3 gene cluster (p = 1.62 × 10-9) and cg09448088 (p = 3.95 × 10-9) annotated to MCF2L in the IFG. These genes were previously reported to harbor DMPs and/or differentially methylated regions (DMRs). Previously reported DMPs annotated to RMGA, GNG7, HOXA3, GPR56, SPG7, PCNT, RP11-961A15.1, MCF2L, RHBDF2, ANK1, PCNT, TPRG1, and RASGEF1C were replicated (p < 0.0001). One hundred twenty-one and 173 DMRs associated with pathology in the STG and IFG, respectively, were additionally identified. Of these, DMRs annotated to 30 unique genes were also identified as significant DMRs in the same brain region in a recent meta-analysis, while additional DMRs annotated to 12 genes were reported as DMRs in a different brain region or in a cross-cortex meta-analysis. The significant DMRs were enriched in promoters, CpG islands, and exons in the genome. Gene set enrichment analysis of DMPs and DMRs showed that gene sets involved in neuroinflammation (e.g., microglia differentiation), neurogenesis, and cognition were enriched (false discovery rate (FDR) < 0.05). CONCLUSIONS Twenty-two DMPs and 30 DMRs associated with pathology were replicated, and novel DMPs and DMRs were discovered.
Collapse
Affiliation(s)
- Qingqin S Li
- Neuroscience, Janssen Research & Development, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ, 08560, USA.
| | - Yu Sun
- Neuroscience, Janssen Research & Development, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ, 08560, USA
- Discovery Science, Janssen Research & Development, LLC, Spring House, PA, USA
| | - Tania Wang
- AccuraScience, LLC, Johnston, IA, USA
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410083, China
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
34
|
Venkataraman L, He P, Khan G, Harris BT, Sierks MR. Isolation and characterization of antibody fragments selective for human FTD brain derived TDP-43 variants. BMC Neurosci 2020; 21:36. [PMID: 32887544 PMCID: PMC7472585 DOI: 10.1186/s12868-020-00586-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Frontotemporal dementia (FTD) is the second leading cause of early onset dementia following Alzheimer's disease. It involves atrophy of the frontal and temporal regions of the brain affecting language, memory, and behavior. Transactive response DNA-binding protein 43 (TDP-43) pathology is found in most FTD and ALS cases. It plays a role in transcription, translation and serves as a shuttle between the nucleus and cytoplasm. Prior to its aggregation, TDP-43 exists as polyubiquitinated, hyperphosphorylated C-terminal fragments that correlate well with FTD disease progression. Because of the importance of TDP-43 in these diseases, reagents that can selectively recognize specific toxic TDP variants associated with onset and progression of FTD can be effective diagnostic and therapeutic tools. RESULTS We utilized a novel atomic force microscopy (AFM) based biopanning protocol to isolate single chain variable fragments (scFvs) from a phage display library that selectively bind TDP variants present in human FTD but not cognitively normal age matched brain tissue. We then used the scFvs (FTD-TDP1 through 5) to probe post-mortem brain tissue and sera samples for the presence of FTD related TDP variants. The scFvs readily selected the FTD tissue and sera samples over age matched controls. The scFvs were used in immunohistochemical analysis of FTD and control brain slices where the reagents showed strong staining with TDP in FTD brain tissue slice. FTD-TDP1, FTD-TDP2, FTD-TDP4 and FTD-TDP5 all protected neuronal cells against FTD TDP induced toxicity suggesting potential therapeutic value. CONCLUSIONS These results show existence of different disease specific TDP variants in FTD individuals. We have identified a panel of scFvs capable of recognizing these disease specific TDP variants in postmortem FTD tissue and sera samples over age matched controls and can thus serve as a biomarker tool.
Collapse
Affiliation(s)
| | - Ping He
- Chemical Engineering, School for Engineering, Matter, Transport and Energy, Arizona State University, ECG301-501 Tyler Mall, Tempe, AZ, 85281-6106, USA
| | - Galam Khan
- Departments of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Brent T Harris
- Departments of Neurology, Georgetown University Medical Center, Washington, DC, USA.,Departments of Pathology, Georgetown University Medical Center, Washington, DC, USA
| | - Michael R Sierks
- Chemical Engineering, School for Engineering, Matter, Transport and Energy, Arizona State University, ECG301-501 Tyler Mall, Tempe, AZ, 85281-6106, USA.
| |
Collapse
|
35
|
Tang K, Wan M, Zhang H, Zhang Q, Yang Q, Chen K, Wang N, Zhang D, Qiu W, Ma C. The top 100 most-cited articles citing human brain banking from 1970 to 2020: a bibliometric analysis. Cell Tissue Bank 2020; 21:685-697. [PMID: 32761559 DOI: 10.1007/s10561-020-09849-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/09/2020] [Indexed: 02/08/2023]
Abstract
Many articles involving human brain banks have been published. Bibliometric analysis can determine the history of the development of research and future research trends in a specific field. Three independent researchers retrieved and reviewed articles from the Web of Science database using the following strategy: "TS = (((brain OR cerebral) AND (bank* OR biobank*)) OR brainbank*)." The top 100 most-cited articles were identified and listed in descending order by total citations. Web of Science was used to identify ten recent articles describing bank construction. GeenMedical ( https://www.geenmedical.com/ ) was used to identify ten recent articles from journals with an impact factor (IF) > 20. The top 100 most-cited articles citing human brain banks were published between 1991 and 2017. Fifty-two percent of the articles focused on a specific type of neurodegenerative disease, and 16% discussed the construction and development of human brain banks. Articles using brain tissue had more total and annual citations than those on bank construction. Ten articles with high IFs were published from 2017 to 2019, and they were primarily studies using novel research techniques such RNA sequencing and genome-wide association studies. Most studies were published in journals specializing in neurology or neuroscience such as Movement Disorders (10%), and had been conducted in the United States (52%) by neurologists (62%). The top 100 most-cited articles and recent publications citing human brain banks and their bibliometric characteristics were identified and analyzed, which may serve as a useful reference and pave the way for further research.
