1
|
Du J, Nakachi Y, Murata Y, Kiyota E, Kato T, Bundo M, Iwamoto K. Exploration of cell type-specific somatic mutations in schizophrenia and the impact of maternal immune activation on the somatic mutation profile in the brain. Psychiatry Clin Neurosci 2024; 78:237-247. [PMID: 38334156 DOI: 10.1111/pcn.13640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 02/10/2024]
Abstract
AIM Schizophrenia (SZ) is a severe psychiatric disorder caused by the interaction of genetic and environmental factors. Although somatic mutations that occur in the brain after fertilization may play an important role in the cause of SZ, their frequencies and patterns in the brains of patients and related animal models have not been well studied. This study aimed to find somatic mutations related to the pathophysiology of SZ. METHODS We performed whole-exome sequencing (WES) of neuronal and nonneuronal nuclei isolated from the postmortem prefrontal cortex of patients with SZ (n = 10) and controls (n = 10). After detecting somatic mutations, we explored the similarities and differences in shared common mutations between two cell types and cell type-specific mutations. We also performed WES of prefrontal cortex samples from an animal model of SZ based on maternal immune activation (MIA) and explored the possible impact of MIA on the patterns of somatic mutations. RESULTS We did not find quantitative differences in somatic mutations but found higher variant allele fractions of neuron-specific mutations in patients with SZ. In the mouse model, we found a larger variation in the number of somatic mutations in the offspring of MIA mice, with the occurrence of somatic mutations in neurodevelopment-related genes. CONCLUSION Somatic mutations occurring at an earlier stage of brain cell differentiation toward neurons may be important for the cause of SZ. MIA may affect somatic mutation profiles in the brain.
Collapse
Affiliation(s)
- Jianbin Du
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yutaka Nakachi
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yui Murata
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
| | - Emi Kiyota
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miki Bundo
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
| | - Kazuya Iwamoto
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
| |
Collapse
|
2
|
Wang P, Yao M, Yuan J, Han F, Zhai F, Zhang D, Zhou L, Ni J, Zhang S, Cui L, Zhu Y. Association of Rare NOTCH3 Variants With Prevalent and Incident Stroke and Dementia in the General Population. J Am Heart Assoc 2024; 13:e032668. [PMID: 38348813 PMCID: PMC11010104 DOI: 10.1161/jaha.123.032668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/05/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND It is uncertain whether rare NOTCH3 variants are associated with stroke and dementia in the general population and whether they lead to alterations in cognitive function. This study aims to determine the associations of rare NOTCH3 variants with prevalent and incident stroke and dementia, as well as cognitive function changes. METHODS AND RESULTS In the prospective community-based Shunyi Study, a total of 1007 participants were included in the baseline analysis. For the follow-up analysis, 1007 participants were included in the stroke analysis, and 870 participants in the dementia analysis. All participants underwent baseline brain magnetic resonance imaging, carotid ultrasound, and whole exome sequencing. Rare NOTCH3 variants were defined as variants with minor allele frequency <1%. A total of 137 rare NOTCH3 carriers were enrolled in the baseline study. At baseline, rare NOTCH3 variant carriers had higher rates of stroke (8.8% versus 5.6%) and dementia (2.9% versus 0.8%) compared with noncarriers. After adjustment for associated risk factors, the epidermal growth factor-like repeats (EGFr)-involving rare NOTCH3 variants were associated with a higher risk of prevalent stroke (odds ratio [OR], 2.697 [95% CI, 1.266-5.745]; P=0.040) and dementia (OR, 8.498 [95% CI, 1.727-41.812]; P=0.032). After 5 years of follow-up, we did not find that the rare NOTCH3 variants increased the risk of incident stroke and dementia. There was no statistical difference in the change in longitudinal cognitive scale scores. CONCLUSIONS Rare NOTCH3 EGFr-involving variants are genetic risk factors for stroke and dementia in the general Chinese population.
Collapse
Affiliation(s)
- Pei Wang
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ming Yao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jing Yuan
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fei Han
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fei‐Fei Zhai
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ding‐Ding Zhang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li‐Xin Zhou
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jun Ni
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shu‐Yang Zhang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li‐Ying Cui
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yi‐Cheng Zhu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
3
|
Area-Gomez E, Schon EA. Towards a Unitary Hypothesis of Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2024; 98:1243-1275. [PMID: 38578892 DOI: 10.3233/jad-231318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
The "amyloid cascade" hypothesis of Alzheimer's disease (AD) pathogenesis invokes the accumulation in the brain of plaques (containing the amyloid-β protein precursor [AβPP] cleavage product amyloid-β [Aβ]) and tangles (containing hyperphosphorylated tau) as drivers of pathogenesis. However, the poor track record of clinical trials based on this hypothesis suggests that the accumulation of these peptides is not the only cause of AD. Here, an alternative hypothesis is proposed in which the AβPP cleavage product C99, not Aβ, is the main culprit, via its role as a regulator of cholesterol metabolism. C99, which is a cholesterol sensor, promotes the formation of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a cholesterol-rich lipid raft-like subdomain of the ER that communicates, both physically and biochemically, with mitochondria. We propose that in early-onset AD (EOAD), MAM-localized C99 is elevated above normal levels, resulting in increased transport of cholesterol from the plasma membrane to membranes of intracellular organelles, such as ER/endosomes, thereby upregulating MAM function and driving pathology. By the same token, late-onset AD (LOAD) is triggered by any genetic variant that increases the accumulation of intracellular cholesterol that, in turn, boosts the levels of C99 and again upregulates MAM function. Thus, the functional cause of AD is upregulated MAM function that, in turn, causes the hallmark disease phenotypes, including the plaques and tangles. Accordingly, the MAM hypothesis invokes two key interrelated elements, C99 and cholesterol, that converge at the MAM to drive AD pathogenesis. From this perspective, AD is, at bottom, a lipid disorder.
Collapse
Affiliation(s)
- Estela Area-Gomez
- Department of Neurology, Columbia University, New York, NY, USA
- Centro de Investigaciones Biológicas "Margarita Salas", Spanish National Research Council, Madrid, Spain
| | - Eric A Schon
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Genetics and Development>, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Karachanak-Yankova S, Serbezov D, Mihaylova M, Nikolova D, Balabanski L, Damyanova V, Antonova O, Staneva R, Ganev M, Spasova V, Rukova B, Nesheva D, Josifovska S, Stancheva M, Belejanska D, Petrova M, Mehrabian S, Traykov L, Hadjidekova S, Toncheva D. Detection of pathogenic variants in Alzheimer’s disease related genes in Bulgarian patients by pooled whole-exome sequencing. BIOTECHNOL BIOTEC EQ 2023. [DOI: 10.1080/13102818.2022.2155572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Sena Karachanak-Yankova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
- Department of Genetics, Faculty of Biology, Sofia University ‘St. Kliment Ohridski’, Sofia, Bulgaria
| | - Dimitar Serbezov
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Marta Mihaylova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Dragomira Nikolova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | | | - Vera Damyanova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Olga Antonova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Rada Staneva
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Mihail Ganev
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Victoria Spasova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Blaga Rukova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Desislava Nesheva
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Slavica Josifovska
- Department of Molecular Biology, Genetics and Microbiology, Faculty of Natural Sciences and Mathematics, ‘Ss. Cyril and Methodius’ University, Skopje, North Macedonia
| | - Mikaela Stancheva
- Department of Genetics, Faculty of Biology, Sofia University ‘St. Kliment Ohridski’, Sofia, Bulgaria
| | - Diana Belejanska
- Department of Neurology, University Hospital ‘Alexandrovska’, Sofia, Bulgaria
| | - Mariya Petrova
- Department of Neurology, University Hospital ‘Alexandrovska’, Sofia, Bulgaria
| | - Shima Mehrabian
- Department of Neurology, University Hospital ‘Alexandrovska’, Sofia, Bulgaria
| | - Latchezar Traykov
- Department of Neurology, University Hospital ‘Alexandrovska’, Sofia, Bulgaria
| | - Savina Hadjidekova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Draga Toncheva
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
- Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
5
|
Li D, Farrell JJ, Mez J, Martin ER, Bush WS, Ruiz A, Boada M, de Rojas I, Mayeux R, Haines JL, Vance MAP, Wang LS, Schellenberg GD, Lunetta KL, Farrer LA. Novel loci for Alzheimer's disease identified by a genome-wide association study in Ashkenazi Jews. Alzheimers Dement 2023; 19:5550-5562. [PMID: 37260021 PMCID: PMC10689571 DOI: 10.1002/alz.13117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 06/02/2023]
Abstract
INTRODUCTION Most Alzheimer's disease (AD) loci have been discovered in individuals with European ancestry (EA). METHODS We applied principal component analysis using Gaussian mixture models and an Ashkenazi Jewish (AJ) reference genome-wide association study (GWAS) data set to identify Ashkenazi Jews ascertained in GWAS (n = 42,682), whole genome sequencing (WGS, n = 16,815), and whole exome sequencing (WES, n = 20,504) data sets. The association of AD was tested genome wide (GW) in the GWAS and WGS data sets and exome wide (EW) in all three data sets (EW). Gene-based analyses were performed using aggregated rare variants. RESULTS In addition to apolipoprotein E (APOE), GW analyses (1355 cases and 1661 controls) revealed associations with TREM2 R47H (p = 9.66 × 10-9 ), rs541586606 near RAB3B (p = 5.01 × 10-8 ), and rs760573036 between SPOCK3 and ANXA10 (p = 6.32 × 10-8 ). In EW analyses (1504 cases and 2047 controls), study-wide significant association was observed with rs1003710 near SMAP2 (p = 1.91 × 10-7 ). A significant gene-based association was identified with GIPR (p = 7.34 × 10-7 ). DISCUSSION Our results highlight the efficacy of founder populations for AD genetic studies.
Collapse
Affiliation(s)
- Donghe Li
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - John J Farrell
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Jesse Mez
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Eden R. Martin
- Dr. John T. Macdonald Foundation, University of Miami, Miami, FL 33136, USA
- Department of Human Genetics, University of Miami, Miami, FL 33136, USA
| | - William S. Bush
- Department of Population & Quantitative Health Science and Cleveland Institute for Computational Biology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Agustin Ruiz
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Richard Mayeux
- Taub Institute on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center Department of Neurology, Columbia University, 710 West 168th Street, New York, NY 10032, USA
| | - Jonathan L. Haines
- Department of Population & Quantitative Health Science and Cleveland Institute for Computational Biology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Margaret A. Pericak Vance
- Dr. John T. Macdonald Foundation, University of Miami, Miami, FL 33136, USA
- Department of Human Genetics, University of Miami, Miami, FL 33136, USA
- Department of Neurology, University of Miami, Miami, FL 33136, USA
| | - Li-San Wang
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Gerard D. Schellenberg
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| |
Collapse
|
6
|
Swain PS, Panda S, Pati S, Dehury B. Computational saturation mutagenesis to explore the effect of pathogenic mutations on extra-cellular domains of TREM2 associated with Alzheimer's and Nasu-Hakola disease. J Mol Model 2023; 29:360. [PMID: 37924367 DOI: 10.1007/s00894-023-05770-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023]
Abstract
CONTEXT The specialised family of triggering receptors expressed on myeloid cells (TREMs) plays a pivotal role in causing neurodegenerative disorders and activating microglial anti-inflammatory responses. Nasu-Hakola disease (NHD), a rare autosomal recessive disorder, has been associated with mutations in TREM2, which is also responsible for raising the risk of Alzheimer's disease (AD). Herein, we have made an endeavour to differentiate the confirmed pathogenic variants in TREM2 extra-cellular domain (ECD) linked with NHD and AD using mutation-induced fold stability change (∆∆G), with the computation of 12distinct structure-based methods through saturation mutagenesis. Correlation analysis between relative solvent accessibility (RSA) and ∆∆G expresses the discrete distributive behaviour of mutants associated with TREM2 in AD (R2 = 0.061) and NHD (R2 = 0.601). Our findings put an emphasis on W50 and V126 as major players in maintaining V-like domain in TREM2. Interestingly, we discern that both of them interact with a common residue Y108, which is dissolved upon mutation. This Y108 could have structural or functional role for TREM2 which can be an ideal candidate for further study. Furthermore, the residual interaction network highlights the importance of R47 and R62 in maintaining the CDR loops that are crucial for ligand binding. Future studies using biophysical characterisation of ligand interactions in TREM2-ECD would be helpful for the development of novel therapeutics for AD and NHD. METHODS ConSurf algorithm and ENDscript were used to determine the position and conservation of each residue in the wild-type ECD of TREM2. The mutation-induced fold stability change (∆∆G) of confirmed pathogenic mutants associated with NHD and AD was estimated using 12 state-of-the-art structure-based protein stability tools. Furthermore, we also computed the effect of random mutation on these sites using computational saturation mutagenesis. Linear regression analysis was performed using mutants ∆∆G and RSA through GraphPad software. In addition, a comprehensive non-bonded residual interaction network (RIN) of wild type and its mutants of TREM2-ECD was enumerated using RING3.0.
