1
|
Wang J, Wen S, Liu W, Meng X, Jiao Z. Deep joint learning diagnosis of Alzheimer's disease based on multimodal feature fusion. BioData Min 2024; 17:48. [PMID: 39501294 PMCID: PMC11536794 DOI: 10.1186/s13040-024-00395-9] [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: 08/28/2024] [Accepted: 10/08/2024] [Indexed: 11/09/2024] Open
Abstract
Alzheimer's disease (AD) is an advanced and incurable neurodegenerative disease. Genetic variations are intrinsic etiological factors contributing to the abnormal expression of brain function and structure in AD patients. A new multimodal feature fusion called "magnetic resonance imaging (MRI)-p value" was proposed to construct 3D fusion images by introducing genes as a priori knowledge. Moreover, a new deep joint learning diagnostic model was constructed to fully learn images features. One branch trained a residual network (ResNet) to learn the features of local pathological regions. The other branch learned the position information of brain regions with different changes in the different categories of subjects' brains by introducing attention convolution, and then obtained the discriminative probability information from locations via convolution and global average pooling. The feature and position information of the two branches were linearly interacted to acquire the diagnostic basis for classifying the different categories of subjects. The diagnoses of AD and health control (HC), AD and mild cognitive impairment (MCI), HC and MCI were performed with data from the Alzheimer's Disease Neuroimaging Initiative (ADNI). The results showed that the proposed method achieved optimal results in AD-related diagnosis. The classification accuracy (ACC) and area under the curve (AUC) of the three experimental groups were 93.44% and 96.67%, 89.06% and 92%, and 84% and 81.84%, respectively. Moreover, a total of six novel genes were found to be significantly associated with AD, namely NTM, MAML2, NAALADL2, FHIT, TMEM132D and PCSK5, which provided new targets for the potential treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jingru Wang
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, China
| | - Shipeng Wen
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, China
| | - Wenjie Liu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, 213032, China
| | - Xianglian Meng
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, 213032, China.
- Wangzheng School of Microelectronics, Changzhou University, Changzhou, 213164, China.
| | - Zhuqing Jiao
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, China.
- Wangzheng School of Microelectronics, Changzhou University, Changzhou, 213164, China.
| |
Collapse
|
2
|
Shade LMP, Katsumata Y, Abner EL, Aung KZ, Claas SA, Qiao Q, Heberle BA, Brandon JA, Page ML, Hohman TJ, Mukherjee S, Mayeux RP, Farrer LA, Schellenberg GD, Haines JL, Kukull WA, Nho K, Saykin AJ, Bennett DA, Schneider JA, Ebbert MTW, Nelson PT, Fardo DW. GWAS of multiple neuropathology endophenotypes identifies new risk loci and provides insights into the genetic risk of dementia. Nat Genet 2024; 56:2407-2421. [PMID: 39379761 PMCID: PMC11549054 DOI: 10.1038/s41588-024-01939-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/30/2024] [Indexed: 10/10/2024]
Abstract
Genome-wide association studies (GWAS) have identified >80 Alzheimer's disease and related dementias (ADRD)-associated genetic loci. However, the clinical outcomes used in most previous studies belie the complex nature of underlying neuropathologies. Here we performed GWAS on 11 ADRD-related neuropathology endophenotypes with participants drawn from the following three sources: the National Alzheimer's Coordinating Center, the Religious Orders Study and Rush Memory and Aging Project, and the Adult Changes in Thought study (n = 7,804 total autopsied participants). We identified eight independent significantly associated loci, of which four were new (COL4A1, PIK3R5, LZTS1 and APOC2). Separately testing known ADRD loci, 19 loci were significantly associated with at least one neuropathology after false-discovery rate adjustment. Genetic colocalization analyses identified pleiotropic effects and quantitative trait loci. Methylation in the cerebral cortex at two sites near APOC2 was associated with cerebral amyloid angiopathy. Studies that include neuropathology endophenotypes are an important step in understanding the mechanisms underlying genetic ADRD risk.
Collapse
Affiliation(s)
- Lincoln M P Shade
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Yuriko Katsumata
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Epidemiology and Environmental Health, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Khine Zin Aung
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
| | - Steven A Claas
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Qi Qiao
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
| | - Bernardo Aguzzoli Heberle
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - J Anthony Brandon
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Madeline L Page
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Richard P Mayeux
- Department of Neurology, Columbia University, New York City, NY, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Penn Neurodegeneration Genomics Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan L Haines
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Walter A Kukull
- National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
- Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
| | - Julie A Schneider
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
- Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
- Department of Pathology, Rush Medical College, Chicago, IL, USA
| | - Mark T W Ebbert
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY, USA
| | - David W Fardo
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA.
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
3
|
Niedowicz DM, Wang WX, Prajapati P, Zhong Y, Fister S, Rogers CB, Sompol P, Powell DK, Patel I, Norris CM, Saatman KE, Nelson PT. Nicorandil treatment improves survival and spatial learning in aged granulin knockout mice. Brain Pathol 2024:e13312. [PMID: 39438022 DOI: 10.1111/bpa.13312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Mutations in the human granulin (GRN) gene are associated with multiple diseases, including dementia disorders such as frontotemporal dementia (FTD) and limbic-predominant age-related TDP-43 encephalopathy (LATE). We studied a Grn knockout (Grn-KO) mouse model in order to evaluate a potential therapeutic strategy for these diseases using nicorandil, a commercially available agonist for the ABCC9/Abcc9-encoded regulatory subunit of the "K+ATP" channel that is well-tolerated in humans. Aged (13 months) Grn-KO and wild-type (WT) mice were treated as controls or with nicorandil (15 mg/kg/day) in drinking water for 7 months, then tested for neurobehavioral performance, neuropathology, and gene expression. Mortality was significantly higher for aged Grn-KO mice (particularly females), but there was a conspicuous improvement in survival for both sexes treated with nicorandil. Grn-KO mice performed worse on some cognitive tests than WT mice, but Morris Water Maze performance was improved with nicorandil treatment. Neuropathologically, Grn-KO mice had significantly increased levels of glial fibrillary acidic protein (GFAP)-immunoreactive astrocytosis but not ionized calcium binding adaptor molecule 1 (IBA-1)-immunoreactive microgliosis, indicating cell-specific inflammation in the brain. Expression of several astrocyte-enriched genes, including Gfap, were also elevated in the Grn-KO brain. Nicorandil treatment was associated with a subtle shift in a subset of detected brain transcript levels, mostly related to attenuated inflammatory markers. Nicorandil treatment improved survival outcomes, cognition, and inflammation in aged Grn-KO mice.