Collapse
Affiliation(s)
- Keyun Tang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Mengyao Wan
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Hanlin Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Qing Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qian Yang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Kang Chen
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Eight-Year MD Program, Peking Union Medical College, Beijing, China
| | - Naili Wang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Experimental Demonstration Center of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Di Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,National Experimental Demonstration Center of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China. .,Joint Laboratory of Anesthesia and Pain, Peking Union Medical College, Beijing, China.
| |
Collapse
|
36
|
Raghunathan R, Hogan JD, Labadorf A, Myers RH, Zaia J. A glycomics and proteomics study of aging and Parkinson's disease in human brain. Sci Rep 2020; 10:12804. [PMID: 32733076 PMCID: PMC7393382 DOI: 10.1038/s41598-020-69480-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/04/2020] [Indexed: 01/08/2023] Open
Abstract
Previous studies on Parkinson’s disease mechanisms have shown dysregulated extracellular transport of α-synuclein and growth factors in the extracellular space. In the human brain these consist of perineuronal nets, interstitial matrices, and basement membranes, each composed of a set of collagens, non-collagenous glycoproteins, proteoglycans, and hyaluronan. The manner by which amyloidogenic proteins spread extracellularly, become seeded, oligomerize, and are taken up by cells, depends on intricate interactions with extracellular matrix molecules. We sought to assess the alterations to structure of glycosaminoglycans and proteins that occur in PD brain relative to controls of similar age. We found that PD differs markedly from normal brain in upregulation of extracellular matrix structural components including collagens, proteoglycans and glycosaminoglycan binding molecules. We also observed that levels of hemoglobin chains, possibly related to defects in iron metabolism, were enriched in PD brains. These findings shed important new light on disease processes that occur in association with PD.
Collapse
Affiliation(s)
- Rekha Raghunathan
- Graduate Program in Molecular and Translational Medicine, Boston University School of Medicine, Boston, 02118, USA
| | - John D Hogan
- Bioinformatics Program, Boston University Graduate School of Arts and Sciences, Boston, 02118, USA
| | - Adam Labadorf
- Bioinformatics Program, Boston University Graduate School of Arts and Sciences, Boston, 02118, USA.,Department of Neurology, Boston University School of Medicine, Boston, 02118, USA
| | - Richard H Myers
- Graduate Program in Molecular and Translational Medicine, Boston University School of Medicine, Boston, 02118, USA.,Bioinformatics Program, Boston University Graduate School of Arts and Sciences, Boston, 02118, USA.,Department of Neurology, Boston University School of Medicine, Boston, 02118, USA
| | - Joseph Zaia
- Graduate Program in Molecular and Translational Medicine, Boston University School of Medicine, Boston, 02118, USA. .,Department of Biochemistry, Boston University School of Medicine, 670 Albany St., Rm. 509, Boston, 02118, USA. .,Bioinformatics Program, Boston University Graduate School of Arts and Sciences, Boston, 02118, USA.
| |
Collapse
|
37
|
Driver-Dunckley ED, Zhang N, Adler CH, Serrano GE, Sue LI, Shill HA, Mehta SH, Belden CM, Zamrini EY, Davis K, Beach TG. Brain Lewy-Type Synucleinopathy Density Is Associated with a Lower Prevalence of Atherosclerotic Cardiovascular Disease Risk Factors in Patients with Parkinson's Disease1. JOURNAL OF PARKINSONS DISEASE 2020; 9:543-552. [PMID: 31282425 DOI: 10.3233/jpd-191610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Some epidemiology studies suggest that atherosclerotic cardiovascular disease (ASCVD) risk factors increase the risk of developing Parkinson's disease (PD). However, conflicting data suggest lower rates of ASCVD in PD. OBJECTIVE The objective of this study is to determine, with data from a longitudinal clinicopathological study, whether ASCVD risk factors are associated with a PD diagnosis and/or increased brain or peripheral load of Lewy-type synucleinopathy (LTS). METHODS All subjects were followed to autopsy and neuropathological examination in the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND). Multivariable regression models, including age, gender, and smoking history, were used to investigate the association of a PD diagnosis or brain or submandibular gland LTS load with ASCVD risk factors. RESULTS 150 subjects were included (PD n = 60, controls n = 90). Univariable comparisons and regression models showed a general trend to inverse associations. The multivariable odds ratio (OR) of brain LTS load for carotid artery disease was 0.93 (95% CI: 0.86 to 0.98; p = 0.02), for anticoagulant use 0.95 (95% CI: 0.90 to 0.99; p = 0.04) and for abnormal heart weight 0.96 (95% CI: 0.92 to 0.99; p = 0.01). Composite clinical and overall (clinical + pathology composite risk scores) composite risk scores were also significantly lower in the PD subjects (p = 0.0164 and 0.0187, respectively). Submandibular gland LTS load was not significantly related to ASCVD conditions. CONCLUSIONS This study shows associations of higher brain LTS with lower prevalence of both clinical and pathological indices of ASCVD in PD subjects versus age-similar controls. We suggest that this is due to α-synuclein pathology-induced sympathetic denervation in PD.