Collapse
Affiliation(s)
- Preety Sthutika Swain
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, 751023, Odisha, India
| | - Sunita Panda
- Mycology Laboratory, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, 751023, Odisha, India
| | - Sanghamitra Pati
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, 751023, Odisha, India.
| | - Budheswar Dehury
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, 751023, Odisha, India.
| |
Collapse
|
7
|
Myllykangas L. Alzheimer's disease with a CADASIL twist - neuropathological examination proves its value in clarifying the genetics behind familial dementia. J Neurol Sci 2023; 452:120769. [PMID: 37625337 DOI: 10.1016/j.jns.2023.120769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Affiliation(s)
- Liisa Myllykangas
- Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki, Finland.
| |
Collapse
|
8
|
Korvatska O, Bucks SA, Yoda RA, Nolan A, Dorschner MO, Tsuang D, Jayadev S, Raskind WH, Bird TD. NOTCH3 C201R variant causes cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) that can be confused with early-onset Alzheimer's disease. J Neurol Sci 2023; 452:120763. [PMID: 37598468 PMCID: PMC10863424 DOI: 10.1016/j.jns.2023.120763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/08/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND NOTCH3 is the causative gene for autosomal dominant cerebral arteriopathy with subcortical infarctions and leukoencephalopathy (CADASIL) which is associated with both stroke and dementia. When CADASIL presents primarily as dementia it can be difficult to distinguish from Alzheimer's disease (AD) at both the clinical and neuropathological levels. METHODS We performed exome sequencing of several affected individuals from a large family affected with AD. PCR amplification and direct Sanger sequencing were used to verify variants detected by exome analysis and to screen family members at-risk to carry those variants. Neuropathologic brain evaluation by immunohistochemistry and MRI were performed for the carriers of the NOTCH3 variant. RESULTS In a three-generation family with AD, we found a c.601 T > C p.Cys201Arg variant in the NOTCH3 gene that caused clinical and neuropathological manifestations of CADASIL. These features included earlier onset of dementia accompanied by behavioral abnormalities in the father and son and white matter abnormalities in the asymptomatic grandson. The family is one branch of a large pedigree studied by the Alzheimer's Disease Sequencing Project (ADSP). As part of the ADSP linkage analysis and whole genome sequencing endeavor, an ABCA1 variant, p.Ala937Val, was previously found associated with AD in this pedigree. CONCLUSIONS Our findings, together with other reported pathogenic missense variants of the C201 codon in NOTCH3, support the role of cysteine 201 as a mutation hotspot for CADASIL and highlight the genetic complexity both clinically and pathologically of AD and related dementia.
Collapse
Affiliation(s)
- Olena Korvatska
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, USA.
| | - Stephanie A Bucks
- Department of Neurology, University of Washington, Seattle, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Rebecca A Yoda
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Amber Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Michael O Dorschner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA; Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, USA
| | - Debby Tsuang
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Medical Center, Seattle, USA
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, USA
| | - Wendy H Raskind
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, USA; Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Medical Center, Seattle, USA; Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Medical Center, Seattle, USA
| | - Thomas D Bird
- Department of Neurology, University of Washington, Seattle, USA; Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Medical Center, Seattle, USA
| |
Collapse
|
9
|
Alatrany AS, Khan W, Hussain A, Al-Jumeily D. Wide and deep learning based approaches for classification of Alzheimer's disease using genome-wide association studies. PLoS One 2023; 18:e0283712. [PMID: 37126509 PMCID: PMC10150974 DOI: 10.1371/journal.pone.0283712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/15/2023] [Indexed: 05/02/2023] Open
Abstract
The increasing incidence of Alzheimer's disease (AD) has been leading towards a significant growth in socioeconomic challenges. A reliable prediction of AD might be useful to mitigate or at-least slow down its progression for which, identification of the factors affecting the AD and its accurate diagnoses, are vital. In this study, we use Genome-Wide Association Studies (GWAS) dataset which comprises significant genetic markers of complex diseases. The original dataset contains large number of attributes (620901) for which we propose a hybrid feature selection approach based on association test, principal component analysis, and the Boruta algorithm, to identify the most promising predictors of AD. The selected features are then forwarded to a wide and deep neural network models to classify the AD cases and healthy controls. The experimental outcomes indicate that our approach outperformed the existing methods when evaluated on standard dataset, producing an accuracy and f1-score of 99%. The outcomes from this study are impactful particularly, the identified features comprising AD-associated genes and a reliable classification model that might be useful for other chronic diseases.
Collapse
Affiliation(s)
- Abbas Saad Alatrany
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
- University of Information Technology and Communications, Baghdad, Iraq
- Imam Ja’afar Al-Sadiq University, Baghdad, Iraq
| | - Wasiq Khan
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
| | - Abir Hussain
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
- Department of Electrical Engineering, University of Sharjah, Sharjah, UAE
| | - Dhiya Al-Jumeily
- School of Computer Science and Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
| | | |
Collapse
|
10
|
Minaya MA, Mahali S, Iyer AK, Eteleeb AM, Martinez R, Huang G, Budde J, Temple S, Nana AL, Seeley WW, Spina S, Grinberg LT, Harari O, Karch CM. Conserved gene signatures shared among MAPT mutations reveal defects in calcium signaling. Front Mol Biosci 2023; 10:1051494. [PMID: 36845551 PMCID: PMC9948093 DOI: 10.3389/fmolb.2023.1051494] [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: 09/22/2022] [Accepted: 01/13/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction: More than 50 mutations in the MAPT gene result in heterogeneous forms of frontotemporal lobar dementia with tau inclusions (FTLD-Tau). However, early pathogenic events that lead to disease and the degree to which they are common across MAPT mutations remain poorly understood. The goal of this study is to determine whether there is a common molecular signature of FTLD-Tau. Methods: We analyzed genes differentially expressed in induced pluripotent stem cell-derived neurons (iPSC-neurons) that represent the three major categories of MAPT mutations: splicing (IVS10 + 16), exon 10 (p.P301L), and C-terminal (p.R406W) compared with isogenic controls. The genes that were commonly differentially expressed in MAPT IVS10 + 16, p.P301L, and p.R406W neurons were enriched in trans-synaptic signaling, neuronal processes, and lysosomal function. Many of these pathways are sensitive to disruptions in calcium homeostasis. One gene, CALB1, was significantly reduced across the three MAPT mutant iPSC-neurons and in a mouse model of tau accumulation. We observed a significant reduction in calcium levels in MAPT mutant neurons compared with isogenic controls, pointing to a functional consequence of this disrupted gene expression. Finally, a subset of genes commonly differentially expressed across MAPT mutations were also dysregulated in brains from MAPT mutation carriers and to a lesser extent in brains from sporadic Alzheimer disease and progressive supranuclear palsy, suggesting that molecular signatures relevant to genetic and sporadic forms of tauopathy are captured in a dish. The results from this study demonstrate that iPSC-neurons capture molecular processes that occur in human brains and can be used to pinpoint common molecular pathways involving synaptic and lysosomal function and neuronal development, which may be regulated by disruptions in calcium homeostasis.
Collapse
Affiliation(s)
- Miguel A. Minaya
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
| | - Sidhartha Mahali
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
| | - Abhirami K. Iyer
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
| | - Abdallah M. Eteleeb
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
| | - Rita Martinez
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
| | - Guangming Huang
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
| | - John Budde
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY, United States
| | - Alissa L. Nana
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - William W. Seeley
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Salvatore Spina
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Lea T. Grinberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Department of Pathology, University of Sao Paulo, Sao Paulo, Brazil
| | - Oscar Harari
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, United States
- NeuroGenomics and Informatics Center, Washington University in St Louis, St Louis, MO, United States
| | - Celeste M. Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University in St Louis, St Louis, MO, United States
- NeuroGenomics and Informatics Center, Washington University in St Louis, St Louis, MO, United States
| |
Collapse
|
11
|
Dong W, Huang Y. Common Genetic Factors and Pathways in Alzheimer's Disease and Ischemic Stroke: Evidences from GWAS. Genes (Basel) 2023; 14:353. [PMID: 36833280 PMCID: PMC9957001 DOI: 10.3390/genes14020353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/19/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Alzheimer's disease (AD) and ischemic stroke (IS) are common neurological disorders, and the comorbidity of these two brain diseases is often seen. Although AD and IS were regarded as two distinct disease entities, in terms of different etiologies and clinical presentation, recent genome-wide association studies (GWASs) revealed that there were common risk genes between AD and IS, indicating common molecular pathways and their common pathophysiology. In this review, we summarize AD and IS risk single nucleotide polymorphisms (SNPs) and their representative genes from the GWAS Catalog database, and find thirteen common risk genes, but no common risk SNPs. Furthermore, the common molecular pathways associated with these risk gene products are summarized from the GeneCards database and clustered into inflammation and immunity, G protein-coupled receptor, and signal transduction. At least seven of these thirteen genes can be regulated by 23 microRNAs identified from the TargetScan database. Taken together, the imbalance of these molecular pathways may give rise to these two common brain disorders. This review sheds light on the pathogenesis of comorbidity of AD and IS, and provides molecular targets for disease prevention, manipulation, and brain health maintenance.
Collapse
Affiliation(s)
- Wei Dong
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yue Huang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Department of Pharmacology, School of Medical Sciences, Faculty of Medicine & Health, UNSW, Sydney, NSW 2052, Australia
| |
Collapse
|
12
|
Belloy ME, Le Guen Y, Eger SJ, Napolioni V, Greicius MD, He Z. A Fast and Robust Strategy to Remove Variant-Level Artifacts in Alzheimer Disease Sequencing Project Data. Neurol Genet 2022; 8:e200012. [PMID: 35966919 PMCID: PMC9372872 DOI: 10.1212/nxg.0000000000200012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/31/2022] [Indexed: 02/02/2023]
Abstract
Background and Objectives Exome sequencing (ES) and genome sequencing (GS) are expected to be critical to further elucidate the missing genetic heritability of Alzheimer disease (AD) risk by identifying rare coding and/or noncoding variants that contribute to AD pathogenesis. In the United States, the Alzheimer Disease Sequencing Project (ADSP) has taken a leading role in sequencing AD-related samples at scale, with the resultant data being made publicly available to researchers to generate new insights into the genetic etiology of AD. To achieve sufficient power, the ADSP has adapted a study design where subsets of larger AD cohorts are collected and sequenced across multiple centers, using a variety of sequencing platforms. This approach may lead to variable variant quality across sequencing centers and/or platforms. In this study, we sought to implement and evaluate filters that can be applied fast to robustly remove variant-level artifacts in the ADSP data. Methods We implemented a robust quality control procedure to handle ADSP data. We evaluated this procedure while performing exome-wide and genome-wide association analyses on AD risk using the latest ADSP whole ES (WES) and whole GS (WGS) data releases (NG00067.v5). Results We observed that many variants displayed large variation in allele frequencies across sequencing centers/platforms and contributed to spurious association signals with AD risk. We also observed that sequencing platform/center adjustment in association models could not fully account for these spurious signals. To address this issue, we designed and implemented variant filters that could capture and remove these center-specific/platform-specific artifactual variants. Discussion We derived a fast and robust approach to filter variants that represent sequencing center-related or platform-related artifacts underlying spurious associations with AD risk in ADSP WES and WGS data. This approach will be important to support future robust genetic association studies on ADSP data, as well as other studies with similar designs.