Collapse
Affiliation(s)
- Dana M Niedowicz
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Wang-Xia Wang
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| | - Paresh Prajapati
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Yu Zhong
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Shuling Fister
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Colin B Rogers
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Pradoldej Sompol
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - David K Powell
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Indumati Patel
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Kathryn E Saatman
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Peter T Nelson
- Sanders Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Nelson PT, Fardo DW, Wu X, Aung KZ, Cykowski MD, Katsumata Y. Limbic-predominant age-related TDP-43 encephalopathy (LATE-NC): Co-pathologies and genetic risk factors provide clues about pathogenesis. J Neuropathol Exp Neurol 2024; 83:396-415. [PMID: 38613823 PMCID: PMC11110076 DOI: 10.1093/jnen/nlae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is detectable at autopsy in more than one-third of people beyond age 85 years and is robustly associated with dementia independent of other pathologies. Although LATE-NC has a large impact on public health, there remain uncertainties about the underlying biologic mechanisms. Here, we review the literature from human studies that may shed light on pathogenetic mechanisms. It is increasingly clear that certain combinations of pathologic changes tend to coexist in aging brains. Although "pure" LATE-NC is not rare, LATE-NC often coexists in the same brains with Alzheimer disease neuropathologic change, brain arteriolosclerosis, hippocampal sclerosis of aging, and/or age-related tau astrogliopathy (ARTAG). The patterns of pathologic comorbidities provide circumstantial evidence of mechanistic interactions ("synergies") between the pathologies, and also suggest common upstream influences. As to primary mediators of vulnerability to neuropathologic changes, genetics may play key roles. Genes associated with LATE-NC include TMEM106B, GRN, APOE, SORL1, ABCC9, and others. Although the anatomic distribution of TDP-43 pathology defines the condition, important cofactors for LATE-NC may include Tau pathology, endolysosomal pathways, and blood-brain barrier dysfunction. A review of the human phenomenology offers insights into disease-driving mechanisms, and may provide clues for diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - David W Fardo
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Xian Wu
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Khine Zin Aung
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Yuriko Katsumata
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
5
|
Efthymiou S, Scala M, Nagaraj V, Ochenkowska K, Komdeur FL, Liang RA, Abdel-Hamid MS, Sultan T, Barøy T, Van Ghelue M, Vona B, Maroofian R, Zafar F, Alkuraya FS, Zaki MS, Severino M, Duru KC, Tryon RC, Brauteset LV, Ansari M, Hamilton M, van Haelst MM, van Haaften G, Zara F, Houlden H, Samarut É, Nichols CG, Smeland MF, McClenaghan C. Novel loss-of-function variants expand ABCC9-related intellectual disability and myopathy syndrome. Brain 2024; 147:1822-1836. [PMID: 38217872 PMCID: PMC11068106 DOI: 10.1093/brain/awae010] [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: 06/25/2023] [Revised: 11/22/2023] [Accepted: 12/30/2023] [Indexed: 01/15/2024] Open
Abstract
Loss-of-function mutation of ABCC9, the gene encoding the SUR2 subunit of ATP sensitive-potassium (KATP) channels, was recently associated with autosomal recessive ABCC9-related intellectual disability and myopathy syndrome (AIMS). Here we identify nine additional subjects, from seven unrelated families, harbouring different homozygous loss-of-function variants in ABCC9 and presenting with a conserved range of clinical features. All variants are predicted to result in severe truncations or in-frame deletions within SUR2, leading to the generation of non-functional SUR2-dependent KATP channels. Affected individuals show psychomotor delay and intellectual disability of variable severity, microcephaly, corpus callosum and white matter abnormalities, seizures, spasticity, short stature, muscle fatigability and weakness. Heterozygous parents do not show any conserved clinical pathology but report multiple incidences of intra-uterine fetal death, which were also observed in an eighth family included in this study. In vivo studies of abcc9 loss-of-function in zebrafish revealed an exacerbated motor response to pentylenetetrazole, a pro-convulsive drug, consistent with impaired neurodevelopment associated with an increased seizure susceptibility. Our findings define an ABCC9 loss-of-function-related phenotype, expanding the genotypic and phenotypic spectrum of AIMS and reveal novel human pathologies arising from KATP channel dysfunction.
Collapse
Affiliation(s)
- Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Marcello Scala
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16147 Genoa, Italy
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Vini Nagaraj
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers the State University of New Jersey, Piscatway, NJ 08854, USA
| | - Katarzyna Ochenkowska
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), and Department of Neuroscience, Université de Montréal, Montreal H2X 0A9, Quebec, Canada
| | - Fenne L Komdeur
- Section Clinical Genetics, Department of Human Genetics and Amsterdam Reproduction and Development, Amsterdam University Medical Centers, 1105 AZ, Amsterdam, The Netherlands
| | - Robin A Liang
- Department of Medical Genetics, Division of Child and Adolescent Health, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Mohamed S Abdel-Hamid
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Tipu Sultan
- Department of Pediatric Neurology, Children Hospital, University of Child Health Sciences, Lahore, Punjab 54000, Pakistan
| | - Tuva Barøy
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Marijke Van Ghelue
- Department of Medical Genetics, Division of Child and Adolescent Health, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Barbara Vona
- Institute of Human Genetics and Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Reza Maroofian
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Faisal Zafar
- Department of Paediatric Neurology, Children’s Hospital and Institute of Child Health, Multan, Punjab 60000, Pakistan
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 12713, Saudi Arabia
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | | | - Kingsley C Duru
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers the State University of New Jersey, Piscatway, NJ 08854, USA
| | - Robert C Tryon
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO 63110, USA
| | - Lin Vigdis Brauteset
- Division of Habilitation for Children, Innlandet Hospital Sanderud, Hamar 2312, Norway
| | - Morad Ansari
- South East Scotland Genetic Service, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Mark Hamilton
- West of Scotland Clinical Genetics Service, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Mieke M van Haelst
- Section Clinical Genetics, Department of Human Genetics and Amsterdam Reproduction and Development, Amsterdam University Medical Centers, 1105 AZ, Amsterdam, The Netherlands
| | - Gijs van Haaften
- Department of Genetics, University Medical Center, Utrecht, 3584 CX, The Netherlands
| | - Federico Zara
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Éric Samarut
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), and Department of Neuroscience, Université de Montréal, Montreal H2X 0A9, Quebec, Canada
| | - Colin G Nichols
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO 63110, USA
| | - Marie F Smeland
- Department of Pediatric Rehabilitation, University Hospital of North Norway, 9019 Tromsø, Norway
- Institute of Clinical Medicine, UiT The Arctic University of Norway, 9019, Tromsø, Norway
| | - Conor McClenaghan
- Center for Advanced Biotechnology and Medicine, and Departments of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers the State University of New Jersey, Piscatway, NJ 08854, USA
| |
Collapse
|
6
|
Picard C, Miron J, Poirier J. Association of TMEM106B with Cortical APOE Gene Expression in Neurodegenerative Conditions. Genes (Basel) 2024; 15:416. [PMID: 38674351 PMCID: PMC11049136 DOI: 10.3390/genes15040416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
The e4 allele of the apolipoprotein E gene is the strongest genetic risk factor for sporadic Alzheimer's disease. Nevertheless, how APOE is regulated is still elusive. In a trans-eQTL analysis, we found a genome-wide significant association between transmembrane protein 106B (TMEM106B) genetic variants and cortical APOE mRNA levels in human brains. The goal of this study is to determine whether TMEM106B is mis-regulated in Alzheimer's disease or in other neurodegenerative conditions. Available genomic, transcriptomic and proteomic data from human brains were downloaded from the Mayo Clinic Brain Bank and the Religious Orders Study and Memory and Aging Project. An in-house mouse model of the hippocampal deafferentation/reinnervation was achieved via a stereotaxic lesioning surgery to the entorhinal cortex, and mRNA levels were measured using RNAseq technology. In human temporal cortices, the mean TMEM106B expression was significantly higher in Alzheimer's disease compared to cognitively unimpaired individuals. In the mouse model, hippocampal Tmem106b reached maximum levels during the early phase of reinnervation. These results suggest an active response to tissue damage that is consistent with compensatory synaptic and terminal remodeling.