Collapse
Affiliation(s)
| | - Nan Zhang
- Section of Biostatistics, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Lucia I Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | | | | | - Christine M Belden
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Edward Y Zamrini
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Kathryn Davis
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| |
Collapse
|
38
|
Chouliaras L, Kumar GS, Thomas AJ, Lunnon K, Chinnery PF, O'Brien JT. Epigenetic regulation in the pathophysiology of Lewy body dementia. Prog Neurobiol 2020; 192:101822. [PMID: 32407744 DOI: 10.1016/j.pneurobio.2020.101822] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 04/09/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022]
Abstract
Lewy body dementia encompasses both dementia with Lewy bodies and Parkinson's disease dementia. Although both are common causes of dementia, they remain relatively understudied. The review summarises the clinico-pathologic characteristics of Lewy Body dementia and discusses the genetic and environmental evidence contributing to the risk of developing the condition. Considering that the pathophysiology of Lewy body dementia is not yet fully understood, here we focus on the role of epigenetic mechanisms as potential key mediators of gene-environment interactions in the development of the disease. We examine available important data on genomics, epigenomics, gene expression and proteomic studies in Lewy body dementia on human post-mortem brain and peripheral tissues. Genetic variation and epigenetic modifications in key genes involved in the disorder, such as apolipoprotein E (APOE), α-synuclein (SNCA) and glucocerobrosidase (GBA), suggest a central involvement of epigenetics in DLB but conclusive evidence is scarce. This is due to limitations of existing literature, such as small sample sizes, lack of replication and lack of studies interrogating cell-type specific epigenetic modifications in the brain. Future research in the field can improve the understanding of this common but complex and rapidly progressing type of dementia and potentially open early diagnostic and effective therapeutic targets.
Collapse
Affiliation(s)
| | - Gautham S Kumar
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Alan J Thomas
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Katie Lunnon
- College of Medicine and Health, University of Exeter Medical School, Exeter University, Exeter, UK
| | - Patrick F Chinnery
- Department of Clinical Neurosciences and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
39
|
Serrano GE, Shprecher D, Callan M, Cutler B, Glass M, Zhang N, Walker J, Intorcia A, Adler CH, Shill HA, Driver-Dunckley E, Mehta SH, Belden CM, Zamrini E, Sue LI, Vargas D, Beach TG. Cardiac sympathetic denervation and synucleinopathy in Alzheimer's disease with brain Lewy body disease. Brain Commun 2020; 2:fcaa004. [PMID: 32064463 PMCID: PMC7008146 DOI: 10.1093/braincomms/fcaa004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/11/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022] Open
Abstract
Comorbid Lewy body pathology is very common in Alzheimer’s disease and may confound clinical trial design, yet there is no in vivo test to identify patients with this. Tissue (and/or radioligand imaging) studies have shown cardiac sympathetic denervation in Parkinson’s disease and dementia with Lewy bodies, but this has not been explored in Alzheimer’s subjects with Lewy bodies not meeting dementia with Lewy bodies clinicopathological criteria. To determine if Alzheimer’s disease with Lewy bodies subjects show sympathetic cardiac denervation, we analysed epicardial and myocardial tissue from autopsy-confirmed cases using tyrosine hydroxylase and neurofilament immunostaining. Comparison of tyrosine hydroxylase fibre density in 19 subjects with Alzheimer’s disease/dementia with Lewy bodies, 20 Alzheimer’s disease with Lewy bodies, 12 Alzheimer’s disease subjects without Lewy body disease, 19 Parkinson’s disease, 30 incidental Lewy body disease and 22 cognitively normal without Alzheimer’s disease or Lewy body disease indicated a significant group difference (P < 0.01; Kruskal–Wallis analysis of variance) and subsequent pair-wise Mann–Whitney U tests showed that Parkinson’s disease (P < 0.05) and Alzheimer’s disease/dementia with Lewy bodies (P < 0.01) subjects, but not Alzheimer’s disease with Lewy bodies subjects, had significantly reduced tyrosine hydroxylase fibre density as compared with cognitively normal. Both Parkinson’s disease and Alzheimer’s disease/dementia with Lewy bodies subjects also showed significant epicardial losses of neurofilament protein-immunoreactive nerve fibre densities within the fibre bundles as compared with cognitively normal subjects (P < 0.01) and both groups showed high pathologic alpha-synuclein densities (P < 0.0001). Cardiac alpha-synuclein densities correlated significantly with brain alpha-synuclein (P < 0.001), while cardiac tyrosine hydroxylase and neurofilament immunoreactive nerve fibre densities were negatively correlated with the densities of both brain and cardiac alpha-synuclein, as well as Unified Parkinson’s Disease Rating Scale scores (P < 0.05). The clear separation of Alzheimer’s disease/dementia with Lewy bodies subjects from Alzheimer’s disease and cognitively normal, based on cardiac tyrosine hydroxylase fibre density, is the first report of a statistically significant difference between these groups. Our data do not show significant sympathetic cardiac denervation in Alzheimer’s disease with Lewy bodies, but strongly confirm that cardiac nuclear imaging with a noradrenergic radioligand is worthy of further study as a potential means to separate Alzheimer’s disease from Alzheimer’s disease/dementia with Lewy bodies during life.