Collapse
Affiliation(s)
- Michael E. Belloy
- From the Department of Neurology and Neurological Sciences (M.E.B., Y.L.G., S.J.E., M.D.G., Z.H.), Stanford University, CA; Institut du Cerveau—Paris Brain Institute—ICM (Y.L.G.), France; School of Biosciences and Veterinary Medicine (V.N.), University of Camerino, Italy; and Quantitative Sciences Unit (Z.H.), Department of Medicine, Stanford University, CA
| | - Yann Le Guen
- From the Department of Neurology and Neurological Sciences (M.E.B., Y.L.G., S.J.E., M.D.G., Z.H.), Stanford University, CA; Institut du Cerveau—Paris Brain Institute—ICM (Y.L.G.), France; School of Biosciences and Veterinary Medicine (V.N.), University of Camerino, Italy; and Quantitative Sciences Unit (Z.H.), Department of Medicine, Stanford University, CA
| | - Sarah J. Eger
- From the Department of Neurology and Neurological Sciences (M.E.B., Y.L.G., S.J.E., M.D.G., Z.H.), Stanford University, CA; Institut du Cerveau—Paris Brain Institute—ICM (Y.L.G.), France; School of Biosciences and Veterinary Medicine (V.N.), University of Camerino, Italy; and Quantitative Sciences Unit (Z.H.), Department of Medicine, Stanford University, CA
| | - Valerio Napolioni
- From the Department of Neurology and Neurological Sciences (M.E.B., Y.L.G., S.J.E., M.D.G., Z.H.), Stanford University, CA; Institut du Cerveau—Paris Brain Institute—ICM (Y.L.G.), France; School of Biosciences and Veterinary Medicine (V.N.), University of Camerino, Italy; and Quantitative Sciences Unit (Z.H.), Department of Medicine, Stanford University, CA
| | - Michael D. Greicius
- From the Department of Neurology and Neurological Sciences (M.E.B., Y.L.G., S.J.E., M.D.G., Z.H.), Stanford University, CA; Institut du Cerveau—Paris Brain Institute—ICM (Y.L.G.), France; School of Biosciences and Veterinary Medicine (V.N.), University of Camerino, Italy; and Quantitative Sciences Unit (Z.H.), Department of Medicine, Stanford University, CA
| | - Zihuai He
- From the Department of Neurology and Neurological Sciences (M.E.B., Y.L.G., S.J.E., M.D.G., Z.H.), Stanford University, CA; Institut du Cerveau—Paris Brain Institute—ICM (Y.L.G.), France; School of Biosciences and Veterinary Medicine (V.N.), University of Camerino, Italy; and Quantitative Sciences Unit (Z.H.), Department of Medicine, Stanford University, CA
| |
Collapse
|
13
|
Bartoletti-Stella A, Tarozzi M, Mengozzi G, Asirelli F, Brancaleoni L, Mometto N, Stanzani-Maserati M, Baiardi S, Linarello S, Spallazzi M, Pantieri R, Ferriani E, Caffarra P, Liguori R, Parchi P, Capellari S. Dementia-related genetic variants in an Italian population of early-onset Alzheimer’s disease. Front Aging Neurosci 2022; 14:969817. [PMID: 36133075 PMCID: PMC9484406 DOI: 10.3389/fnagi.2022.969817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Early-onset Alzheimer’s disease (EOAD) is the most common form of early-onset dementia. Although three major genes have been identified as causative, the genetic contribution to the disease remains unsolved in many patients. Recent studies have identified pathogenic variants in genes representing a risk factor for developing Alzheimer’s disease (AD) and in causative genes for other degenerative dementias as responsible for EOAD. To study them further, we investigated a panel of candidate genes in 102 Italian EOAD patients, 45.10% of whom had a positive family history and 21.74% with a strong family history of dementia. We found that 10.78% of patients carried pathogenic or likely pathogenic variants, including a novel variant, in PSEN1, PSEN2, or APP, and 7.84% showed homozygosity for the ε4 APOE allele. Additionally, 7.84% of patients had a moderate risk allele in PSEN1, PSEN2, or TREM2 genes. Besides, we observed that 12.75% of our patients carried only a variant in genes associated with other neurodegenerative diseases. The combination of these variants contributes to explain 46% of cases with a definite familiarity and 32% of sporadic forms. Our results confirm the importance of extensive genetic screening in EOAD for clinical purposes, to select patients for future treatments and to contribute to the definition of overlapping pathogenic mechanisms between AD and other forms of dementia.
Collapse
Affiliation(s)
- Anna Bartoletti-Stella
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Martina Tarozzi
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Giacomo Mengozzi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Francesca Asirelli
- Department of Medical Science and Surgery (DIMEC), University of Bologna, Bologna, Italy
| | - Laura Brancaleoni
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Neurologia e Rete Stroke Metropolitana, Ospedale Maggiore, Bologna, Italy
| | - Nicola Mometto
- UOC Neurologia, Ospedale Guglielmo da Saliceto, Piacenza, Italy
| | | | - Simone Baiardi
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Simona Linarello
- Programma Cure Intermedie - Azienda USL di Bologna, Bologna, Italy
| | - Marco Spallazzi
- U.O. di Neurologia, Azienda Ospedaliero-Universitaria, Parma, Italy
| | - Roberta Pantieri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Elisa Ferriani
- UOC Psicologia Clinica Ospedaliera, Ospedale Bellaria, Azienda USL di Bologna, Bologna, Italy
| | - Paolo Caffarra
- Unità di Neuroscienze, Università di Parma, Parma, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
- *Correspondence: Sabina Capellari,
| |
Collapse
|
14
|
Sirkis DW, Bonham LW, Johnson TP, La Joie R, Yokoyama JS. Dissecting the clinical heterogeneity of early-onset Alzheimer's disease. Mol Psychiatry 2022; 27:2674-2688. [PMID: 35393555 PMCID: PMC9156414 DOI: 10.1038/s41380-022-01531-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) is a rare but particularly devastating form of AD. Though notable for its high degree of clinical heterogeneity, EOAD is defined by the same neuropathological hallmarks underlying the more common, late-onset form of AD. In this review, we describe the various clinical syndromes associated with EOAD, including the typical amnestic phenotype as well as atypical variants affecting visuospatial, language, executive, behavioral, and motor functions. We go on to highlight advances in fluid biomarker research and describe how molecular, structural, and functional neuroimaging can be used not only to improve EOAD diagnostic acumen but also enhance our understanding of fundamental pathobiological changes occurring years (and even decades) before the onset of symptoms. In addition, we discuss genetic variation underlying EOAD, including pathogenic variants responsible for the well-known mendelian forms of EOAD as well as variants that may increase risk for the much more common forms of EOAD that are either considered to be sporadic or lack a clear autosomal-dominant inheritance pattern. Intriguingly, specific pathogenic variants in PRNP and MAPT-genes which are more commonly associated with other neurodegenerative diseases-may provide unexpectedly important insights into the formation of AD tau pathology. Genetic analysis of the atypical clinical syndromes associated with EOAD will continue to be challenging given their rarity, but integration of fluid biomarker data, multimodal imaging, and various 'omics techniques and their application to the study of large, multicenter cohorts will enable future discoveries of fundamental mechanisms underlying the development of EOAD and its varied clinical presentations.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Taylor P Johnson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
15
|
Chen F, Wang N, He X. Identification of Differential Genes of DNA Methylation Associated With Alzheimer's Disease Based on Integrated Bioinformatics and Its Diagnostic Significance. Front Aging Neurosci 2022; 14:884367. [PMID: 35615586 PMCID: PMC9125150 DOI: 10.3389/fnagi.2022.884367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Background Alzheimer's disease (AD) is a common neurodegenerative disease. The pathogenesis is complex and has not been clearly elucidated, and there is no effective treatment. Recent studies have demonstrated that DNA methylation is closely associated with the pathogenesis of AD, which sheds light on investigating potential biomarkers for the diagnosis of early AD and related possible therapeutic approaches. Methods Alzheimer's disease patients samples and healthy controls samples were collected from two datasets in the GEO database. Using LIMMA software package in R language to find differentially expressed genes (DEGs). Afterward, DEGs have been subjected to enrichment analysis of GO and KEGG pathways. The PPI networks and Hub genes were created and visualized based on the STRING database and Cytoscape. ROC curves were further constructed to analyze the accuracy of these genes for AD diagnosis. Results Analysis of the GSE109887 and GSE97760 datasets showed 477 significant DEGs. GO and KEGG enrichment analysis showed terms related to biological processes related to these genes. The top ten Hub genes were found on the basis of the PPI network using the CytoHubba plugin, and the AUC areas of these top ranked genes were all greater than 0.7, showing satisfactory diagnostic accuracy. Conclusion The study identified the top 10 Hub genes associated with AD-related DNA methylation, of which RPSA, RPS23, and RPLP0 have high diagnostic accuracy and excellent AD biomarker potential.
Collapse
Affiliation(s)
| | | | - Xiaping He
- School of Basic Medical Sciences, Dali University, Dali, China
| |
Collapse
|
16
|
Tarozzi M, Bartoletti-Stella A, Dall'Olio D, Matteuzzi T, Baiardi S, Parchi P, Castellani G, Capellari S. Identification of recurrent genetic patterns from targeted sequencing panels with advanced data science: a case-study on sporadic and genetic neurodegenerative diseases. BMC Med Genomics 2022; 15:26. [PMID: 35144616 PMCID: PMC8830183 DOI: 10.1186/s12920-022-01173-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background Targeted Next Generation Sequencing is a common and powerful approach used in both clinical and research settings. However, at present, a large fraction of the acquired genetic information is not used since pathogenicity cannot be assessed for most variants. Further complicating this scenario is the increasingly frequent description of a poli/oligogenic pattern of inheritance showing the contribution of multiple variants in increasing disease risk. We present an approach in which the entire genetic information provided by target sequencing is transformed into binary data on which we performed statistical, machine learning, and network analyses to extract all valuable information from the entire genetic profile. To test this approach and unbiasedly explore the presence of recurrent genetic patterns, we studied a cohort of 112 patients affected either by genetic Creutzfeldt–Jakob (CJD) disease caused by two mutations in the PRNP gene (p.E200K and p.V210I) with different penetrance or by sporadic Alzheimer disease (sAD). Results Unsupervised methods can identify functionally relevant sources of variation in the data, like haplogroups and polymorphisms that do not follow Hardy–Weinberg equilibrium, such as the NOTCH3 rs11670823 (c.3837 + 21 T > A). Supervised classifiers can recognize clinical phenotypes with high accuracy based on the mutational profile of patients. In addition, we found a similar alteration of allele frequencies compared the European population in sporadic patients and in V210I-CJD, a poorly penetrant PRNP mutation, and sAD, suggesting shared oligogenic patterns in different types of dementia. Pathway enrichment and protein–protein interaction network revealed different altered pathways between the two PRNP mutations. Conclusions We propose this workflow as a possible approach to gain deeper insights into the genetic information derived from target sequencing, to identify recurrent genetic patterns and improve the understanding of complex diseases. This work could also represent a possible starting point of a predictive tool for personalized medicine and advanced diagnostic applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01173-4.
Collapse
Affiliation(s)
- M Tarozzi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - A Bartoletti-Stella
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - D Dall'Olio
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - T Matteuzzi
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - S Baiardi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - P Parchi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - G Castellani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - S Capellari
- IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
17
|
Evidence for an Inherited Contribution to Sepsis Susceptibility Among a Cohort of U.S. Veterans. Crit Care Explor 2022; 4:e0603. [PMID: 35036923 PMCID: PMC8754185 DOI: 10.1097/cce.0000000000000603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Analyze a unique clinical and genealogical resource for evidence of familial clustering of sepsis to test for an inherited contribution to sepsis predisposition.
Collapse
|
18
|
Khani M, Gibbons E, Bras J, Guerreiro R. Challenge accepted: uncovering the role of rare genetic variants in Alzheimer's disease. Mol Neurodegener 2022; 17:3. [PMID: 35000612 PMCID: PMC8744312 DOI: 10.1186/s13024-021-00505-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
The search for rare variants in Alzheimer's disease (AD) is usually deemed a high-risk - high-reward situation. The challenges associated with this endeavor are real. Still, the application of genome-wide technologies to large numbers of cases and controls or to small, well-characterized families has started to be fruitful.Rare variants associated with AD have been shown to increase risk or cause disease, but also to protect against the development of AD. All of these can potentially be targeted for the development of new drugs.Multiple independent studies have now shown associations of rare variants in NOTCH3, TREM2, SORL1, ABCA7, BIN1, CLU, NCK2, AKAP9, UNC5C, PLCG2, and ABI3 with AD and suggested that they may influence disease via multiple mechanisms. These genes have reported functions in the immune system, lipid metabolism, synaptic plasticity, and apoptosis. However, the main pathway emerging from the collective of genes harboring rare variants associated with AD is the Aβ pathway. Associations of rare variants in dozens of other genes have also been proposed, but have not yet been replicated in independent studies. Replication of this type of findings is one of the challenges associated with studying rare variants in complex diseases, such as AD. In this review, we discuss some of these primary challenges as well as possible solutions.Integrative approaches, the availability of large datasets and databases, and the development of new analytical methodologies will continue to produce new genes harboring rare variability impacting AD. In the future, more extensive and more diverse genetic studies, as well as studies of deeply characterized families, will enhance our understanding of disease pathogenesis and put us on the correct path for the development of successful drugs.