Collapse
Affiliation(s)
- Cynthia Picard
- Douglas Mental Health University Institute, Montreal, QC H4H 1R3, Canada; (C.P.); (J.M.)
- Centre for the Studies on Prevention of Alzheimer’s Disease, Montreal, QC H4H 1R3, Canada
| | - Justin Miron
- Douglas Mental Health University Institute, Montreal, QC H4H 1R3, Canada; (C.P.); (J.M.)
- Centre for the Studies on Prevention of Alzheimer’s Disease, Montreal, QC H4H 1R3, Canada
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC H3A 0E7, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montreal, QC H4H 1R3, Canada; (C.P.); (J.M.)
- Centre for the Studies on Prevention of Alzheimer’s Disease, Montreal, QC H4H 1R3, Canada
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC H3A 0E7, Canada
| |
Collapse
|
7
|
Nag S, Schneider JA. Limbic-predominant age-related TDP43 encephalopathy (LATE) neuropathological change in neurodegenerative diseases. Nat Rev Neurol 2023; 19:525-541. [PMID: 37563264 PMCID: PMC10964248 DOI: 10.1038/s41582-023-00846-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 08/12/2023]
Abstract
TAR DNA-binding protein 43 (TDP43) is a focus of research in late-onset dementias. TDP43 pathology in the brain was initially identified in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, and later in Alzheimer disease (AD), other neurodegenerative diseases and ageing. Limbic-predominant age-related TDP43 encephalopathy (LATE), recognized as a clinical entity in 2019, is characterized by amnestic dementia resembling AD dementia and occurring most commonly in adults over 80 years of age. Neuropathological findings in LATE, referred to as LATE neuropathological change (LATE-NC), consist of neuronal and glial cytoplasmic TDP43 localized predominantly in limbic areas with or without coexisting hippocampal sclerosis and/or AD neuropathological change and without frontotemporal lobar degeneration or amyotrophic lateral sclerosis pathology. LATE-NC is frequently associated with one or more coexisting pathologies, mainly AD neuropathological change. The focus of this Review is the pathology, genetic risk factors and nature of the cognitive impairments and dementia in pure LATE-NC and in LATE-NC associated with coexisting pathologies. As the clinical and cognitive profile of LATE is currently not easily distinguishable from AD dementia, it is important to develop biomarkers to aid in the diagnosis of this condition in the clinic. The pathogenesis of LATE-NC should be a focus of future research to form the basis for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Sukriti Nag
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
8
|
Cochran JN, Acosta-Uribe J, Esposito BT, Madrigal L, Aguillón D, Giraldo MM, Taylor JW, Bradley J, Fulton-Howard B, Andrews SJ, Acosta-Baena N, Alzate D, Garcia GP, Piedrahita F, Lopez HE, Anderson AG, Rodriguez-Nunez I, Roberts K, Dominantly Inherited Alzheimer Network, Absher D, Myers RM, Beecham GW, Reitz C, Rizzardi LF, Fernandez MV, Goate AM, Cruchaga C, Renton AE, Lopera F, Kosik KS. Genetic associations with age at dementia onset in the PSEN1 E280A Colombian kindred. Alzheimers Dement 2023; 19:3835-3847. [PMID: 36951251 PMCID: PMC10514237 DOI: 10.1002/alz.13021] [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: 10/31/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 03/24/2023]
Abstract
INTRODUCTION Genetic associations with Alzheimer's disease (AD) age at onset (AAO) could reveal genetic variants with therapeutic applications. We present a large Colombian kindred with autosomal dominant AD (ADAD) as a unique opportunity to discover AAO genetic associations. METHODS A genetic association study was conducted to examine ADAD AAO in 340 individuals with the PSEN1 E280A mutation via TOPMed array imputation. Replication was assessed in two ADAD cohorts, one sporadic early-onset AD study and four late-onset AD studies. RESULTS 13 variants had p<1×10-7 or p<1×10-5 with replication including three independent loci with candidate associations with clusterin including near CLU. Other suggestive associations were identified in or near HS3ST1, HSPG2, ACE, LRP1B, TSPAN10, and TSPAN14. DISCUSSION Variants with suggestive associations with AAO were associated with biological processes including clusterin, heparin sulfate, and amyloid processing. The detection of these effects in the presence of a strong mutation for ADAD reinforces their potentially impactful role.
Collapse
Affiliation(s)
| | - Juliana Acosta-Uribe
- Neuroscience Research Institute, University of California, Santa Barbara, California, and Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, California, USA
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Bianca T Esposito
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lucia Madrigal
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - David Aguillón
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Margarita M Giraldo
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Jared W Taylor
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Joseph Bradley
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brian Fulton-Howard
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shea J Andrews
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Natalia Acosta-Baena
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Diana Alzate
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Gloria P Garcia
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Francisco Piedrahita
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Hugo E Lopez
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | | | | | - Kevin Roberts
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | | | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Gary W Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Christiane Reitz
- Department of Epidemiology, Sergievsky Center, Taub Institute for Research on the Aging Brain, Columbia University, New York, New York, USA
| | | | | | - Alison M Goate
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carlos Cruchaga
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alan E Renton
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia. School of Medicine. Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Kenneth S Kosik
- Neuroscience Research Institute, University of California, Santa Barbara, California, and Department of Molecular Cellular and Developmental Biology University of California, Santa Barbara, California, USA
| |
Collapse
|
9
|
Katsumata Y, Fardo DW, Shade LMP, Nelson PT. LATE-NC risk alleles (in TMEM106B, GRN, and ABCC9 genes) among persons with African ancestry. J Neuropathol Exp Neurol 2023; 82:760-768. [PMID: 37528055 PMCID: PMC10440720 DOI: 10.1093/jnen/nlad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy (LATE) affects approximately one-third of older individuals and is associated with cognitive impairment. However, there is a highly incomplete understanding of the genetic determinants of LATE neuropathologic changes (LATE-NC) in diverse populations. The defining neuropathologic feature of LATE-NC is TDP-43 proteinopathy, often with comorbid hippocampal sclerosis (HS). In terms of genetic risk factors, LATE-NC and/or HS are associated with single nucleotide variants (SNVs) in 3 genes-TMEM106B (rs1990622), GRN (rs5848), and ABCC9 (rs1914361 and rs701478). We evaluated these 3 genes in convenience samples of individuals of African ancestry. The allele frequencies of the LATE-associated alleles were significantly different between persons of primarily African (versus European) ancestry: In persons of African ancestry, the risk-associated alleles for TMEM106B and ABCC9 were less frequent, whereas the risk allele in GRN was more frequent. We performed an exploratory analysis of data from African-American subjects processed by the Alzheimer's Disease Genomics Consortium, with a subset of African-American participants (n = 166) having corroborating neuropathologic data through the National Alzheimer's Coordinating Center (NACC). In this limited-size sample, the ABCC9/rs1914361 SNV was associated with HS pathology. More work is required concerning the genetic factors influencing non-Alzheimer disease pathology such as LATE-NC in diverse cohorts.