Collapse
Affiliation(s)
- Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - David Shprecher
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Michael Callan
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Brett Cutler
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Michael Glass
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Nan Zhang
- Section of Biostatistics, Department of Health Science Research, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Jessica Walker
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Anthony Intorcia
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Holly A Shill
- Muhammad Ali Parkinson Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Erika Driver-Dunckley
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Shyamal H Mehta
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | - Christine M Belden
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Edward Zamrini
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Lucia I Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Daisy Vargas
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| |
Collapse
|
40
|
Patterns of Expression of Purinergic Receptor P2RY12, a Putative Marker for Non-Activated Microglia, in Aged and Alzheimer's Disease Brains. Int J Mol Sci 2020; 21:ijms21020678. [PMID: 31968618 PMCID: PMC7014248 DOI: 10.3390/ijms21020678] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is considered a key pathological process in neurodegenerative diseases of aging, including Alzheimer's disease (AD). Many studies have defined phenotypes of reactive microglia, the brain-resident macrophages, with different antigenic markers to identify those potentially causing inflammatory damage. We took an alternative approach with the goal of characterizing the distribution of purinergic receptor P2RY12-positive microglia, a marker previously defined as identifying homeostatic or non-activated microglia. We examined the expression of P2RY12 by dual-color light and fluorescence immunohistochemistry using sections of middle temporal gyrus from AD, high plaque and low plaque non-demented cases in relation to amyloid beta (Aβ) plaques and phosphorylated tau, markers of pathology, and HLA-DR, IBA-1, CD68, and progranulin, microglial phenotype markers. In low plaque cases, P2RY12-positive microglia mostly had non-activated morphologies, while the morphologies of P2RY12-positive microglia in AD brains were highly variable, suggesting its expression could encompass a wider range of phenotypes than originally hypothesized. P2RY12 expression by microglia differed depending on the types of plaques or tangles they were associated with. Areas of inflammation characterized by lack of P2RY12-positive microglia around mature plaques could be observed, but many diffuse plaques showed colocalization with P2RY12-positive microglia. Based on these results, P2RY12 expression by microglia should not be considered solely a marker of resting microglia as P2RY12 immunoreactivity was identifying microglia positive for CD68, progranulin and to a limited extent HLA-DR, markers of activation.
Collapse
|
41
|
Ornelas AS, Adler CH, Serrano GE, Curry JR, Shill HA, Kopyov O, Beach TG. Co-Existence of tau and α-synuclein pathology in fetal graft tissue at autopsy: A case report. Parkinsonism Relat Disord 2020; 71:36-39. [PMID: 32004818 DOI: 10.1016/j.parkreldis.2019.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 12/21/2019] [Accepted: 12/23/2019] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Transplant of fetal ventral mesencephalic tissue into the striatum of Parkinson's disease (PD) patients has been performed to increase dopamine production and stimulate neuronal regeneration. Analysis of fetal graft tissue at autopsy has demonstrated 6 cases of α-synuclein pathology in PD patients, one case with both α-synuclein and tau pathology in a PD patient, and two cases of tau pathology within a Huntington's Disease patient. METHODS A 49 year old man with PD underwent bilateral fetal ventral mesencephalic cell transplants into the striatum. Autopsy at age 70 included immunohistochemical staining of host and graft tissue with antibodies to phosphorylated α-synuclein and phosphorylated tau protein. RESULTS Autopsy confirmed the diagnosis of PD. Immunohistochemical staining of graft tissue demonstrated frequent neuronal perikaryal inclusions of phosphorylated α -synuclein and tau in the left graft only. CONCLUSION Speculations on the formation of pathology include: 1) α-synuclein and tau pathology spread from host to the graft in a neuron-neuron manner. 2) The nature of the fetal cells themselves, or transplantation process, may render fetal tissue more susceptible to the spontaneous generation of pathology. 3) Factors within host environment caused native tau and α-synuclein in fetal tissue graft to become phosphorylated.
Collapse
Affiliation(s)
- Abbie S Ornelas
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA.
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA.
| | - Geidy E Serrano
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, 10515 W Santa Fe Dr, Sun City, AZ, 85351, USA.
| | - Jasmine R Curry
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, 10515 W Santa Fe Dr, Sun City, AZ, 85351, USA.
| | - Holly A Shill
- Barrow Neurological Institute, 350 W. Thomas Rd, Phoenix, AZ, 85013, USA. https://
| | | | - Thomas G Beach
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, 10515 W Santa Fe Dr, Sun City, AZ, 85351, USA.
| |
Collapse
|
42
|
Manivannan B, Yegambaram M, Supowit S, Beach TG, Halden RU. Assessment of Persistent, Bioaccumulative and Toxic Organic Environmental Pollutants in Liver and Adipose Tissue of Alzheimer’s Disease Patients and Age-matched Controls. Curr Alzheimer Res 2019; 16:1039-1049. [DOI: 10.2174/1567205016666191010114744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 09/19/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Background:
Lifetime exposure to environmental (neuro) toxicants may contribute to the
pathogenesis of Alzheimer’s Disease (AD). Since many contaminants do not cross the blood-brain
barrier, brain tissue alone cannot serve to assess the spectrum of environmental exposures.