Collapse
Affiliation(s)
- Marzieh Khani
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Elizabeth Gibbons
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave. N.E., Grand Rapids, Michigan 49503-2518 USA
| | - Jose Bras
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave. N.E., Grand Rapids, Michigan 49503-2518 USA
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave. N.E., Grand Rapids, Michigan 49503-2518 USA
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI USA
| |
Collapse
|
19
|
Karlsson IK, Escott-Price V, Gatz M, Hardy J, Pedersen NL, Shoai M, Reynolds CA. Measuring heritable contributions to Alzheimer's disease: polygenic risk score analysis with twins. Brain Commun 2022; 4:fcab308. [PMID: 35169705 PMCID: PMC8833403 DOI: 10.1093/braincomms/fcab308] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/17/2021] [Accepted: 10/18/2021] [Indexed: 11/23/2022] Open
Abstract
The heritability of Alzheimer's disease estimated from twin studies is greater than the heritability derived from genome-based studies, for reasons that remain unclear. We apply both approaches to the same twin sample, considering both Alzheimer's disease polygenic risk scores and heritability from twin models, to provide insight into the role of measured genetic variants and to quantify uncaptured genetic risk. A population-based heritability and polygenic association study of Alzheimer's disease was conducted between 1986 and 2016 and is the first study to incorporate polygenic risk scores into biometrical twin models of Alzheimer's disease. The sample included 1586 twins drawn from the Swedish Twin Registry which were nested within 1137 twin pairs (449 complete pairs and 688 incomplete pairs) with clinically based diagnoses and registry follow-up (M age = 85.28, SD = 7.02; 44% male; 431 cases and 1155 controls). We report contributions of polygenic risk scores at P < 1 × 10-5, considering a full polygenic risk score (PRS), PRS without the APOE region (PRS.no.APOE) and PRS.no.APOE plus directly measured APOE alleles. Biometric twin models estimated the contribution of environmental influences and measured (PRS) and unmeasured genes to Alzheimer's disease risk. The full PRS and PRS.no.APOE contributed 10.1 and 2.4% to Alzheimer's disease risk, respectively. When APOE ɛ4 alleles were added to the model with the PRS.no.APOE, the total contribution was 11.4% to Alzheimer's disease risk, where APOE ɛ4 explained 9.3% and PRS.no.APOE dropped from 2.4 to 2.1%. The total genetic contribution to Alzheimer's disease risk, measured and unmeasured, was 71% while environmental influences unique to each twin accounted for 29% of the risk. The APOE region accounts for much of the measurable genetic contribution to Alzheimer's disease, with a smaller contribution from other measured polygenic influences. Importantly, substantial background genetic influences remain to be understood.
Collapse
Affiliation(s)
- Ida K. Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Aging Research Network—Jönköping (ARN-J), School of Health and 6 Welfare, Jönköping University, Jönköping, Sweden
| | - Valentina Escott-Price
- UK Dementia Research Institute at Cardiff, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Margaret Gatz
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK
- UCL Movement Disorders Centre, University College London, London, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Nancy L. Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Maryam Shoai
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Chandra A. Reynolds
- Department of Psychology, University of California—Riverside, Riverside, CA, USA
| |
Collapse
|
20
|
Yang SA, Salazar JL, Li-Kroeger D, Yamamoto S. Functional Studies of Genetic Variants Associated with Human Diseases in Notch Signaling-Related Genes Using Drosophila. Methods Mol Biol 2022; 2472:235-276. [PMID: 35674905 PMCID: PMC9396741 DOI: 10.1007/978-1-0716-2201-8_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Rare variants in the many genes related to Notch signaling cause diverse Mendelian diseases that affect myriad organ systems. In addition, genome- and exome-wide association studies have linked common and rare variants in Notch-related genes to common diseases and phenotypic traits. Moreover, somatic mutations in these genes have been observed in many types of cancer, some of which are classified as oncogenic and others as tumor suppressive. While functional characterization of some of these variants has been performed through experimental studies, the number of "variants of unknown significance" identified in patients with diverse conditions keeps increasing as high-throughput sequencing technologies become more commonly used in the clinic. Furthermore, as disease gene discovery efforts identify rare variants in human genes that have yet to be linked to a disease, the demand for functional characterization of variants in these "genes of unknown significance" continues to increase. In this chapter, we describe a workflow to functionally characterize a rare variant in a Notch signaling related gene that was found to be associated with late-onset Alzheimer's disease. This pipeline involves informatic analysis of the variant of interest using diverse human and model organism databases, followed by in vivo experiments in the fruit fly Drosophila melanogaster. The protocol described here can be used to study variants that affect amino acids that are not conserved between human and fly. By "humanizing" the almondex gene in Drosophila with mutant alleles and heterologous genomic rescue constructs, a missense variant in TM2D3 (TM2 Domain Containing 3) was shown to be functionally damaging. This, and similar approaches, greatly facilitate functional interpretations of genetic variants in the human genome and propel personalized medicine.
Collapse
Affiliation(s)
- Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Jose L Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - David Li-Kroeger
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA.
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
21
|
Xue D, Bush WS, Renton AE, Marcora EA, Bis JC, Kunkle BW, Boerwinkle E, DeStefano AL, Farrer L, Goate A, Mayeux R, Pericak‐Vance M, Schellenberg G, Seshadri S, Wijsman E, Haines JL, Blue EE. Large-scale sequencing studies expand the known genetic architecture of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12255. [PMID: 35005195 PMCID: PMC8720139 DOI: 10.1002/dad2.12255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Genes implicated by genome-wide association studies and family-based studies of Alzheimer's disease (AD) are largely discordant. We hypothesized that genes identified by sequencing studies like the Alzheimer's Disease Sequencing Project (ADSP) may bridge this gap and highlight shared biological mechanisms. METHODS We performed structured literature review of genes prioritized by ADSP studies, genes underlying familial dementias, and genes nominated by genome-wide association studies. Gene set enrichment analyses of each list identified enriched pathways. RESULTS The genes prioritized by the ADSP, familial dementia studies, and genome-wide association studies minimally overlapped. Each gene set identified dozens of enriched pathways, several of which were shared (e.g., regulation of amyloid beta clearance). DISCUSSION Alternative study designs provide unique insights into AD genetics. Shared pathways enriched by different genes highlight their relevance to AD pathogenesis, while the patterns of pathway enrichment unique to each gene set provide additional targets for functional studies.
Collapse
Affiliation(s)
- Diane Xue
- Institute for Public Health GeneticsUniversity of WashingtonSeattleWashingtonUSA
| | - William S. Bush
- Department of Population and Quantitative Health Sciences and Department of Genetics and Genome SciencesCase Western Reserve UniversityClevelandOhioUSA
- Cleveland Institute for Computational BiologyClevelandOhioUSA
| | - Alan E. Renton
- Department of Genetics and Genomic SciencesNash Family Department of Neuroscienceand Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Edoardo A. Marcora
- Department of Genetics and Genomic SciencesNash Family Department of Neuroscienceand Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Joshua C. Bis
- Cardiovascular Health Research UnitDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Brian W. Kunkle
- The John P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFloridaUSA
- Dr. John T Macdonald Foundation Department of Human GeneticsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | | | - Eric Boerwinkle
- Human Genome Sequencing CenterDepartment of Molecular and Human GeneticsBaylor College of MedicineHoustonTexasUSA
- School of Public HealthUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Anita L. DeStefano
- Department of BiostatisticsBoston UniversityBostonMassachusettsUSA
- Department of NeurologyBoston UniversityBostonMassachusettsUSA
| | - Lindsay Farrer
- Department of BiostatisticsBoston UniversityBostonMassachusettsUSA
- Department of NeurologyBoston UniversityBostonMassachusettsUSA
- Division of Biomedical GeneticsDepartment of MedicineDepartment of Epidemiologyand Department of OphthalmologyBoston UniversityBostonMassachusettsUSA
| | - Alison Goate
- Department of Genetics and Genomic SciencesNash Family Department of Neuroscienceand Ronald M. Loeb Center for Alzheimer's DiseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genetics and Genomics Sciences and Friedman Brain InstituteMount Sinai School of MedicineNew YorkNew YorkUSA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainGertrude H. Sergievsky CenterDepartment of NeurologyDepartment of Psychiatryand EpidemiologyColumbia UniversityNew YorkNew YorkUSA
| | - Margaret Pericak‐Vance
- The John P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFloridaUSA
- Dr. John T Macdonald Foundation Department of Human GeneticsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Gerard Schellenberg
- Penn Neurodegeneration Genomics CenterDepartment of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases and Department of NeurologyUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - Ellen Wijsman
- Institute for Public Health GeneticsUniversity of WashingtonSeattleWashingtonUSA
- Division of Medical GeneticsUniversity of WashingtonSeattleWashingtonUSA
- Department of BiostatisticsUniversity of WashingtonSeattleWashingtonUSA
| | - Jonathan L. Haines
- Department of Population and Quantitative Health Sciences and Department of Genetics and Genome SciencesCase Western Reserve UniversityClevelandOhioUSA
- Cleveland Institute for Computational BiologyClevelandOhioUSA
| | - Elizabeth E. Blue
- Institute for Public Health GeneticsUniversity of WashingtonSeattleWashingtonUSA
- Division of Medical GeneticsUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
22
|
Association of low-frequency and rare coding variants with information processing speed. Transl Psychiatry 2021; 11:613. [PMID: 34864818 PMCID: PMC8643353 DOI: 10.1038/s41398-021-01736-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/20/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
Measures of information processing speed vary between individuals and decline with age. Studies of aging twins suggest heritability may be as high as 67%. The Illumina HumanExome Bead Chip genotyping array was used to examine the association of rare coding variants with performance on the Digit-Symbol Substitution Test (DSST) in community-dwelling adults participating in the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium. DSST scores were available for 30,576 individuals of European ancestry from nine cohorts and for 5758 individuals of African ancestry from four cohorts who were older than 45 years and free of dementia and clinical stroke. Linear regression models adjusted for age and gender were used for analysis of single genetic variants, and the T5, T1, and T01 burden tests that aggregate the number of rare alleles by gene were also applied. Secondary analyses included further adjustment for education. Meta-analyses to combine cohort-specific results were carried out separately for each ancestry group. Variants in RNF19A reached the threshold for statistical significance (p = 2.01 × 10-6) using the T01 test in individuals of European descent. RNF19A belongs to the class of E3 ubiquitin ligases that confer substrate specificity when proteins are ubiquitinated and targeted for degradation through the 26S proteasome. Variants in SLC22A7 and OR51A7 were suggestively associated with DSST scores after adjustment for education for African-American participants and in the European cohorts, respectively. Further functional characterization of its substrates will be required to confirm the role of RNF19A in cognitive function.
Collapse
|
23
|
Xiao X, Guo L, Liao X, Zhou Y, Zhang W, Zhou L, Wang X, Liu X, Liu H, Xu T, Zhu Y, Yang Q, Hao X, Liu Y, Wang J, Li J, Jiao B, Shen L. The role of vascular dementia associated genes in patients with Alzheimer's disease: A large case-control study in the Chinese population. CNS Neurosci Ther 2021; 27:1531-1539. [PMID: 34551193 PMCID: PMC8611771 DOI: 10.1111/cns.13730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/01/2021] [Accepted: 09/05/2021] [Indexed: 12/16/2022] Open
Abstract
Aim The role of vascular dementia (VaD)‐associated genes in Alzheimer's disease (AD) remains elusive despite similar clinical and pathological features. We aimed to explore the relationship between these genes and AD in the Chinese population. Methods Eight VaD‐associated genes were screened by a targeted sequencing panel in a sample of 3604 individuals comprising 1192 AD patients and 2412 cognitively normal controls. Variants were categorized into common variants and rare variants according to minor allele frequency (MAF). Common variant (MAF ≥ 0.01)‐based association analysis was conducted by PLINK 1.9. Rare variant (MAF < 0.01) association study and gene‐based aggregation testing of rare variants were performed by PLINK 1.9 and Sequence Kernel Association Test‐Optimal (SKAT‐O test), respectively. Age at onset (AAO) and Mini‐Mental State Examination (MMSE) association studies were performed with PLINK 1.9. Analyses were adjusted for age, gender, and APOE ε4 status. Results Four common COL4A1 variants, including rs874203, rs874204, rs16975492, and rs1373744, exhibited suggestive associations with AD. Five rare variants, NOTCH3 rs201436750, COL4A1 rs747972545, COL4A1 rs201481886, CST3 rs765692764, and CST3 rs140837441, showed nominal association with AD risk. Gene‐based aggregation testing revealed that HTRA1 was nominally associated with AD. In the AAO and MMSE association studies, variants in GSN, ITM2B, and COL4A1 reached suggestive significance. Conclusion Common variants in COL4A1 and rare variants in HTRA1, NOTCH3, COL4A1, and CST3 may be implicated in AD pathogenesis. Besides, GSN, ITM2B, and COL4A1 are probably involved in the development of AD endophenotypes.