Collapse
Affiliation(s)
- Yuriko Katsumata
- University of Kentucky Sanders-Brown Center on Aging, Lexington, Kentucky, USA
- University of Kentucky Department of Biostatistics, Lexington, Kentucky, USA
| | - David W Fardo
- University of Kentucky Sanders-Brown Center on Aging, Lexington, Kentucky, USA
- University of Kentucky Department of Biostatistics, Lexington, Kentucky, USA
| | - Lincoln M P Shade
- University of Kentucky Department of Biostatistics, Lexington, Kentucky, USA
| | - Peter T Nelson
- University of Kentucky Sanders-Brown Center on Aging, Lexington, Kentucky, USA
- University of Kentucky Department of Pathology and Laboratory Medicine, Lexington, Kentucky, USA
| |
Collapse
|
10
|
Nelson PT, Schneider JA, Jicha GA, Duong MT, Wolk DA. When Alzheimer's is LATE: Why Does it Matter? Ann Neurol 2023; 94:211-222. [PMID: 37245084 PMCID: PMC10516307 DOI: 10.1002/ana.26711] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/07/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
Recent therapeutic advances provide heightened motivation for accurate diagnosis of the underlying biologic causes of dementia. This review focuses on the importance of clinical recognition of limbic-predominant age-related TDP-43 encephalopathy (LATE). LATE affects approximately one-quarter of older adults and produces an amnestic syndrome that is commonly mistaken for Alzheimer's disease (AD). Although AD and LATE often co-occur in the same patients, these diseases differ in the protein aggregates driving neuropathology (Aβ amyloid/tau vs TDP-43). This review discusses signs and symptoms, relevant diagnostic testing, and potential treatment implications for LATE that may be helpful for physicians, patients, and families. ANN NEUROL 2023;94:211-222.
Collapse
Affiliation(s)
| | | | | | | | - David A. Wolk
- University of Pennsylvania Alzheimer’s Disease Research Center
| |
Collapse
|
11
|
Zhao B, Li T, Fan Z, Yang Y, Shu J, Yang X, Wang X, Luo T, Tang J, Xiong D, Wu Z, Li B, Chen J, Shan Y, Tomlinson C, Zhu Z, Li Y, Stein JL, Zhu H. Heart-brain connections: Phenotypic and genetic insights from magnetic resonance images. Science 2023; 380:abn6598. [PMID: 37262162 DOI: 10.1126/science.abn6598] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/11/2023] [Indexed: 06/03/2023]
Abstract
Cardiovascular health interacts with cognitive and mental health in complex ways, yet little is known about the phenotypic and genetic links of heart-brain systems. We quantified heart-brain connections using multiorgan magnetic resonance imaging (MRI) data from more than 40,000 subjects. Heart MRI traits displayed numerous association patterns with brain gray matter morphometry, white matter microstructure, and functional networks. We identified 80 associated genomic loci (P < 6.09 × 10-10) for heart MRI traits, which shared genetic influences with cardiovascular and brain diseases. Genetic correlations were observed between heart MRI traits and brain-related traits and disorders. Mendelian randomization suggests that heart conditions may causally contribute to brain disorders. Our results advance a multiorgan perspective on human health by revealing heart-brain connections and shared genetic influences.
Collapse
Affiliation(s)
- Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiarui Tang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Di Xiong
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhenyi Wu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Bingxuan Li
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yue Shan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chalmer Tomlinson
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Nicks R, Clement NF, Alvarez VE, Tripodis Y, Baucom ZH, Huber BR, Mez J, Alosco ML, Aytan N, Cherry JD, Cormier KA, Kubilius C, Mathias R, Svirsky SE, Pothast MJ, Hildebrandt AM, Chung J, Han X, Crary JF, McKee AC, Frosch MP, Stein TD. Repetitive head impacts and chronic traumatic encephalopathy are associated with TDP-43 inclusions and hippocampal sclerosis. Acta Neuropathol 2023; 145:395-408. [PMID: 36681782 PMCID: PMC11360224 DOI: 10.1007/s00401-023-02539-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/22/2023]
Abstract
Hippocampal sclerosis (HS) is associated with advanced age as well as transactive response DNA-binding protein with 43 kDa (TDP-43) deposits. Both hippocampal sclerosis and TDP-43 proteinopathy have also been described in chronic traumatic encephalopathy (CTE), a neurodegenerative disease linked to exposure to repetitive head impacts (RHI). However, the prevalence of HS in CTE, the pattern of TDP-43 pathology, and associations of HS and TDP-43 with RHI are unknown. A group of participants with a history of RHI and CTE at autopsy (n = 401) as well as a group with HS-aging without CTE (n = 33) was examined to determine the prevalence of HS and TDP-43 inclusions in CTE and to compare the clinical and pathological features of HS and TDP-43 inclusions in CTE to HS-aging. In CTE, HS was present in 23.4%, and TDP-43 inclusions were present in 43.3% of participants. HS in CTE occurred at a relatively young age (mean 77.0 years) and was associated with a greater number of years of RHI than CTE without HS adjusting for age (p = 0.029). In CTE, TDP-43 inclusions occurred frequently in the frontal cortex and occurred both with and without limbic TDP-43. Additionally, structural equation modeling demonstrated that RHI exposure years were associated with hippocampal TDP-43 inclusions (p < 0.001) through increased CTE stage (p < 0.001). Overall, RHI and the development of CTE pathology may contribute to TDP-43 deposition and hippocampal sclerosis.
Collapse
Affiliation(s)
- Raymond Nicks
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
| | - Nathan F Clement
- C.S. Kubik Laboratory for Neuropathology, Pathology Service, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pathology and Laboratory Services, Brooke Army Medical Center, Fort Sam Houston, San Antonio, TX, USA
| | - Victor E Alvarez
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Zachery H Baucom
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Bertrand R Huber
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Nurgul Aytan
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Jonathan D Cherry
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Kerry A Cormier
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- VA Boston Healthcare System, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Carol Kubilius
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
| | - Rebecca Mathias
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
| | - Sarah E Svirsky
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
| | - Morgan J Pothast
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
| | | | - Jaeyoon Chung
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Xudong Han
- Boston University Bioinformatics Graduate Program, Boston, MA, USA
| | - John F Crary
- Department of Pathology, Nash Family Department of Neuroscience, Department of Artificial Intelligence and Human Health, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ann C McKee
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Matthew P Frosch
- C.S. Kubik Laboratory for Neuropathology, Pathology Service, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, 02130, USA.