Methods:
We used liquid and gas chromatography tandem mass spectrometry to monitor, in postmortem
liver and adipose tissues of AD patients and age-matched controls, the occurrence and
concentrations of 11 environmental contaminants.
Results:
Seven toxicants were detected at 100% frequency: p,p'-DDE, dieldrin, triclosan,
methylparaben, bisphenol A, fipronil and tetrabromobisphenol A (TBBPA). Intra-individual, tissuedependent
differences were detected for triclosan, methylparaben, fipronil and TBBPA. High
concentrations of p,p’-DDE and dieldrin were observed in adipose tissue when compared to liver values
for both AD cases and controls.
Conclusion:
This study provides vital data on organ-specific human body burdens to select analytes and
demonstrate the feasibility of analyzing small sample quantities for toxicants suspected to constitute AD
risk factors.
Collapse
Affiliation(s)
- Bhagyashree Manivannan
- Biodesign Center for Environmental Health Engineering, Arizona State University, Tempe, AZ, United States
| | - Manivannan Yegambaram
- Biodesign Center for Environmental Health Engineering, Arizona State University, Tempe, AZ, United States
| | - Samuel Supowit
- Biodesign Center for Environmental Health Engineering, Arizona State University, Tempe, AZ, United States
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ-85287, United States
| | - Rolf U. Halden
- Biodesign Center for Environmental Health Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
43
|
Lardenoije R, Roubroeks JAY, Pishva E, Leber M, Wagner H, Iatrou A, Smith AR, Smith RG, Eijssen LMT, Kleineidam L, Kawalia A, Hoffmann P, Luck T, Riedel-Heller S, Jessen F, Maier W, Wagner M, Hurlemann R, Kenis G, Ali M, del Sol A, Mastroeni D, Delvaux E, Coleman PD, Mill J, Rutten BPF, Lunnon K, Ramirez A, van den Hove DLA. Alzheimer's disease-associated (hydroxy)methylomic changes in the brain and blood. Clin Epigenetics 2019; 11:164. [PMID: 31775875 PMCID: PMC6880587 DOI: 10.1186/s13148-019-0755-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/26/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Late-onset Alzheimer's disease (AD) is a complex multifactorial affliction, the pathogenesis of which is thought to involve gene-environment interactions that might be captured in the epigenome. The present study investigated epigenome-wide patterns of DNA methylation (5-methylcytosine, 5mC) and hydroxymethylation (5-hydroxymethylcytosine, 5hmC), as well as the abundance of unmodified cytosine (UC), in relation to AD. RESULTS We identified epigenetic differences in AD patients (n = 45) as compared to age-matched controls (n = 35) in the middle temporal gyrus, pertaining to genomic regions close to or overlapping with genes such as OXT (- 3.76% 5mC, pŠidák = 1.07E-06), CHRNB1 (+ 1.46% 5hmC, pŠidák = 4.01E-04), RHBDF2 (- 3.45% UC, pŠidák = 4.85E-06), and C3 (- 1.20% UC, pŠidák = 1.57E-03). In parallel, in an independent cohort, we compared the blood methylome of converters to AD dementia (n = 54) and non-converters (n = 42), at a preclinical stage. DNA methylation in the same region of the OXT promoter as found in the brain was found to be associated with subsequent conversion to AD dementia in the blood of elderly, non-demented individuals (+ 3.43% 5mC, pŠidák = 7.14E-04). CONCLUSIONS The implication of genome-wide significant differential methylation of OXT, encoding oxytocin, in two independent cohorts indicates it is a promising target for future studies on early biomarkers and novel therapeutic strategies in AD.
Collapse
Affiliation(s)
- Roy Lardenoije
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Janou A. Y. Roubroeks
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Ehsan Pishva
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Markus Leber
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
| | - Holger Wagner
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
| | - Artemis Iatrou
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Adam R. Smith
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Rebecca G. Smith
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Lars M. T. Eijssen
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
- Department of Bioinformatics—BiGCaT, Maastricht University, Maastricht, The Netherlands
| | - Luca Kleineidam
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Amit Kawalia
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany
- Division of Medical Genetics, University Hospital and Department of Biomedicine, University of Basel, CH-4058 Basel, Switzerland
| | - Tobias Luck
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, 04103 Leipzig, Germany
| | - Steffi Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, 04103 Leipzig, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
| | - Wolfgang Maier
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Michael Wagner
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - René Hurlemann
- Department of Psychiatry and Division of Medical Psychology, University of Bonn, 53105 Bonn, Germany
| | - Gunter Kenis
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Muhammad Ali
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow, Russian Federation
- CIC bioGUNE, Bizkaia Technology Park, 801 Building, 48160 Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Dolgoprudny Bilbao, Spain
| | - Diego Mastroeni
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
- L.J. Roberts Center for Alzheimer’s Research Banner Sun Health Research Institute, Sun City, AZ USA
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ USA
| | - Elaine Delvaux
- L.J. Roberts Center for Alzheimer’s Research Banner Sun Health Research Institute, Sun City, AZ USA
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ USA
| | - Paul D. Coleman