Collapse
Affiliation(s)
- Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lina Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Liao
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Yafang Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Weiwei Zhang
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xixi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Tianyan Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoli Hao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yingzi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Jinchen Li
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
24
|
Panegyres PK. The Clinical Spectrum of Young Onset Dementia Points to Its Stochastic Origins. J Alzheimers Dis Rep 2021; 5:663-679. [PMID: 34632303 PMCID: PMC8461730 DOI: 10.3233/adr-210309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Dementia is a major global health problem and the search for improved therapies is ongoing. The study of young onset dementia (YOD)-with onset prior to 65 years-represents a challenge owing to the variety of clinical presentations, pathology, and gene mutations. The advantage of the investigation of YOD is the lack of comorbidities that complicate the clinical picture in older adults. Here we explore the origins of YOD. OBJECTIVE To define the clinical diversity of YOD in terms of its demography, range of presentations, neurological examination findings, comorbidities, medical history, cognitive findings, imaging abnormalities both structural and functional, electroencephagraphic (EEG) data, neuropathology, and genetics. METHODS A prospective 20-year study of 240 community-based patients referred to specialty neurology clinics established to elucidate the nature of YOD. RESULTS Alzheimer's disease (AD; n = 139) and behavioral variant frontotemporal (bvFTD; n = 58) were the most common causes with a mean age of onset of 56.5 years for AD (±1 SD 5.45) and 57.1 years for bvFTD (±1 SD 5.66). Neuropathology showed a variety of diagnoses from multiple sclerosis, Lewy body disease, FTD-MND, TDP-43 proteinopathy, adult-onset leukoencephalopathy with axonal steroids and pigmented glia, corticobasal degeneration, unexplained small vessel disease, and autoimmune T-cell encephalitis. Non-amnestic forms of AD and alternative forms of FTD were discovered. Mutations were only found in 11 subjects (11/240 = 4.6%). APOE genotyping was not divergent between the two populations. CONCLUSION There are multiple kinds of YOD, and most are sporadic. These observations point to their stochastic origins.
Collapse
Affiliation(s)
- Peter K Panegyres
- Neurodegenerative Disorders Research Pty Ltd, West Perth, Australia
- The University of Western Australia, Nedlands, Australia
| |
Collapse
|
25
|
Balabanski L, Serbezov D, Atanasoska M, Karachanak-Yankova S, Hadjidekova S, Nikolova D, Boyanova O, Staneva R, Vazharova R, Mihailova M, Damyanova V, Nesheva D, Belejanska D, Mehrabian S, Traykov L, Toncheva D. Rare genetic variants prioritize molecular pathways for semaphorin interactions in Alzheimer’s disease patients. BIOTECHNOL BIOTEC EQ 2021. [DOI: 10.1080/13102818.2021.1964382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Lubomir Balabanski
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Malinov MD”, Sofia, Bulgaria
| | - Dimitar Serbezov
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Maya Atanasoska
- Genetic Laboratory, Gynecology and Assisted Reproduction Hospital “Malinov MD”, Sofia, Bulgaria
- Department of Genetics, Faculty of Biology, Sofia University St Kliment Ohridski, Sofia, Bulgaria
| | - Sena Karachanak-Yankova
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
- Department of Genetics, Faculty of Biology, Sofia University St Kliment Ohridski, Sofia, Bulgaria
| | - Savina Hadjidekova
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Dragomira Nikolova
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Olga Boyanova
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Rada Staneva
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Radoslava Vazharova
- Department of Biology, Medical Genetics and Microbiology, Faculty of Medicine, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Marta Mihailova
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Vera Damyanova
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Desislava Nesheva
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
| | - Diana Belejanska
- Depatment of Neurology, University Hospital “Alexandrovska”, Medical University of Sofia, Sofia, Bulgaria
| | - Shima Mehrabian
- Depatment of Neurology, University Hospital “Alexandrovska”, Medical University of Sofia, Sofia, Bulgaria
| | - Lachezar Traykov
- Depatment of Neurology, University Hospital “Alexandrovska”, Medical University of Sofia, Sofia, Bulgaria
| | - Draga Toncheva
- Department of Medical Genetics, Medical Faculty, Medical University of Sofia, Sofia, Bulgaria
- Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
26
|
Teerlink CC, Miller JB, Vance EL, Staley LA, Stevens J, Tavana JP, Cloward ME, Page ML, Dayton L, Cannon-Albright LA, Kauwe JSK. Analysis of high-risk pedigrees identifies 11 candidate variants for Alzheimer's disease. Alzheimers Dement 2021; 18:307-317. [PMID: 34151536 PMCID: PMC9291865 DOI: 10.1002/alz.12397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/15/2021] [Accepted: 05/11/2021] [Indexed: 11/08/2022]
Abstract
Introduction Analysis of sequence data in high‐risk pedigrees is a powerful approach to detect rare predisposition variants. Methods Rare, shared candidate predisposition variants were identified from exome sequencing 19 Alzheimer's disease (AD)‐affected cousin pairs selected from high‐risk pedigrees. Variants were further prioritized by risk association in various external datasets. Candidate variants emerging from these analyses were tested for co‐segregation to additional affected relatives of the original sequenced pedigree members. Results AD‐affected high‐risk cousin pairs contained 564 shared rare variants. Eleven variants spanning 10 genes were prioritized in external datasets: rs201665195 (ABCA7), and rs28933981 (TTR) were previously implicated in AD pathology; rs141402160 (NOTCH3) and rs140914494 (NOTCH3) were previously reported; rs200290640 (PIDD1) and rs199752248 (PIDD1) were present in more than one cousin pair; rs61729902 (SNAP91), rs140129800 (COX6A2, AC026471), and rs191804178 (MUC16) were not present in a longevity cohort; and rs148294193 (PELI3) and rs147599881 (FCHO1) approached significance from analysis of AD‐related phenotypes. Three variants were validated via evidence of co‐segregation to additional relatives (PELI3, ABCA7, and SNAP91). Discussion These analyses support ABCA7 and TTR as AD risk genes, expand on previously reported NOTCH3 variant identification, and prioritize seven additional candidate variants.
Collapse
Affiliation(s)
- Craig C Teerlink
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Justin B Miller
- Department of Biomedical Informatics, University of Kentucky Sanders-Brown Center on Aging, Lexington, Kentucky, USA
| | | | - Lyndsay A Staley
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | - Jeffrey Stevens
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Justina P Tavana
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | | | - Madeline L Page
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | - Louisa Dayton
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | | | - Lisa A Cannon-Albright
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.,George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA.,Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, Utah, USA
| |
Collapse
|
27
|
Monk B, Rajkovic A, Petrus S, Rajkovic A, Gaasterland T, Malinow R. A Machine Learning Method to Identify Genetic Variants Potentially Associated With Alzheimer's Disease. Front Genet 2021; 12:647436. [PMID: 34194466 PMCID: PMC8238203 DOI: 10.3389/fgene.2021.647436] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/06/2021] [Indexed: 01/17/2023] Open
Abstract
There is hope that genomic information will assist prediction, treatment, and understanding of Alzheimer's disease (AD). Here, using exome data from ∼10,000 individuals, we explore machine learning neural network (NN) methods to estimate the impact of SNPs (i.e., genetic variants) on AD risk. We develop an NN-based method (netSNP) that identifies hundreds of novel potentially protective or at-risk AD-associated SNPs (along with an effect measure); the majority with frequency under 0.01. For case individuals, the number of "protective" (or "at-risk") netSNP-identified SNPs in their genome correlates positively (or inversely) with their age of AD diagnosis and inversely (or positively) with autopsy neuropathology. The effect measure increases correlations. Simulations suggest our results are not due to genetic linkage, overfitting, or bias introduced by netSNP. These findings suggest that netSNP can identify SNPs associated with AD pathophysiology that may assist with the diagnosis and mechanistic understanding of the disease.
Collapse
Affiliation(s)
- Bradley Monk
- Department of Neurosciences, Center for Neural Circuits and Behavior, School of Medicine, University of California, San Diego, San Diego, CA, United States
- Cognitive Science & Psychology IDP, University of California, San Diego, San Diego, CA, United States
| | - Andrei Rajkovic
- Department of Computer Science, Royal Holloway, University of London, Egham, United Kingdom
| | - Semar Petrus
- Institute for Genomic Medicine, Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA, United States
| | - Aleks Rajkovic
- Department of Pathology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Terry Gaasterland
- Institute for Genomic Medicine, Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA, United States
| | - Roberto Malinow
- Department of Neurosciences, Center for Neural Circuits and Behavior, School of Medicine, University of California, San Diego, San Diego, CA, United States
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
28
|
Teerlink CC, Stevens J, Hernandez R, Facelli JC, Cannon-Albright LA. An intronic variant in the CELF4 gene is associated with risk for colorectal cancer. Cancer Epidemiol 2021; 72:101941. [PMID: 33930674 PMCID: PMC8158787 DOI: 10.1016/j.canep.2021.101941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/17/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Germline predisposition variants associated with colorectal cancer (CRC) have been identified but all are not yet identified. We sought to identify the responsible predisposition germline variant in an extended high-risk CRC pedigree that exhibited evidence of linkage to the 18q12.2 region (TLOD = +2.81). METHODS DNA from two distantly related carriers of the hypothesized predisposition haplotype on 18q12.2 was sequenced to identify candidate variants. The candidate rare variants shared by the related sequenced subjects were screened in 3,094 CRC cases and 5x population-matched controls from UKBiobank to test for association. Further segregation of the variant was tested via Taqman assay in other sampled individuals in the pedigree. RESULTS Analysis of whole genome sequence data for the two related hypothesized predisposition haplotype carriers, restricted to the shared haplotype boundaries, identified multiple (n = 6) rare candidate non-coding variants that were tested for association with CRC risk in UKBiobank. A rare intronic variant ofCELF4 gene, rs568643870, was significantly associated with CRC (p = 0.004, OR = 5.0), and segregated with CRC in other members of the linked pedigree. CONCLUSION Evidence of segregation in a high-risk pedigree, case-control association in an external dataset, and identification of additional CRC-affected carriers in the linked pedigree support a role for a rareCELF4 intronic variant in CRC risk.
Collapse
Affiliation(s)
- Craig C Teerlink
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - Jeff Stevens
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - Rolando Hernandez
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA.
| | - Julio C Facelli
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; Center for Clinical and Translational Science, University of Utah School of Medicine, Salt Lake City, UT 84108, USA.
| | - Lisa A Cannon-Albright
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA; Huntsman Cancer Institute, Salt Lake City, UT 84112, USA.
| |
Collapse
|
29
|
Lu H, Zhang J, Chen YE, Garcia-Barrio MT. Integration of Transformative Platforms for the Discovery of Causative Genes in Cardiovascular Diseases. Cardiovasc Drugs Ther 2021; 35:637-654. [PMID: 33856594 PMCID: PMC8216854 DOI: 10.1007/s10557-021-07175-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Genome-wide association studies (GWAS) are powerful epidemiological tools to find genes and variants associated with cardiovascular diseases while follow-up biological studies allow to better understand the etiology and mechanisms of disease and assign causality. Improved methodologies and reduced costs have allowed wider use of bulk and single-cell RNA sequencing, human-induced pluripotent stem cells, organoids, metabolomics, epigenomics, and novel animal models in conjunction with GWAS. In this review, we feature recent advancements relevant to cardiovascular diseases arising from the integration of genetic findings with multiple enabling technologies within multidisciplinary teams to highlight the solidifying transformative potential of this approach. Well-designed workflows integrating different platforms are greatly improving and accelerating the unraveling and understanding of complex disease processes while promoting an effective way to find better drug targets, improve drug design and repurposing, and provide insight towards a more personalized clinical practice.
Collapse
Affiliation(s)
- Haocheng Lu
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Y Eugene Chen
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA.
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| | - Minerva T Garcia-Barrio
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA.
| |
Collapse
|
30
|
Elahi FM, Ashimatey SB, Bennett DJ, Walters SM, La Joie R, Jiang X, Wolf A, Cobigo Y, Staffaroni AM, Rosen HJ, Miller BL, Rabinovici GD, Kramer JH, Green AJ, Kashani AH. Retinal imaging demonstrates reduced capillary density in clinically unimpaired APOE ε4 gene carriers. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12181. [PMID: 34013017 PMCID: PMC8111703 DOI: 10.1002/dad2.12181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Apolipoprotein E (APOE) ε4, the strongest non-Mendelian genetic risk factor for Alzheimer's disease (AD), has been shown to affect brain capillaries in mice, with potential implications for AD-related neurodegenerative disease. However, human brain capillaries cannot be directly visualized in vivo. We therefore used retinal imaging to test APOE ε4 effects on human central nervous system capillaries. METHODS We collected retinal optical coherence tomography angiography, cognitive testing, and brain imaging in research participants and built statistical models to test genotype-phenotype associations. RESULTS Our analyses demonstrate lower retinal capillary densities in early disease, in cognitively normal APOE ε4 gene carriers. Furthermore, through regression modeling with a measure of brain perfusion (arterial spin labeling), we provide support for the relevance of these findings to cerebral vasculature. DISCUSSION These results suggest that APOE ε4 affects capillary health in humans and that retinal capillary measures could serve as surrogates for brain capillaries, providing an opportunity to study microangiopathic contributions to neurodegenerative disorders directly in humans.