- VA Boston Healthcare System, Boston, MA, USA.
- VA Bedford Healthcare System, Bedford, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
13
|
I F. The unique neuropathological vulnerability of the human brain to aging. Ageing Res Rev 2023; 87:101916. [PMID: 36990284 DOI: 10.1016/j.arr.2023.101916] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD)-related neurofibrillary tangles (NFT), argyrophilic grain disease (AGD), aging-related tau astrogliopathy (ARTAG), limbic predominant TDP-43 proteinopathy (LATE), and amygdala-predominant Lewy body disease (LBD) are proteinopathies that, together with hippocampal sclerosis, progressively appear in the elderly affecting from 50% to 99% of individuals aged 80 years, depending on the disease. These disorders usually converge on the same subject and associate with additive cognitive impairment. Abnormal Tau, TDP-43, and α-synuclein pathologies progress following a pattern consistent with an active cell-to-cell transmission and abnormal protein processing in the host cell. However, cell vulnerability and transmission pathways are specific for each disorder, albeit abnormal proteins may co-localize in particular neurons. All these alterations are unique or highly prevalent in humans. They all affect, at first, the archicortex and paleocortex to extend at later stages to the neocortex and other regions of the telencephalon. These observations show that the phylogenetically oldest areas of the human cerebral cortex and amygdala are not designed to cope with the lifespan of actual humans. New strategies aimed at reducing the functional overload of the human telencephalon, including optimization of dream repair mechanisms and implementation of artificial circuit devices to surrogate specific brain functions, appear promising.
Collapse
Affiliation(s)
- Ferrer I
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain; Biomedical Research Network of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
14
|
Duong MT, Wolk DA. Limbic-Predominant Age-Related TDP-43 Encephalopathy: LATE-Breaking Updates in Clinicopathologic Features and Biomarkers. Curr Neurol Neurosci Rep 2022; 22:689-698. [PMID: 36190653 PMCID: PMC9633415 DOI: 10.1007/s11910-022-01232-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Limbic-predominant age-related TDP-43 encephalopathy (LATE) is a recently defined neurodegenerative disease characterized by amnestic phenotype and pathological inclusions of TAR DNA-binding protein 43 (TDP-43). LATE is distinct from rarer forms of TDP-43 diseases such as frontotemporal lobar degeneration with TDP-43 but is also a common copathology with Alzheimer's disease (AD) and cerebrovascular disease and accelerates cognitive decline. LATE contributes to clinicopathologic heterogeneity in neurodegenerative diseases, so it is imperative to distinguish LATE from other etiologies. RECENT FINDINGS Novel biomarkers for LATE are being developed with magnetic resonance imaging (MRI) and positron emission tomography (PET). When cooccurring with AD, LATE exhibits identifiable patterns of limbic-predominant atrophy on MRI and hypometabolism on 18F-fluorodeoxyglucose PET that are greater than expected relative to levels of local AD pathology. Efforts are being made to develop TDP-43-specific radiotracers, molecularly specific biofluid measures, and genomic predictors of TDP-43. LATE is a highly prevalent neurodegenerative disease distinct from previously characterized cognitive disorders.
Collapse
Affiliation(s)
- Michael Tran Duong
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute On Aging, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce St, Philadelphia, PA, 19104, USA.
| |
Collapse
|
15
|
Katsumata Y, Shade LM, Hohman TJ, Schneider JA, Bennett DA, Farfel JM, Kukull WA, Fardo DW, Nelson PT. Multiple gene variants linked to Alzheimer's-type clinical dementia via GWAS are also associated with non-Alzheimer's neuropathologic entities. Neurobiol Dis 2022; 174:105880. [PMID: 36191742 PMCID: PMC9641973 DOI: 10.1016/j.nbd.2022.105880] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/26/2022] Open
Abstract
The classic pathologic hallmarks of Alzheimer's disease (AD) are amyloid plaques and neurofibrillary tangles (AD neuropathologic changes, or ADNC). However, brains from individuals clinically diagnosed with "AD-type" (amnestic) dementia usually harbor heterogeneous neuropathologies in addition to, or other than, ADNC. We hypothesized that some AD-type dementia associated genetic single nucleotide variants (SNVs) identified from large genomewide association studies (GWAS) were associated with non-ADNC neuropathologies. To test this hypothesis, we analyzed data from multiple studies with available genotype and neuropathologic phenotype information. Clinical AD/dementia risk alleles of interest were derived from the very large GWAS by Bellenguez et al. (2022) who reported 83 clinical AD/dementia-linked SNVs in addition to the APOE risk alleles. To query the pathologic phenotypes associated with variation of those SNVs, National Alzheimer's disease Coordinating Center (NACC) neuropathologic data were linked to AD Sequencing Project (ADSP) and AD Genomics Consortium (ADGC) data. Separate data were obtained from the harmonized Religious Orders Study and the Rush Memory and Aging Project (ROSMAP). A total of 4811 European participants had at least ADNC neuropathology data and also genotype data available; data were meta-analyzed across cohorts. As expected, a subset of dementia-associated SNVs were associated with ADNC risk in Europeans-e.g., BIN1, PICALM, CR1, MME, and COX7C. Other gene variants linked to (clinical) AD dementia were associated with non-ADNC pathologies. For example, the associations of GRN and TMEM106B SNVs with limbic-predominant age-related TDP-43 neuropathologic changes (LATE-NC) were replicated. In addition, SNVs in TNIP1 and WNT3 previously reported as AD-related were instead associated with hippocampal sclerosis pathology. Some genotype/neuropathology association trends were not statistically significant at P < 0.05 after correcting for multiple testing, but were intriguing. For example, variants in SORL1 and TPCN1 showed trends for association with LATE-NC whereas Lewy body pathology trended toward association with USP6NL and BIN1 gene variants. A smaller cohort of non-European subjects (n = 273, approximately one-half of whom were African-Americans) provided the basis for additional exploratory analyses. Overall, these findings were consistent with the hypothesis that some genetic variants linked to AD dementia risk exert their affect by influencing non-ADNC neuropathologies.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Lincoln M Shade
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julie A Schneider
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jose M Farfel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Walter A Kukull
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Pathology, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
16
|
Ruffini N, Klingenberg S, Heese R, Schweiger S, Gerber S. The Big Picture of Neurodegeneration: A Meta Study to Extract the Essential Evidence on Neurodegenerative Diseases in a Network-Based Approach. Front Aging Neurosci 2022; 14:866886. [PMID: 35832065 PMCID: PMC9271745 DOI: 10.3389/fnagi.2022.866886] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
The common features of all neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), and Huntington's disease, are the accumulation of aggregated and misfolded proteins and the progressive loss of neurons, leading to cognitive decline and locomotive dysfunction. Still, they differ in their ultimate manifestation, the affected brain region, and the kind of proteinopathy. In the last decades, a vast number of processes have been described as associated with neurodegenerative diseases, making it increasingly harder to keep an overview of the big picture forming from all those data. In this meta-study, we analyzed genomic, transcriptomic, proteomic, and epigenomic data of the aforementioned diseases using the data of 234 studies in a network-based approach to study significant general coherences but also specific processes in individual diseases or omics levels. In the analysis part, we focus on only some of the emerging findings, but trust that the meta-study provided here will be a valuable resource for various other researchers focusing on specific processes or genes contributing to the development of neurodegeneration.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research, Leibniz Association, Mainz, Germany
| | - Susanne Klingenberg
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Raoul Heese
- Fraunhofer Institute for Industrial Mathematics (ITWM), Kaiserslautern, Germany
| | - Susann Schweiger
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
17
|
Ogura J, Yamaguchi H. The Effectiveness of Antidiabetic Drugs in Treating Dementia: A Peek into Pharmacological and Pharmacokinetic Properties. Int J Mol Sci 2022; 23:6542. [PMID: 35742986 PMCID: PMC9223777 DOI: 10.3390/ijms23126542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/04/2022] Open
Abstract
Dementia dramatically affects the activities of daily living and quality of life; thus, many therapeutic approaches for overcoming dementia have been developed. However, an effective treatment regimen is yet to be developed. As diabetes is a well-known risk factor for dementia, drug repositioning and repurposing of antidiabetic drugs are expected to be effective dementia treatments. Several observational studies have been useful for understanding the effectiveness of antidiabetic drugs in treating dementia, but it is difficult to conclusively analyze the association between antidiabetic drug treatment and the risk of developing dementia after correcting for potential confounding factors. Mechanism-based approaches may provide a better understanding of the effectiveness of antidiabetic drugs for treating dementia. Since the peripheral circulation and the central nerve system are separated by the blood-brain barrier, it is important to understand the regulation of the central glucose metabolism. In this review, we discuss the pharmacological and pharmacokinetic properties of antidiabetic drugs in relation to treating dementia.