- L.J. Roberts Center for Alzheimer’s Research Banner Sun Health Research Institute, Sun City, AZ USA
- Biodesign Institute, Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ USA
| | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, Exeter, UK
- Institute of Psychiatry, King’s College London, London, UK
| | - Bart P. F. Rutten
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Katie Lunnon
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Medical Faculty, 50937 Cologne, Germany
- Department of Neurodegeneration and Gerontopsychiatry, University of Bonn, 53127 Bonn, Germany
| | - Daniël L. A. van den Hove
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| |
Collapse
|
44
|
Manne S, Kondru N, Jin H, Anantharam V, Huang X, Kanthasamy A, Kanthasamy AG. α-Synuclein real-time quaking-induced conversion in the submandibular glands of Parkinson's disease patients. Mov Disord 2019; 35:268-278. [PMID: 31758740 DOI: 10.1002/mds.27907] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Identification of a peripheral biomarker is a major roadblock in the diagnosis of PD. Immunohistological identification of p-serine 129 α-synuclein in the submandibular gland tissues of PD patients has been recently reported. OBJECTIVE We report on a proof-of-principle study for using an ultra-sensitive and specific, real-time quaking-induced conversion assay to detect pathological α-synuclein in the submandibular gland tissues of PD patients. METHODS The α-synuclein real-time quaking-induced conversion assay was used to detect and quantify pathological α-synuclein levels in PD, incidental Lewy body disease, and control submandibular gland tissues as well as in formalin-fixed paraffin-embedded sections. RESULTS We determined the quantitative seeding kinetics of pathological α-synuclein present in submandibular gland tissues from autopsied subjects using the α-synuclein real-time quaking-induced conversion assay. A total of 32 cases comprising 13 PD, 3 incidental Lewy body disease, and 16 controls showed 100% sensitivity and 94% specificity. Interestingly, both PD and incidental Lewy body disease tissues showed 100% concordance for elevated levels of pathological α-synuclein seeding activity compared to control tissues. End-point dilution kinetic analyses revealed that the submandibular gland had a wide dynamic range of pathological α-synuclein seeding activity. CONCLUSIONS Our results are the first to demonstrate the utility of using the real-time quaking-induced conversion assay on peripherally accessible submandibular gland tissues and formalin-fixed paraffin-embedded tissue sections to detect PD-related pathological changes with high sensitivity and specificity. Additionally, the detection of seeding activity from incidental Lewy body disease cases containing immunohistochemically undetected pathological α-synuclein demonstrates the α-synuclein real-time quaking-induced conversion assay's potential utility for identifying prodromal PD in submandibular gland tissues. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sireesha Manne
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Naveen Kondru
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Xuemei Huang
- Department of Neurology and Pharmacology, Neurosurgery, Radiology, and Kinesiology, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
45
|
African-American and Caucasian participation in postmortem human brain donation for neuropsychiatric research. PLoS One 2019; 14:e0222565. [PMID: 31644530 PMCID: PMC6808324 DOI: 10.1371/journal.pone.0222565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 09/03/2019] [Indexed: 01/15/2023] Open
Abstract
Increased African-American research participation is critical to the applicability and generalizability of biomedical research, as population diversity continues to increase both domestically and abroad. Yet numerous studies document historical origins of mistrust, as well as other barriers that may contribute to resistance in the African-American community towards participation in biomedical research. However, a growing body of more recent scientific evidence suggests that African-Americans value research and are willing to participate when asked. In the present study, we set out to determine factors associated with research participation of African-American families in postmortem human brain tissue donation for neuropsychiatric disorders as compared with Caucasian families, from same-day medical examiner autopsy referrals. We retrospectively reviewed brain donation rates, as well as demographic and clinical factors associated with donation in 1,421 consecutive referrals to three medical examiner’s offices from 2010–2015. Overall, 69.7% of all next-of-kin contacted agreed to brain donation. While Caucasian families consented to donate brain tissue at a significantly higher rate (74.1%) than African-American families (57.0%) (p<0.001), African-American brain donation rates were as high as 60.5% in referrals from Maryland. Neither African-American nor Caucasian donors differed significantly from non-donors on any demographic or clinical factors ascertained, including age, sex, diagnosis of the donor, or in the relationship of the next-of-kin being contacted (p>0.05). However, Caucasian donors were significantly older, had more years of education, were more likely to be referred for study due to a psychiatric diagnosis, more likely to have comorbid substance abuse, and more likely to have died via suicide, as compared with African-American donors (p<0.05). When African-American participants are identified and approached, African-American families as well as Caucasian families are indeed willing to donate brain tissue on the spot for neuropsychiatric research, which supports the belief that African-American attitudes towards biomedical research may be more favorable than previously thought.