Collapse
Affiliation(s)
- Fanny M. Elahi
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
- San Francisco Veterans Affairs Health Care SystemSan FranciscoCaliforniaUSA
| | - Senyo B. Ashimatey
- Department of OphthalmologyUSC Roski Eye InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Daniel J. Bennett
- Department of NeurologyDivision of Neuroimmunology and Glial BiologyWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Samantha M. Walters
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Renaud La Joie
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Xuejuan Jiang
- Department of OphthalmologyUSC Roski Eye InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Amy Wolf
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Yann Cobigo
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Adam M. Staffaroni
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Howie J. Rosen
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Bruce L. Miller
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Gil D. Rabinovici
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Joel H. Kramer
- Department of NeurologyMemory and Aging CenterWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ari J. Green
- Department of NeurologyDivision of Neuroimmunology and Glial BiologyWeill Institute for NeurosciencesSan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of OphthalmologySan FranciscoUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Amir H. Kashani
- Department of OphthalmologyUSC Roski Eye InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
- USC Ginsberg Institute for Biomedical TherapeuticsLos AngelesCaliforniaUSA
| |
Collapse
|
31
|
Guo L, Jiao B, Liao X, Xiao X, Zhang W, Yuan Z, Liu X, Zhou L, Wang X, Zhu Y, Yang Q, Wang J, Tang B, Shen L. The role of NOTCH3 variants in Alzheimer's disease and subcortical vascular dementia in the Chinese population. CNS Neurosci Ther 2021; 27:930-940. [PMID: 33942994 PMCID: PMC8265940 DOI: 10.1111/cns.13647] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 01/05/2023] Open
Abstract
AIMS NOTCH3 gene mutations predominantly cause cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, a common etiology of subcortical vascular dementia (SVaD). Besides, there may be a pathogenic link between NOTCH3 variants and Alzheimer's disease (AD). We aimed to study the role of NOTCH3 variants in AD and SVaD patients. METHODS We recruited 763 patients with dementia (667 AD and 96 SVaD) and 365 healthy controls from the Southern Han Chinese population. Targeted capture sequencing was performed on NOTCH3 coding and adjacent intron regions to detect the pathogenic variants in AD and SVaD. The relationship between common or rare NOTCH3 variants and AD was further analyzed using Plink1.9. RESULTS Five known pathogenic variants (p.R182C, p.C201S, p.R544C, p.R607C, and p.R1006C) and two novel likely pathogenic variants (p.C201F and p.C1061F) were detected in 16 SVaD patients. Additionally, no pathogenic or likely pathogenic variants were found in AD patients. NOTCH3 was not associated with AD in either single-variant association analysis or gene-based association analysis. CONCLUSION Our findings broaden the mutational spectrum of NOTCH3 and validate the pathogenic role of NOTCH3 mutations in SVaD, but do not support the notion that NOTCH3 variation influences the risk of AD.
Collapse
Affiliation(s)
- Lina Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Xinxin Liao
- Department of Geriatrics Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Weiwei Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenhua Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xixi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
32
|
iPSCs: A Preclinical Drug Research Tool for Neurological Disorders. Int J Mol Sci 2021; 22:ijms22094596. [PMID: 33925625 PMCID: PMC8123805 DOI: 10.3390/ijms22094596] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
The development and commercialization of new drugs is an articulated, lengthy, and very expensive process that proceeds through several steps, starting from target identification, screening new leading compounds for testing in preclinical studies, and subsequently in clinical trials to reach the final approval for therapeutic use. Preclinical studies are usually performed using both cell cultures and animal models, although they do not completely resume the complexity of human diseases, in particular neurodegenerative conditions. To this regard, stem cells represent a powerful tool in all steps of drug discovery. The recent advancement in induced Pluripotent Stem Cells (iPSCs) technology has opened the possibility to obtain patient-specific disease models for drug screening and development. Here, we report the use of iPSCs as a disease model for drug development in the contest of neurological disorders, including Alzheimer’s (AD) and Parkinson’s disease (PD), Amyotrophic lateral Sclerosis (ALS), and Fragile X syndrome (FRAX).
Collapse
|
33
|
Dehghani N, Bras J, Guerreiro R. How understudied populations have contributed to our understanding of Alzheimer's disease genetics. Brain 2021; 144:1067-1081. [PMID: 33889936 DOI: 10.1093/brain/awab028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/30/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
The majority of genome-wide association studies have been conducted using samples with a broadly European genetic background. As a field, we acknowledge this limitation and the need to increase the diversity of populations studied. A major challenge when designing and conducting such studies is to assimilate large samples sizes so that we attain enough statistical power to detect variants associated with disease, particularly when trying to identify variants with low and rare minor allele frequencies. In this review, we aimed to illustrate the benefits to genetic characterization of Alzheimer's disease, in researching currently understudied populations. This is important for both fair representation of world populations and the translatability of findings. To that end, we conducted a literature search to understand the contributions of studies, on different populations, to Alzheimer's disease genetics. Using both PubMed and Alzforum Mutation Database, we systematically quantified the number of studies reporting variants in known disease-causing genes, in a worldwide manner, and discuss the contributions of research in understudied populations to the identification of novel genetic factors in this disease. Additionally, we compared the effects of genome-wide significant single nucleotide polymorphisms across populations by focusing on loci that show different association profiles between populations (a key example being APOE). Reports of variants in APP, PSEN1 and PSEN2 can initially determine whether patients from a country have been studied for Alzheimer's disease genetics. Most genome-wide significant associations in non-Hispanic white genome-wide association studies do not reach genome-wide significance in such studies of other populations, with some suggesting an opposite effect direction; this is likely due to much smaller sample sizes attained. There are, however, genome-wide significant associations first identified in understudied populations which have yet to be replicated. Familial studies in understudied populations have identified rare, high effect variants, which have been replicated in other populations. This work functions to both highlight how understudied populations have furthered our understanding of Alzheimer's disease genetics, and to help us gauge our progress in understanding the genetic architecture of this disease in all populations.
Collapse
Affiliation(s)
- Nadia Dehghani
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Jose Bras
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA.,Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, Michigan, USA.,Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| |
Collapse
|
34
|
Le Guen Y, Belloy ME, Napolioni V, Eger SJ, Kennedy G, Tao R, He Z, Greicius MD. A novel age-informed approach for genetic association analysis in Alzheimer's disease. Alzheimers Res Ther 2021; 13:72. [PMID: 33794991 PMCID: PMC8017764 DOI: 10.1186/s13195-021-00808-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/11/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Many Alzheimer's disease (AD) genetic association studies disregard age or incorrectly account for it, hampering variant discovery. METHODS Using simulated data, we compared the statistical power of several models: logistic regression on AD diagnosis adjusted and not adjusted for age; linear regression on a score integrating case-control status and age; and multivariate Cox regression on age-at-onset. We applied these models to real exome-wide data of 11,127 sequenced individuals (54% cases) and replicated suggestive associations in 21,631 genotype-imputed individuals (51% cases). RESULTS Modeling variable AD risk across age results in 5-10% statistical power gain compared to logistic regression without age adjustment, while incorrect age adjustment leads to critical power loss. Applying our novel AD-age score and/or Cox regression, we discovered and replicated novel variants associated with AD on KIF21B, USH2A, RAB10, RIN3, and TAOK2 genes. CONCLUSION Our AD-age score provides a simple means for statistical power gain and is recommended for future AD studies.
Collapse
Affiliation(s)
- Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA.
| | - Michael E Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| | - Valerio Napolioni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Sarah J Eger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| | - Gabriel Kennedy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| | - Ran Tao
- Department of Biostatistics and Vanderbilt Genetic Institute, Vanderbilt University, Nashville, TN, 37203, USA
| | - Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94304, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94304, USA
| |
Collapse
|
35
|
Patel D, Zhang X, Farrell JJ, Lunetta KL, Farrer LA. Set-Based Rare Variant Expression Quantitative Trait Loci in Blood and Brain from Alzheimer Disease Study Participants. Genes (Basel) 2021; 12:419. [PMID: 33804025 PMCID: PMC7999141 DOI: 10.3390/genes12030419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Because studies of rare variant effects on gene expression have limited power, we investigated set-based methods to identify rare expression quantitative trait loci (eQTL) related to Alzheimer disease (AD). Gene-level and pathway-level cis rare-eQTL mapping was performed genome-wide using gene expression data derived from blood donated by 713 Alzheimer's Disease Neuroimaging Initiative participants and from brain tissues donated by 475 Religious Orders Study/Memory and Aging Project participants. The association of gene or pathway expression with a set of all cis potentially regulatory low-frequency and rare variants within 1 Mb of genes was evaluated using SKAT-O. A total of 65 genes expressed in the brain were significant targets for rare expression single nucleotide polymorphisms (eSNPs) among which 17% (11/65) included established AD genes HLA-DRB1 and HLA-DRB5. In the blood, 307 genes were significant targets for rare eSNPs. In the blood and the brain, GNMT, LDHC, RBPMS2, DUS2, and HP were targets for significant eSNPs. Pathway enrichment analysis revealed significant pathways in the brain (n = 9) and blood (n = 16). Pathways for apoptosis signaling, cholecystokinin receptor (CCKR) signaling, and inflammation mediated by chemokine and cytokine signaling were common to both tissues. Significant rare eQTLs in inflammation pathways included five genes in the blood (ALOX5AP, CXCR2, FPR2, GRB2, IFNAR1) that were previously linked to AD. This study identified several significant gene- and pathway-level rare eQTLs, which further confirmed the importance of the immune system and inflammation in AD and highlighted the advantages of using a set-based eQTL approach for evaluating the effect of low-frequency and rare variants on gene expression.
Collapse
Affiliation(s)
- Devanshi Patel
- Bioinformatics Graduate Program, Boston University, Boston, MA 02215, USA;
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA; (X.Z.); (J.J.F.)
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA; (X.Z.); (J.J.F.)
| | - John J. Farrell
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA; (X.Z.); (J.J.F.)
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA;
| | - Lindsay A. Farrer
- Bioinformatics Graduate Program, Boston University, Boston, MA 02215, USA;
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA; (X.Z.); (J.J.F.)
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA;
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| |
Collapse
|
36
|
Lin A, Peiris NJ, Dhaliwal H, Hakim M, Li W, Ganesh S, Ramaswamy Y, Patel S, Misra A. Mural Cells: Potential Therapeutic Targets to Bridge Cardiovascular Disease and Neurodegeneration. Cells 2021; 10:cells10030593. [PMID: 33800271 PMCID: PMC7999039 DOI: 10.3390/cells10030593] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Mural cells collectively refer to the smooth muscle cells and pericytes of the vasculature. This heterogenous population of cells play a crucial role in the regulation of blood pressure, distribution, and the structural integrity of the vascular wall. As such, dysfunction of mural cells can lead to the pathogenesis and progression of a number of diseases pertaining to the vascular system. Cardiovascular diseases, particularly atherosclerosis, are perhaps the most well-described mural cell-centric case. For instance, atherosclerotic plaques are most often described as being composed of a proliferative smooth muscle cap accompanied by a necrotic core. More recently, the role of dysfunctional mural cells in neurodegenerative diseases, such as Alzheimer’s and Parkinson’s disease, is being recognized. In this review, we begin with an exploration of the mechanisms underlying atherosclerosis and neurodegenerative diseases, such as mural cell plasticity. Next, we highlight a selection of signaling pathways (PDGF, Notch and inflammatory signaling) that are conserved across both diseases. We propose that conserved mural cell signaling mechanisms can be exploited for the identification or development of dual-pronged therapeutics that impart both cardio- and neuroprotective qualities.