Collapse
Affiliation(s)
- Jiro Ogura
- Department of Pharmacy, Yamagata University Hospital, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan;
| | | |
Collapse
|
18
|
Wang L, Jiao Y, Zhao A, Xu X, Ye G, Zhang Y, Wang Y, Deng Y, Xu W, Liu J. Analysis of Genetic Association Between ABCA7 Polymorphism and Alzheimer’s Disease Risk in the Southern Chinese Population. Front Aging Neurosci 2022; 14:819499. [PMID: 35693347 PMCID: PMC9175022 DOI: 10.3389/fnagi.2022.819499] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/19/2022] [Indexed: 11/14/2022] Open
Abstract
Objective The study aimed to clarify the association of the 21 single nucleotide polymorphisms (SNPs) with Alzheimer’s disease (AD) in the population of southern China. Methods A case-control study was conducted with a total sample size of 490 subjects (246 patients with AD and 244 age- and gender-matched healthy controls) enrolled in this study. Twenty-one selected SNPs were detected using SNaPshot assay and polymerase chain reaction (PCR) technique. Then, we assessed how these SNPs correlated with AD susceptibility. Results The results showed that rs3764650 of ABCA7 was closely correlated with risen AD morbidity in the allele [P = 0.010, odds ratio (OR) = 1.43, 95% confidence interval (CI) 1.09–1.89], dominant (P = 0.004, OR = 1.71, 95% CI 1.19–2.46), and additive (P = 0.012, OR = 1.42, 95% CI 1.08–1.86) models. However, rs4147929 of ABCA7 was related to higher AD risk in the allele (P = 0.006, OR = 1.45, 95% CI 1.11–1.89), dominant (P = 0.012, OR = 1.59, 95% CI 1.11–2.27), and additive (P = 0.010, OR = 1.40, 95% CI 1.08–1.81) models. In addition, the frequencies of the G-allele at rs3764650 (P = 0.030) and the A-allele at rs4147929 (P = 0.001) in AD were statistically higher in APOE ε4 carriers in comparison to non-carriers. Conclusion This study demonstrated that the G-allele at rs3764650 and the A-allele at rs4147929 appeared at higher risk for developing AD, particularly in APOE ε4 carriers. Moreover, it was observed that rs3764650 and rs4147929 of ABCA7 were linked to AD. More in-depth research with a relatively large sample is needed to make the results more convincing.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Jiao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aonan Zhao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomeng Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanyu Ye
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichi Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulei Deng
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Wei Xu,
| | - Jun Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Jun Liu,
| |
Collapse
|
19
|
Genome-Wide Association Study of Fluorescent Oxidation Products Accounting for Tobacco Smoking Status in Adults from the French EGEA Study. Antioxidants (Basel) 2022; 11:antiox11050802. [PMID: 35624665 PMCID: PMC9137810 DOI: 10.3390/antiox11050802] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) is the main pathophysiological mechanism involved in several chronic diseases, including asthma. Fluorescent oxidation products (FlOPs), a global biomarker of damage due to OS, is of growing interest in epidemiological studies. We conducted a genome-wide association study (GWAS) of the FlOPs level in 1216 adults from the case-control and family-based EGEA study (mean age 43 years old, 51% women, and 23% current smokers) to identify genetic variants associated with FlOPs. The GWAS was first conducted in the whole sample and then stratified according to smoking status, the main exogenous source of reactive oxygen species. Among the top genetic variants identified by the three GWAS, those located in BMP6 (p = 3 × 10−6), near BMPER (p = 9 × 10−6), in GABRG3 (p = 4 × 10−7), and near ATG5 (p = 2 × 10−9) are the most relevant because of both their link to biological pathways related to OS and their association with several chronic diseases for which the role of OS in their pathophysiology has been pointed out. BMP6 and BMPER are of particular interest due to their involvement in the same biological pathways related to OS and their functional interaction. To conclude, this study, which is the first GWAS of FlOPs, provides new insights into the pathophysiology of chronic OS-related diseases.
Collapse
|
20
|
Dugan AJ, Nelson PT, Katsumata Y, Shade LMP, Teylan MA, Boehme KL, Mukherjee S, Kauwe JSK, Hohman TJ, Schneider JA, Fardo DW. Association between WWOX/MAF variants and dementia-related neuropathologic endophenotypes. Neurobiol Aging 2022; 111:95-106. [PMID: 34852950 PMCID: PMC8761217 DOI: 10.1016/j.neurobiolaging.2021.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/30/2021] [Accepted: 10/22/2021] [Indexed: 11/29/2022]
Abstract
The genetic locus containing the WWOX and MAF genes was implicated as a clinical Alzheimer's disease (AD) risk locus in two recent large meta-analytic genome wide association studies (GWAS). In a prior GWAS, we identified a variant in WWOX as a suggestive risk allele for hippocampal sclerosis. We hypothesized that the WWOX/MAF locus may be preferentially associated with non-plaque- and non-tau-related neuropathological changes (NC). Data from research participants with GWAS and autopsy measures from the National Alzheimer's Coordinating Center and the Religious Orders Study and the Rush Memory and Aging Project were meta-analyzed. Notably, no variants in the locus were significantly associated with ADNC. However, several WWOX/MAF variants had significant adjusted associations with limbic-predominant age-related TDP-43 encephalopathy NC (LATE-NC), HS, and brain arteriolosclerosis. These associations remained largely unchanged after adjustment for ADNC (operationalized with standard semiquantitative staging), suggesting that these associations are independent of ADNC. Thus, WWOX genetic variants were associated pathologically with LATE-NC, not ADNC.