Collapse
|
46
|
Schlenzig D, Cynis H, Hartlage-Rübsamen M, Zeitschel U, Menge K, Fothe A, Ramsbeck D, Spahn C, Wermann M, Roßner S, Buchholz M, Schilling S, Demuth HU. Dipeptidyl-Peptidase Activity of Meprin β Links N-truncation of Aβ with Glutaminyl Cyclase-Catalyzed pGlu-Aβ Formation. J Alzheimers Dis 2019; 66:359-375. [PMID: 30320570 DOI: 10.3233/jad-171183] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The formation of amyloid-β (Aβ) peptides is causally involved in the development of Alzheimer's disease (AD). A significant proportion of deposited Aβ is N-terminally truncated and modified at the N-terminus by a pGlu-residue (pGlu-Aβ). These forms show enhanced neurotoxicity compared to full-length Aβ. Although the truncation may occur by aminopeptidases after formation of Aβ, recently discovered processing pathways of amyloid-β protein precursor (AβPP) by proteases such as meprin β may also be involved. Here, we assessed a role of meprin β in forming Aβ3-40/42, which is the precursor of pGlu-Aβ3-40/42 generated by glutaminyl cyclase (QC). Similar to QC, meprin β mRNA is significantly upregulated in postmortem brain from AD patients. A histochemical analysis supports the presence of meprin β in neurons and astrocytes in the vicinity of pGlu-Aβ containing deposits. Cleavage of AβPP-derived peptides by meprin β in vitro results in peptides Aβ1-x, Aβ2-x, and Aβ3-x. The formation of N-truncated Aβ by meprin β was also corroborated in cell culture. A subset of the generated peptides was converted into pGlu-Aβ3-40 by an addition of glutaminyl cyclase, supporting the preceding formation of Aβ3-40. Further analysis of the meprin β cleavage revealed a yet unknown dipeptidyl-peptidase-like activity specific for the N-terminus of Aβ1-x. Thus, our data suggest that meprin β contributes to the formation of N-truncated Aβ by endopeptidase and exopeptidase activity to generate the substrate for QC-catalyzed pGlu-Aβ formation.
Collapse
Affiliation(s)
- Dagmar Schlenzig
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Holger Cynis
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | | | | | - Katja Menge
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Anja Fothe
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Daniel Ramsbeck
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Claudia Spahn
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Michael Wermann
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, Leipzig, Germany
| | - Mirko Buchholz
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Stephan Schilling
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Hans-Ulrich Demuth
- Department of Molecular Drug Design and Target Validation Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| |
Collapse
|
47
|
Driver-Dunckley ED, Zhang N, Shill HA, Mehta SH, Belden CM, Zamrini EY, Davis K, Beach TG, Adler CH. Correlation Between the Movement Disorder Society's Unified Parkinson's Disease Rating Scale and Nonmotor Scales in Patients with Parkinson's Disease. INNOVATIONS IN CLINICAL NEUROSCIENCE 2019; 16:27-29. [PMID: 32082946 PMCID: PMC7009327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background: The Movement Disorder Society's Unified Parkinson's Disease Rating Scale (MDSUPDRS), Scales for Outcomes in Parkinson's Disease-Autonomic (SCOPA-AUT), Mayo Sleep Questionnaire, Epworth Sleepiness Scale, and Neuropsychiatric Inventory Questionnaire (NPI-Q) are validated instruments for assessing signs and symptoms of Parkinson's disease (PD). Objective: We sought to determine whether responses on the MDS-UPDRS correlate with responses to other scales used in patients with PD. Design: Study subjects were enrolled in the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND). Participants were selected if they had completed all scales within a one-month window. Spearman's rank correlation coefficients were calculated. Results: A total of 96 eligible subjects were identified. High correlation (r-values) was found between the SCOPA-AUT and MDS-UPDRS excessive saliva (0.73; p<0.001), constipation (0.62; p<0.001), and swallowing (0.59; p<0.001) questions. The r-values for the NPI-Q and MDS-UPDRS depression and anxiety questions were 0.53 (p<0.001), and 0.67 (p<0.001). Conclusion: MDS-UPDRS correlates well with some but not all questions from the SCOPA-AUT and NPI-Q. This work emphasizes the importance of employing multiple methods for assessing nonmotor symptoms in patients with PD.
Collapse
Affiliation(s)
- Erika D Driver-Dunckley
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Nan Zhang
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Holly A Shill
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Shyamal H Mehta
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Christine M Belden
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Edward Y Zamrini
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Kathryn Davis
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Thomas G Beach
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| | - Charles H Adler
- Drs. Driver-Dunckley, Mehta, and Adler are with the Department of Neurology of the Parkinson's Disease and Movement Disorders Center at the Mayo Clinic in Scottsdale, Arizona. Zhang is with the Section of Biostatistics at the Mayo Clinic in Scottsdale, Arizona. Shill is with the Barrow Neurological Institute in Phoenix, Arizona. Belden, Zamrini, Davis, and Beach are with the Banner Sun Health Research Institute in Sun City, Arizona
| |
Collapse
|
48
|
Miron J, Picard C, Labonté A, Auld D, Breitner J, Poirier J. Association of PPP2R1A with Alzheimer's disease and specific cognitive domains. Neurobiol Aging 2019; 81:234-243. [PMID: 31349112 DOI: 10.1016/j.neurobiolaging.2019.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/19/2019] [Accepted: 06/23/2019] [Indexed: 01/20/2023]
Abstract
In an attempt to identify novel genetic variants associated with sporadic Alzheimer's disease (AD), a genome-wide association study was performed on a population isolate from Eastern Canada, referred to as the Québec Founder Population (QFP). In the QFP cohort, the rs10406151 C variant on chromosome 19 is associated with higher AD risk and younger age at AD onset in APOE4- individuals. After surveying the region surrounding this intergenic polymorphism for brain cis-eQTL associations in BRAINEAC, we identified PPP2R1A as the most likely target gene modulated by the rs10406151 C variant. PPP2R1A mRNA and protein levels are elevated in multiple regions from QFP autopsy-confirmed AD brains when compared with age-matched controls. Using an independent cohort of cognitively normal individuals with a parental history of AD, we found that the rs10406151 C variant is significantly associated with lower visuospatial and constructional performances. The association of the rs10406151 C variant with AD risk appears to involve brain PPP2R1A gene expression alterations. However, the exact pathological pathway by which this variant modulates AD remains elusive.