Collapse
MESH Headings
- Alzheimer Disease/drug therapy
- Alzheimer Disease/genetics
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Animals
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cardiotonic Agents/pharmacology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neuroprotective Agents/pharmacology
- Parkinson Disease/drug therapy
- Parkinson Disease/genetics
- Parkinson Disease/metabolism
- Parkinson Disease/pathology
- Pericytes/drug effects
- Pericytes/metabolism
- Pericytes/pathology
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Platelet-Derived Growth Factor/genetics
- Platelet-Derived Growth Factor/metabolism
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Alexander Lin
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Niridu Jude Peiris
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Harkirat Dhaliwal
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Maria Hakim
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Weizhen Li
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India;
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
- Cardiac Catheterization Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: ; Tel.: +61-18-0065-1373
| |
Collapse
|
37
|
Hoogmartens J, Cacace R, Van Broeckhoven C. Insight into the genetic etiology of Alzheimer's disease: A comprehensive review of the role of rare variants. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12155. [PMID: 33665345 PMCID: PMC7896636 DOI: 10.1002/dad2.12155] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) is generally known as a dominant disease due to highly penetrant pathogenic mutations in the amyloid precursor protein, presenilin 1 and 2. However, they explain only a fraction of EOAD patients (5% to 10%). Furthermore, only 10% to 15% of EOAD families present with clear autosomal dominant inheritance. Studies showed that only 35% to 60% of EOAD patients have at least one affected first-degree relative. Parent-offspring concordance in EOAD was estimated to be <10%, indicating that full penetrant dominant alleles are not the sole players in EOAD. We aim to summarize current knowledge of rare variants underlying familial and seemingly sporadic Alzheimer's disease (AD) patients. Genetic findings indicate that in addition to the amyloid beta pathway, other pathways are of importance in AD pathophysiology. We discuss the difficulties in interpreting the influence of rare variants on disease onset and we underline the value of carefully selected ethnicity-matched cohorts in AD genetic research.
Collapse
Affiliation(s)
- Julie Hoogmartens
- Neurodegenerative Brain DiseasesVIB Center for Molecular NeurologyAntwerpBelgium
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Rita Cacace
- Neurodegenerative Brain DiseasesVIB Center for Molecular NeurologyAntwerpBelgium
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain DiseasesVIB Center for Molecular NeurologyAntwerpBelgium
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
38
|
Mishra R, Li B. The Application of Artificial Intelligence in the Genetic Study of Alzheimer's Disease. Aging Dis 2020; 11:1567-1584. [PMID: 33269107 PMCID: PMC7673858 DOI: 10.14336/ad.2020.0312] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease in which genetic factors contribute approximately 70% of etiological effects. Studies have found many significant genetic and environmental factors, but the pathogenesis of AD is still unclear. With the application of microarray and next-generation sequencing technologies, research using genetic data has shown explosive growth. In addition to conventional statistical methods for the processing of these data, artificial intelligence (AI) technology shows obvious advantages in analyzing such complex projects. This article first briefly reviews the application of AI technology in medicine and the current status of genetic research in AD. Then, a comprehensive review is focused on the application of AI in the genetic research of AD, including the diagnosis and prognosis of AD based on genetic data, the analysis of genetic variation, gene expression profile, gene-gene interaction in AD, and genetic analysis of AD based on a knowledge base. Although many studies have yielded some meaningful results, they are still in a preliminary stage. The main shortcomings include the limitations of the databases, failing to take advantage of AI to conduct a systematic biology analysis of multilevel databases, and lack of a theoretical framework for the analysis results. Finally, we outlook the direction of future development. It is crucial to develop high quality, comprehensive, large sample size, data sharing resources; a multi-level system biology AI analysis strategy is one of the development directions, and computational creativity may play a role in theory model building, verification, and designing new intervention protocols for AD.
Collapse
Affiliation(s)
- Rohan Mishra
- Washington Institute for Health Sciences, Arlington, VA 22203, USA
| | - Bin Li
- Washington Institute for Health Sciences, Arlington, VA 22203, USA
- Georgetown University Medical Center, Washington D.C. 20057, USA
| |
Collapse
|
39
|
Afshar H, Adelirad F, Kowsari A, Kalhor N, Delbari A, Najafipour R, Foroughan M, Bozorgmehr A, Khamse S, Nazaripanah N, Ohadi M. Natural Selection at the NHLH2 Core Promoter Exceptionally Long CA-Repeat in Human and Disease-Only Genotypes in Late-Onset Neurocognitive Disorder. Gerontology 2020; 66:514-522. [PMID: 32877896 DOI: 10.1159/000509471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/17/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Approximately 2% of the human core promoter short tandem repeats (STRs) reach lengths of ≥6 repeats, which may in part be a result of adaptive evolutionary processes and natural selection. A single-exon transcript of the human nescient helix loop helix 2 (NHLH2) gene is flanked by the longest CA-repeat detected in a human protein-coding gene core promoter (Ensembl transcript ID: ENST00000369506.1). NHLH2 is involved in several biological and pathological pathways, such as motivated exercise, obesity, and diabetes. METHODS The allele and genotype distribution of the NHLH2 CA-repeat were investigated by sequencing in 655 Iranian subjects, consisting of late-onset neurocognitive disorder (NCD) as a clinical entity (n = 290) and matched controls (n = 365). The evolutionary trend of the CA-repeat was also studied across vertebrates. RESULTS The allele range was between 9 and 25 repeats in the NCD cases, and 12 and 24 repeats in the controls. At the frequency of 0.56, the 21-repeat allele was the predominant allele in the controls. While the 21-repeat was also the predominant allele in the NCD patients, we detected significant decline of the frequency (p < 0.0001) and homozygosity (p < 0.006) of this allele in this group. Furthermore, 12 genotypes were detected across 16 patients (5.5% of the entire NCD sample) and not in the controls (disease-only genotypes; p < 0.0003), consisting of at least one extreme allele. The extreme alleles were at 9, 12, 13, 18, and 19 repeats (extreme short end), and 23, 24, and 25 repeats (extreme long end), and their frequencies ranged between 0.001 and 0.04. The frequency of the 21-repeat allele significantly dropped to 0.09 in the disease-only genotype compartment (p < 0.0001). Evolutionarily, while the maximum length of the NHLH2 CA-repeat was 11 repeats in non-primates, this CA-repeat was ≥14 repeats in primates and reached maximum length in human. CONCLUSION We propose a novel locus for late-onset NCD at the NHLH2 core promoter exceptionally long CA-STR and natural selection at this locus. Furthermore, there was indication of genotypes at this locus that unambiguously linked to late-onset NCD. This is the first instance of natural selection in favor of a predominantly abundant STR allele in human and its differential distribution in late-onset NCD.
Collapse
Affiliation(s)
- Hossein Afshar
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Fatemeh Adelirad
- Department of Health Education and Promotion, Faculty of Health Sciences Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Kowsari
- Department of Mesenchymal Stem Cell, The Academic Center for Education, Culture and Research, Qom, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cell, The Academic Center for Education, Culture and Research, Qom, Iran
| | - Ahmad Delbari
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Reza Najafipour
- Cellular and Molecular Research Centre, Research Institute for Prevention of Non Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mahshid Foroughan
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Ali Bozorgmehr
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Safoura Khamse
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Neda Nazaripanah
- Department of Health Education and Promotion, Faculty of Health Sciences Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Ohadi
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran,
| |
Collapse
|
40
|
Errors in Results and Supplement. JAMA Netw Open 2020; 3:e2023811. [PMID: 32965496 PMCID: PMC7512054 DOI: 10.1001/jamanetworkopen.2020.23811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
41
|
Miller JB, Ward E, Staley LA, Stevens J, Teerlink CC, Tavana JP, Cloward M, Page M, Dayton L, Cannon-Albright LA, Kauwe JSK. Identification and genomic analysis of pedigrees with exceptional longevity identifies candidate rare variants. Neurobiol Dis 2020; 143:104972. [PMID: 32574725 PMCID: PMC7461696 DOI: 10.1016/j.nbd.2020.104972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/05/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Longevity as a phenotype entails living longer than average and typically includes living without chronic age-related diseases. Recently, several common genetic components to longevity have been identified. This study aims to identify additional genetic variants associated with longevity using unique and powerful analyses of pedigrees with a statistical excess of healthy elderly individuals identified in the Utah Population Database (UPDB). METHODS From an existing biorepository of Utah pedigrees, six independent cousin pairs were selected from four extended pedigrees that exhibited an excess of healthy elderly individuals; whole exome sequencing (WES) was performed on two elderly individuals from each pedigree who were either first cousins or first cousins once removed. Rare (<.01 population frequency) variants shared by at least one elderly cousin pair in a region likely to be identical by descent were identified as candidates. Ingenuity Variant Analysis was used to prioritize putative causal variants based on quality control, frequency, and gain or loss of function. The variant frequency was compared in healthy cohorts and in an Alzheimer's disease cohort. Remaining variants were filtered based on their presence in genes reported to have an effect on the aging process, aging of cells, or the longevity process. Validation of these candidate variants included tests of segregation on other elderly relatives. RESULTS Fifteen rare candidate genetic variants spanning 17 genes shared within cousins were identified as having passed prioritization criteria. Of those variants, six were present in genes that are known or predicted to affect the aging process: rs78408340 (PAM), rs112892337 (ZFAT), rs61737629 (ESPL1), rs141903485 (CEBPE), rs144369314 (UTP4), and rs61753103 (NUP88 and RABEP1). ESPL1 rs61737629 and CEBPE rs141903485 show additional evidence of segregation with longevity in expanded pedigree analyses (p-values = .001 and .0001, respectively). DISCUSSION This unique pedigree analysis efficiently identified several novel rare candidate variants that may affect the aging process and added support to seven genes that likely contribute to longevity. Further analyses showed evidence for segregation for two rare variants, ESPL1 rs61737629 and CEBPE rs141903485, in the original longevity pedigrees in which they were initially observed. These candidate genes and variants warrant further investigation.
Collapse
Affiliation(s)
- Justin B Miller
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Elizabeth Ward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lyndsay A Staley
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Jeffrey Stevens
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Craig C Teerlink
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Justina P Tavana
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Matthew Cloward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Madeline Page
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Louisa Dayton
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
42
|
Baggiani M, Dell’Anno MT, Pistello M, Conti L, Onorati M. Human Neural Stem Cell Systems to Explore Pathogen-Related Neurodevelopmental and Neurodegenerative Disorders. Cells 2020; 9:E1893. [PMID: 32806773 PMCID: PMC7464299 DOI: 10.3390/cells9081893] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/07/2020] [Accepted: 08/09/2020] [Indexed: 12/18/2022] Open
Abstract
Building and functioning of the human brain requires the precise orchestration and execution of myriad molecular and cellular processes, across a multitude of cell types and over an extended period of time. Dysregulation of these processes affects structure and function of the brain and can lead to neurodevelopmental, neurological, or psychiatric disorders. Multiple environmental stimuli affect neural stem cells (NSCs) at several levels, thus impairing the normal human neurodevelopmental program. In this review article, we will delineate the main mechanisms of infection adopted by several neurotropic pathogens, and the selective NSC vulnerability. In particular, TORCH agents, i.e., Toxoplasma gondii, others (including Zika virus and Coxsackie virus), Rubella virus, Cytomegalovirus, and Herpes simplex virus, will be considered for their devastating effects on NSC self-renewal with the consequent neural progenitor depletion, the cellular substrate of microcephaly. Moreover, new evidence suggests that some of these agents may also affect the NSC progeny, producing long-term effects in the neuronal lineage. This is evident in the paradigmatic example of the neurodegeneration occurring in Alzheimer's disease.