Collapse
Affiliation(s)
- Adam J Dugan
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA; Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| | - Yuriko Katsumata
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA
| | - Lincoln M P Shade
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Merilee A Teylan
- National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kevin L Boehme
- Department of Biology, Brigham Young University, Provo, UT, USA
| | | | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT, USA; Department of Medicine, University of Washington, Seattle, WA, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julie A Schneider
- Departments of Neurology and Pathology, Rush University Medical Center, Chicago, IL, USA
| | - David W Fardo
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging and Alzheimer's Disease Research Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
21
|
Prater KE, Latimer CS, Jayadev S. Glial TDP-43 and TDP-43 induced glial pathology, focus on neurodegenerative proteinopathy syndromes. Glia 2022; 70:239-255. [PMID: 34558120 PMCID: PMC8722378 DOI: 10.1002/glia.24096] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/21/2021] [Accepted: 09/09/2021] [Indexed: 02/03/2023]
Abstract
Since its discovery in 2006, TAR DNA binding protein 43 (TDP-43) has driven rapidly evolving research in neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), and limbic predominant age-related TDP-43 encephalopathy (LATE). TDP-43 mislocalization or aggregation is the hallmark of TDP-43 proteinopathy and is associated with cognitive impairment that can be mapped to its regional deposition. Studies in human tissue and model systems demonstrate that TDP-43 may potentiate other proteinopathies such as the amyloid or tau pathology seen in Alzheimer's Disease (AD) in the combination of AD+LATE. Despite this growing body of literature, there remain gaps in our understanding of whether there is heterogeneity in TDP-43 driven mechanisms across cell types. The growing observations of correlation between TDP-43 proteinopathy and glial pathology suggest a relationship between the two, including pathogenic glial cell-autonomous dysfunction and dysregulated glial immune responses to neuronal TDP-43. In this review, we discuss the available data on TDP-43 in glia within the context of the neurodegenerative diseases ALS and FTLD and highlight the current lack of information about glial TDP-43 interaction in AD+LATE. TDP-43 has proven to be a significant modulator of cognitive and neuropathological outcomes. A deeper understanding of its role in diverse cell types may provide relevant insights into neurodegenerative syndromes.
Collapse
Affiliation(s)
| | - Caitlin S. Latimer
- Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA 98195
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA 98195,Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA 98195
| |
Collapse
|
22
|
Cacabelos R, Naidoo V, Martínez-Iglesias O, Corzo L, Cacabelos N, Pego R, Carril JC. Pharmacogenomics of Alzheimer's Disease: Novel Strategies for Drug Utilization and Development. Methods Mol Biol 2022; 2547:275-387. [PMID: 36068470 DOI: 10.1007/978-1-0716-2573-6_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease (AD) is a priority health problem in developed countries with a high cost to society. Approximately 20% of direct costs are associated with pharmacological treatment. Over 90% of patients require multifactorial treatments, with risk of adverse drug reactions (ADRs) and drug-drug interactions (DDIs) for the treatment of concomitant diseases such as hypertension (>25%), obesity (>70%), diabetes mellitus type 2 (>25%), hypercholesterolemia (40%), hypertriglyceridemia (20%), metabolic syndrome (20%), hepatobiliary disorder (15%), endocrine/metabolic disorders (>20%), cardiovascular disorder (40%), cerebrovascular disorder (60-90%), neuropsychiatric disorders (60-90%), and cancer (10%).For the past decades, pharmacological studies in search of potential treatments for AD focused on the following categories: neurotransmitter enhancers (11.38%), multitarget drugs (2.45%), anti-amyloid agents (13.30%), anti-tau agents (2.03%), natural products and derivatives (25.58%), novel synthetic drugs (8.13%), novel targets (5.66%), repository drugs (11.77%), anti-inflammatory drugs (1.20%), neuroprotective peptides (1.25%), stem cell therapy (1.85%), nanocarriers/nanotherapeutics (1.52%), and other compounds (<1%).Pharmacogenetic studies have shown that the therapeutic response to drugs in AD is genotype-specific in close association with the gene clusters that constitute the pharmacogenetic machinery (pathogenic, mechanistic, metabolic, transporter, pleiotropic genes) under the regulatory control of epigenetic mechanisms (DNA methylation, histone/chromatin remodeling, microRNA regulation). Most AD patients (>60%) are carriers of over ten pathogenic genes. The genes that most frequently (>50%) accumulate pathogenic variants in the same AD case are A2M (54.38%), ACE (78.94%), BIN1 (57.89%), CLU (63.15%), CPZ (63.15%), LHFPL6 (52.63%), MS4A4E (50.87%), MS4A6A (63.15%), PICALM (54.38%), PRNP (80.7059), and PSEN1 (77.19%). There is also an accumulation of 15 to 26 defective pharmagenes in approximately 85% of AD patients. About 50% of AD patients are carriers of at least 20 mutant pharmagenes, and over 80% are deficient metabolizers for the most common drugs, which are metabolized via the CYP2D6, CYP2C9, CYP2C19, and CYP3A4/5 enzymes.The implementation of pharmacogenetics can help optimize drug development and the limited therapeutic resources available to treat AD, and personalize the use of anti-dementia drugs in combination with other medications for the treatment of concomitant disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain.
| | - Vinogran Naidoo
- Department of Neuroscience, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Olaia Martínez-Iglesias
- Department of Medical Epigenetics, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Lola Corzo
- Department of Medical Biochemistry, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Natalia Cacabelos
- Department of Medical Documentation, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Rocío Pego
- Department of Neuropsychology, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| | - Juan C Carril
- Department of Genomics and Pharmacogenomics, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Corunna, Spain
| |
Collapse
|
23
|
Agrawal S, Yu L, Nag S, Arfanakis K, Barnes LL, Bennett DA, Schneider JA. The association of Lewy bodies with limbic-predominant age-related TDP-43 encephalopathy neuropathologic changes and their role in cognition and Alzheimer's dementia in older persons. Acta Neuropathol Commun 2021; 9:156. [PMID: 34563269 PMCID: PMC8466680 DOI: 10.1186/s40478-021-01260-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Lewy bodies (LBs) and limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) are common in older persons and associated with cognitive impairment. However, little is known about the relationship between LBs and LATE-NC and their combined roles in cognitive impairment and Alzheimer's dementia in community-dwelling participants. The study included 1670 community-based participants (mean age-at-death, 89.5 years (SD = 6.65); 69% females) who underwent annual assessments of cognition to create summary measures of global cognition and cognitive domains and evaluation for Alzheimer's dementia. Systematic neuropathologic evaluations were performed to assess LBs, LATE-NC, and Alzheimer's disease (AD) pathology. We excluded cases with pathologically confirmed frontotemporal lobar degeneration in this study. Logistic and linear regression analyses were used, adjusted for demographics and AD pathology. LBs were present in 428 (25.6%) decedents (29 nigra-predominant, 165 limbic-type, and 234 neocortical-type) while 865 (51.7%) decedents exhibited LATE-NC (307 stage 1, 167 stage 2, and 391 stage 3). LBs combined with LATE-NC were common (15% of all participants) and in those with Alzheimer's dementia (25%). Neocortical-type, but not nigral-predominant or limbic-type LBs increased the odds of stage 2/3 LATE-NC (odds ratio = 1.70; 95% confidence interval = 1.26-2.30). The association between neocortical-type LBs and stage 2/3 LATE-NC was stronger in those under 90 years of age and in women. In analyses of cognition and Alzheimer's dementia, LATE-NC and neocortical-type LBs, separately, were related to lower global cognition, five specific cognitive domains, and an increased odds of Alzheimer's dementia, above and beyond the AD pathology. Limbic-type LBs were related to lower global cognition, and the domains of episodic, working, and semantic memory, and increased odds of Alzheimer's dementia. Furthermore, there was no interaction between limbic/neocortical-type LBs and LATE-NC on cognitive function, cognitive domains, or Alzheimer's dementia. These findings suggest that neocortical-type LBs are associated with LATE-NC, specifically in the younger old and in women. Limbic/neocortical-type LBs and LATE-NC have separate and additive effects on cognitive function and odds of Alzheimer's dementia.