Collapse
Affiliation(s)
- Justin Miron
- Douglas Hospital Research Centre, Montréal, Canada; Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, Canada; McGill University, Montréal, Canada
| | - Cynthia Picard
- Douglas Hospital Research Centre, Montréal, Canada; Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, Canada; McGill University, Montréal, Canada
| | - Anne Labonté
- Douglas Hospital Research Centre, Montréal, Canada; Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, Canada
| | - Daniel Auld
- McGill University and Génome Québec Innovation Centre, Montréal, Canada
| | - John Breitner
- Douglas Hospital Research Centre, Montréal, Canada; Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, Canada; McGill University, Montréal, Canada
| | - Judes Poirier
- Douglas Hospital Research Centre, Montréal, Canada; Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, Canada; McGill University, Montréal, Canada.
| |
Collapse
|
49
|
Ohi K, Kuwata A, Shimada T, Kataoka Y, Yasuyama T, Uehara T, Kawasaki Y. Genome-Wide Variants Shared Between Smoking Quantity and Schizophrenia on 15q25 Are Associated With CHRNA5 Expression in the Brain. Schizophr Bull 2019; 45:813-823. [PMID: 30202994 PMCID: PMC6581148 DOI: 10.1093/schbul/sby093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cigarette smokers with schizophrenia consume more cigarettes than smokers in the general population. Schizophrenia and smoking quantity may have shared genetic liability. Genome-wide association studies (GWASs) of schizophrenia and smoking quantity have highlighted a biological pleiotropy in which a robust 15q25 locus affects both traits. To identify the genetic variants shared between these traits on 15q25, we used summary statistics from large-scale GWAS meta-analyses of schizophrenia in the Psychiatric Genomics Consortium 2 and smoking quantity assessed by cigarettes smoked per day in the Tobacco and Genetics Consortium. To evaluate the regulatory potential of the shared genetic variants, expression quantitative trait loci analysis in 10 postmortem brain regions was performed using the BRAINEAC dataset in 134 neuropathologically normal individuals. Twenty-two genetic variants on 15q25 were associated with both smoking quantity and schizophrenia at the genome-wide significance level (P < 5.00 × 10-8). Major alleles of all variants were associated with higher smoking quantity and risk of schizophrenia. These genetic variants were associated with PSMA4, CHRNA3, and CHRNB4 expression in specific brain regions (lowest P = 4.81 × 10-4) and with CHRNA5 expression in multiple brain regions (lowest P = 8.70 × 10-6). Risk-associated major alleles of these variants were commonly associated with higher expression in several brain regions, excluding the medulla, at the transcript level. In addition, the risk-associated major allele at rs637137 was associated with higher CHRNA5 expression at the specific exon level in multiple brain regions (lowest P = 2.37 × 10-5). Our findings suggest that genome-wide variants shared between smoking quantity and schizophrenia contribute to a common pathophysiology underlying these traits involving altered CHRNA5 expression in the brain.
Collapse
Affiliation(s)
- Kazutaka Ohi
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan,Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan,To whom correspondence should be addressed; Department of Neuropsychiatry, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan; tel: +81-76-286-2211, fax: +81-76-286-3341, e-mail:
| | - Aki Kuwata
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Takamitsu Shimada
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Yuzuru Kataoka
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Toshiki Yasuyama
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Takashi Uehara
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| | - Yasuhiro Kawasaki
- Department of Neuropsychiatry, Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|
50
|
Glorioso CA, Pfenning AR, Lee SS, Bennett DA, Sibille EL, Kellis M, Guarente LP. Rate of brain aging and APOE ε4 are synergistic risk factors for Alzheimer's disease. Life Sci Alliance 2019; 2:2/3/e201900303. [PMID: 31133613 PMCID: PMC6537750 DOI: 10.26508/lsa.201900303] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
This study describes a gauge for brain aging predictive of cognitive decline and Alzheimer's disease synergistic with the APOE ε4 allele. Advanced age and the APOE ε4 allele are the two biggest risk factors for Alzheimer’s disease (AD) and declining cognitive function. We describe a universal gauge to measure molecular brain age using transcriptome analysis of four human postmortem cohorts (n = 673, ages 25–97) free of neurological disease. In a fifth cohort of older subjects with or without neurological disease (n = 438, ages 67–108), we show that subjects with brains deviating in the older direction from what would be expected based on chronological age show an increase in AD, Parkinson’s disease, and cognitive decline. Strikingly, a younger molecular age (−5 yr than chronological age) protects against AD even in the presence of APOE ε4. An established DNA methylation gauge for age correlates well with the transcriptome gauge for determination of molecular age and assigning deviations from the expected. Our results suggest that rapid brain aging and APOE ε4 are synergistic risk factors, and interventions that slow aging may substantially reduce risk of neurological disease and decline even in the presence of APOE ε4.
Collapse
Affiliation(s)
- Christin A Glorioso
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA .,Paul F. Glenn Center for Biology of Aging Research at Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andreas R Pfenning
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Sam S Lee
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Paul F. Glenn Center for Biology of Aging Research at Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Etienne L Sibille
- Department of Psychiatry and of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.,Campbell Family Mental Health Research Institute, Toronto, Ontario, Canada
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA.,The Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Leonard P Guarente
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA .,Paul F. Glenn Center for Biology of Aging Research at Massachusetts Institute of Technology, Cambridge, MA, USA.,The Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|