Collapse
Affiliation(s)
- Matteo Baggiani
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56126 Pisa, Italy;
| | - Maria Teresa Dell’Anno
- Cellular Engineering Laboratory, Fondazione Pisana per la Scienza ONLUS, 56017 Pisa, Italy;
| | - Mauro Pistello
- Retrovirus Center and Virology Section, Department of Translational Research, University of Pisa and Virology Division, Pisa University Hospital, 56100 Pisa, Italy;
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology—CIBIO, University of Trento, 38122 Trento, Italy;
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, 56126 Pisa, Italy;
| |
Collapse
|
43
|
Katsumata Y, Fardo DW, Bachstetter AD, Artiushin SC, Wang WX, Wei A, Brzezinski LJ, Nelson BG, Huang Q, Abner EL, Anderson S, Patel I, Shaw BC, Price DA, Niedowicz DM, Wilcock DW, Jicha GA, Neltner JH, Van Eldik LJ, Estus S, Nelson PT. Alzheimer Disease Pathology-Associated Polymorphism in a Complex Variable Number of Tandem Repeat Region Within the MUC6 Gene, Near the AP2A2 Gene. J Neuropathol Exp Neurol 2020; 79:3-21. [PMID: 31748784 PMCID: PMC8204704 DOI: 10.1093/jnen/nlz116] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/18/2019] [Accepted: 10/27/2019] [Indexed: 02/06/2023] Open
Abstract
We found evidence of late-onset Alzheimer disease (LOAD)-associated genetic polymorphism within an exon of Mucin 6 (MUC6) and immediately downstream from another gene: Adaptor Related Protein Complex 2 Subunit Alpha 2 (AP2A2). PCR analyses on genomic DNA samples confirmed that the size of the MUC6 variable number tandem repeat (VNTR) region was highly polymorphic. In a cohort of autopsied subjects with quantitative digital pathology data (n = 119), the size of the polymorphic region was associated with the severity of pTau pathology in neocortex. In a separate replication cohort of autopsied subjects (n = 173), more pTau pathology was again observed in subjects with longer VNTR regions (p = 0.031). Unlike MUC6, AP2A2 is highly expressed in human brain. AP2A2 expression was lower in a subset analysis of brain samples from persons with longer versus shorter VNTR regions (p = 0.014 normalizing with AP2B1 expression). Double-label immunofluorescence studies showed that AP2A2 protein often colocalized with neurofibrillary tangles in LOAD but was not colocalized with pTau proteinopathy in progressive supranuclear palsy, or with TDP-43 proteinopathy. In summary, polymorphism in a repeat-rich region near AP2A2 was associated with neocortical pTau proteinopathy (because of the unique repeats, prior genome-wide association studies were probably unable to detect this association), and AP2A2 was often colocalized with neurofibrillary tangles in LOAD.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - David W Fardo
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Sergey C Artiushin
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Wang-Xia Wang
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Angela Wei
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Lena J Brzezinski
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Bela G Nelson
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Qingwei Huang
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Erin L Abner
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Sonya Anderson
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Indumati Patel
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Benjamin C Shaw
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Douglas A Price
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Dana M Niedowicz
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Donna W Wilcock
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Gregory A Jicha
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Janna H Neltner
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Steven Estus
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| | - Peter T Nelson
- Sanders-Brown Center on Aging (YK, DWF, ADB, SCA, W-XW, AW, LJB, BGN, QH, ELA, SA, IP, DAP, DMN, DWW, GAJ, LJVE, PTN); Department of Biostatistics (YK, DWF); Spinal Cord & Brain Injury Research Center (ADB); Department of Neuroscience (ADB, DWW, LJVE); Department of Epidemiology (ELA); Department of Neurology (DWW, GAJ); Department of Physiology (BCS, SE); and Department of Pathology (W-XW, JHN, PTN), University of Kentucky, Lexington, Kentucky
| |
Collapse
|
44
|
Executive Summary of the 2019 International Conference of Korean Dementia Association: Exploring the Novel Concept of Alzheimer's Disease and Other Dementia: a Report from the Academic Committee of the Korean Dementia Association. Dement Neurocogn Disord 2020; 19:39-53. [PMID: 32602279 PMCID: PMC7326615 DOI: 10.12779/dnd.2020.19.2.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 01/14/2023] Open
Abstract
Because of repeated failures of clinical trials, the concept of Alzheimer's disease (AD) has been changing rapidly in recent years. As suggested by the National Institute on Aging and the Alzheimer's Association Research Framework, the diagnosis and classification of AD is now based on biomarkers rather than on symptoms, allowing more accurate identification of proper candidates for clinical trials by pathogenesis and disease stage. Recent development in neuroimaging has provided a way to reveal the complex dynamics of amyloid and tau in the brain in vivo, and studies of blood biomarkers are taking another leap forward in diagnosis and treatment of AD. In the field of basic and translational research, the development of animal models and a deeper understanding of the role of neuroinflammation are taking a step closer to clarifying the pathogenesis of AD. Development of big data and the Internet of Things is also incorporating dementia care and research into other aspects. Large-scale genetic research has identified genetic abnormalities that can provide a foundation for precision medicine along with the aforementioned digital technologies. Through the first international conference of the Korean Dementia Association, experts from all over the world gathered to exchange opinions with association members on these topics. The Academic Committee of the Korean Dementia Association briefly summarizes the contents of the lectures to convey the depth of the conference and discussions. This will be an important milestone in understanding the latest trends in AD's pathogenesis, diagnostic and therapeutic research and in establishing a future direction.
Collapse
|
45
|
Determination of genetic changes in etiology of autism spectrum disorder in twins by whole-exome sequencing. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
46
|
Nelson PT, Fardo DW, Katsumata Y. The MUC6/AP2A2 Locus and Its Relevance to Alzheimer's Disease: A Review. J Neuropathol Exp Neurol 2020; 79:568-584. [PMID: 32357373 PMCID: PMC7241941 DOI: 10.1093/jnen/nlaa024] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/10/2020] [Indexed: 12/11/2022] Open
Abstract
We recently reported evidence of Alzheimer's disease (AD)-linked genetic variation within the mucin 6 (MUC6) gene on chromosome 11p, nearby the adaptor-related protein complex 2 subunit alpha 2 (AP2A2) gene. This locus has interesting features related to human genomics and clinical research. MUC6 gene variants have been reported to potentially influence viral-including herpesvirus-immunity and the gut microbiome. Within the MUC6 gene is a unique variable number of tandem repeat (VNTR) region. We discovered an association between MUC6 VNTR repeat expansion and AD pathologic severity, particularly tau proteinopathy. Here, we review the relevant literature. The AD-linked VNTR polymorphism may also influence AP2A2 gene expression. AP2A2 encodes a polypeptide component of the adaptor protein complex, AP-2, which is involved in clathrin-coated vesicle function and was previously implicated in AD pathogenesis. To provide background information, we describe some key knowledge gaps in AD genetics research. The "missing/hidden heritability problem" of AD is highlighted. Extensive portions of the human genome, including the MUC6 VNTR, have not been thoroughly evaluated due to limitations of existing high-throughput sequencing technology. We present and discuss additional data, along with cautionary considerations, relevant to the hypothesis that MUC6 repeat expansion influences AD pathogenesis.
Collapse
Affiliation(s)
- Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
- Department of Pathology, University of Kentucky, Lexington, Kentucky
| | - David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
47
|
Curtis D, Bakaya K, Sharma L, Bandyopadhyay S. Weighted burden analysis of exome-sequenced late-onset Alzheimer's cases and controls provides further evidence for a role for PSEN1 and suggests involvement of the PI3K/Akt/GSK-3β and WNT signalling pathways. Ann Hum Genet 2020; 84:291-302. [PMID: 32020597 DOI: 10.1111/ahg.12375] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 11/07/2019] [Accepted: 12/18/2019] [Indexed: 08/01/2024]
Abstract
Previous studies have implicated common and rare genetic variants as risk factors for late-onset Alzheimer's disease (LOAD). Here, weighted burden analysis was applied to over 10,000 exome-sequenced subjects from the Alzheimer's Disease Sequencing Project. Analyses were carried out to investigate whether rare variants predicted to have a functional effect within a gene were more commonly seen in cases or in controls. Confirmatory results were obtained for TREM2, ABCA7, and SORL1. Additional support was provided for PSEN1 (p = 0.0002), which previously had been only weakly implicated in LOAD. There was suggestive evidence that functional variants in PIK3R1, WNT7A, C1R, and EXOC5 might increase risk and that variants in TIAF1 and/or NDRG2 might have a protective effect. Overall, there was strong evidence (p = 5 × 10-6 ) that variants in tyrosine phosphatase genes reduce the risk of developing LOAD. Because PIK3R1 variants are expected to impair PI3K/Akt/GSK-3β signalling while variants in tyrosine phosphatase genes would enhance it, these findings are in line with those from animal models, suggesting that this pathway is protective against Alzheimer's disease.
Collapse
Affiliation(s)
- David Curtis
- UCL Genetics Institute, UCL, London, WC1E 6BT, United Kingdom
- Centre for Psychiatry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Kaushiki Bakaya
- UCL Genetics Institute, UCL, London, WC1E 6BT, United Kingdom
| | - Leona Sharma
- UCL Genetics Institute, UCL, London, WC1E 6BT, United Kingdom
| | | |
Collapse
|
48
|
D’Argenio V, Sarnataro D. New Insights into the Molecular Bases of Familial Alzheimer's Disease. J Pers Med 2020; 10:jpm10020026. [PMID: 32325882 PMCID: PMC7354425 DOI: 10.3390/jpm10020026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Like several neurodegenerative disorders, such as Prion and Parkinson diseases, Alzheimer's disease (AD) is characterized by spreading mechanism of aggregated proteins in the brain in a typical "prion-like" manner. Recent genetic studies have identified in four genes associated with inherited AD (amyloid precursor protein-APP, Presenilin-1, Presenilin-2 and Apolipoprotein E), rare mutations which cause dysregulation of APP processing and alterations of folding of the derived amyloid beta peptide (A). Accumulation and aggregation of A in the brain can trigger a series of intracellular events, including hyperphosphorylation of tau protein, leading to the pathological features of AD. However, mutations in these four genes account for a small of the total genetic risk for familial AD (FAD). Genome-wide association studies have recently led to the identification of additional AD candidate genes. Here, we review an update of well-established, highly penetrant FAD-causing genes with correlation to the protein misfolding pathway, and novel emerging candidate FAD genes, as well as inherited risk factors. Knowledge of these genes and of their correlated biochemical cascade will provide several potential targets for treatment of AD and aging-related disorders.
Collapse
Affiliation(s)
- Valeria D’Argenio
- CEINGE-Biotecnologie Avanzate scarl, via G. Salvatore 486, 80145 Naples, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, via di val Cannuta 247, 00166 Rome, Italy
- Correspondence: (V.D.); (D.S.); Tel.: +39-081-3737909 (V.D.); +39-081-7464575 (D.S.)
| | - Daniela Sarnataro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, via S. Pansini 5, 80131 Naples, Italy
- Correspondence: (V.D.); (D.S.); Tel.: +39-081-3737909 (V.D.); +39-081-7464575 (D.S.)
| |
Collapse
|
49
|
Penney J, Ralvenius WT, Tsai LH. Modeling Alzheimer's disease with iPSC-derived brain cells. Mol Psychiatry 2020; 25:148-167. [PMID: 31391546 PMCID: PMC6906186 DOI: 10.1038/s41380-019-0468-3] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/10/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease is a devastating neurodegenerative disorder with no cure. Countless promising therapeutics have shown efficacy in rodent Alzheimer's disease models yet failed to benefit human patients. While hope remains that earlier intervention with existing therapeutics will improve outcomes, it is becoming increasingly clear that new approaches to understand and combat the pathophysiology of Alzheimer's disease are needed. Human induced pluripotent stem cell (iPSC) technologies have changed the face of preclinical research and iPSC-derived cell types are being utilized to study an array of human conditions, including neurodegenerative disease. All major brain cell types can now be differentiated from iPSCs, while increasingly complex co-culture systems are being developed to facilitate neuroscience research. Many cellular functions perturbed in Alzheimer's disease can be recapitulated using iPSC-derived cells in vitro, and co-culture platforms are beginning to yield insights into the complex interactions that occur between brain cell types during neurodegeneration. Further, iPSC-based systems and genome editing tools will be critical in understanding the roles of the numerous new genes and mutations found to modify Alzheimer's disease risk in the past decade. While still in their relative infancy, these developing iPSC-based technologies hold considerable promise to push forward efforts to combat Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jay Penney
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - William T Ralvenius
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
50
|
Zhang S, Li X, Zhang L, Meng X, Ma L, Zhang G, Wu H, Liang L, Cao M, Mei F. Identification of a Rare PSEN1 Mutation (Thr119Ile) in Late-Onset Alzheimer's Disease With Early Presentation of Behavioral Disturbance. Front Psychiatry 2020; 11:347. [PMID: 32477171 PMCID: PMC7240292 DOI: 10.3389/fpsyt.2020.00347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative dementia. In this study, whole genome sequencing identifies one rare and likely pathogenic mutation in the presenilin 1 (PSEN1) gene (c.356C > T, p.T119I) associated with a frontal variant of AD. Affected individuals in the kindred developed late-onset cognitive decline accompanied with early presentation of psychiatric symptoms. Positive amyloid PiB PET tracing suggested presence of pathophysiological biomarker for AD. Whole genome sequencing analysis evaluated rare coding mutations in susceptible genes for various types of dementia and supported the role of PSEN1 as a causal gene. Identification of this T119I variant in PSEN1 might broaden the spectrum of genetic basis and clinical diversity of familial AD.
Collapse
Affiliation(s)
- Shouzi Zhang
- Psychiatry Department, Beijing Geriatric Hospital, Beijing, China
| | - Xiang Li
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Li Zhang
- Psychiatry Department, Beijing Geriatric Hospital, Beijing, China
| | - Xiangyan Meng
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Li Ma
- Psychiatry Department, Beijing Geriatric Hospital, Beijing, China
| | - Guangze Zhang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Haiyan Wu
- Psychiatry Department, Beijing Geriatric Hospital, Beijing, China
| | - Ling Liang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Meng Cao
- Psychiatry Department, Beijing Geriatric Hospital, Beijing, China
| | - Fan Mei
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|