Collapse
Affiliation(s)
- Sonal Agrawal
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison street, Suite 1000, Chicago, IL, 60612, USA.
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA.
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison street, Suite 1000, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sukriti Nag
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison street, Suite 1000, Chicago, IL, 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison street, Suite 1000, Chicago, IL, 60612, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison street, Suite 1000, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison street, Suite 1000, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W. Harrison street, Suite 1000, Chicago, IL, 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
24
|
Dugan AJ, Nelson PT, Katsumata Y, Shade LMP, Boehme KL, Teylan MA, Cykowski MD, Mukherjee S, Kauwe JSK, Hohman TJ, Schneider JA, Fardo DW. Analysis of genes (TMEM106B, GRN, ABCC9, KCNMB2, and APOE) implicated in risk for LATE-NC and hippocampal sclerosis provides pathogenetic insights: a retrospective genetic association study. Acta Neuropathol Commun 2021; 9:152. [PMID: 34526147 PMCID: PMC8442328 DOI: 10.1186/s40478-021-01250-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is the most prevalent subtype of TDP-43 proteinopathy, affecting up to 1/3rd of aged persons. LATE-NC often co-occurs with hippocampal sclerosis (HS) pathology. It is currently unknown why some individuals with LATE-NC develop HS while others do not, but genetics may play a role. Previous studies found associations between LATE-NC phenotypes and specific genes: TMEM106B, GRN, ABCC9, KCNMB2, and APOE. Data from research participants with genomic and autopsy measures from the National Alzheimer’s Coordinating Center (NACC; n = 631 subjects included) and the Religious Orders Study and Memory and the Rush Aging Project (ROSMAP; n = 780 included) were analyzed in the current study. Our goals were to reevaluate disease-associated genetic variants using newly collected data and to query whether the specific genotype/phenotype associations could provide new insights into disease-driving pathways. Research subjects included in prior LATE/HS genome-wide association studies (GWAS) were excluded. Single nucleotide variants (SNVs) within 10 kb of TMEM106B, GRN, ABCC9, KCNMB2, and APOE were tested for association with HS and LATE-NC, and separately for Alzheimer’s pathologies, i.e. amyloid plaques and neurofibrillary tangles. Significantly associated SNVs were identified. When results were meta-analyzed, TMEM106B, GRN, and APOE had significant gene-based associations with both LATE and HS, whereas ABCC9 had significant associations with HS only. In a sensitivity analysis limited to LATE-NC + cases, ABCC9 variants were again associated with HS. By contrast, the associations of TMEM106B, GRN, and APOE with HS were attenuated when adjusting for TDP-43 proteinopathy, indicating that these genes may be associated primarily with TDP-43 proteinopathy. None of these genes except APOE appeared to be associated with Alzheimer’s-type pathology. In summary, using data not included in prior studies of LATE or HS genomics, we replicated several previously reported gene-based associations and found novel evidence that specific risk alleles can differentially affect LATE-NC and HS.
Collapse
|
25
|
Oveisgharan S, Capuano AW, Nag S, Agrawal S, Barnes LL, Bennett DA, Arvanitakis Z, Schneider JA. Association of Hemoglobin A1C With TDP-43 Pathology in Community-Based Elders. Neurology 2021; 96:e2694-e2703. [PMID: 33853892 PMCID: PMC8205465 DOI: 10.1212/wnl.0000000000012025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/03/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test the hypothesis that an inverse association exists between diabetes mellitus (DM) and hemoglobin A1C (A1C) with transactive response DNA binding protein 43 (TDP-43) levels in older adults. METHODS We leveraged antemortem and postmortem data of decedents from 3 community-based clinical-pathologic studies. DM status, A1C levels, and medications for DM were documented annually. TDP-43 cytoplasmic inclusions, evaluated in 6 brain regions using immunohistochemistry, were used to obtain a semiquantitative TDP-43 score (0-5) in each region, and scores were averaged across regions to obtain a TDP-43 severity score. We used linear regressions to test the association of DM and A1C with the TDP-43 severity score. RESULTS On average, participants (n = 817) were 90 years old at the time of death, three-fourths were women, and one-fourth had DM. The mean A1C was 6.0% (SD 0.6). TDP-43 was observed in 54% of participants, and the mean TDP-43 score was 0.7 (range 0-4.5). A higher level of A1C was associated with a lower TDP-43 score (estimate -0.156, SE 0.060, p = 0.009), while DM had a borderline inverse association with the TDP-43 score (estimate -0.163, SE 0.087, p = 0.060). The association of higher levels of A1C with lower TDP-43 scores persisted after further adjustment by APOE ε4, vascular risk factors, stroke, and hypoglycemic medications. Exclusion of the oldest old participants did not change the results. CONCLUSION Overall, the results suggest that a high level of A1C is associated with less TDP-43 proteinopathy in older persons while the relationship of DM with TDP-43 needs further study.
Collapse
Affiliation(s)
- Shahram Oveisgharan
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL.
| | - Ana W Capuano
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL
| | - Sukriti Nag
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL
| | - Sonal Agrawal
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL
| | - Lisa L Barnes
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL
| | - David A Bennett
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL
| | - Zoe Arvanitakis
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL
| | - Julie A Schneider
- From the Rush Alzheimer's Disease Center (S.O., A.W.C., S.N., S.A., L.L.B., D.A.B., Z.A., J.A.S.) and Departments of Neurological Sciences (S.O., A.W.C., L.L.B., D.A.B., Z.A., J.A.S.), Pathology (S.N., S.A., J.A.S.), and Behavioral Sciences (L.L.B.), Rush University Medical Center, Chicago, IL
| |
Collapse
|
26
|
Zhang L, Huang P, Huang C, Jiang L, Lu Z, Wang P. Varied clinical significance of ATP-binding cassette C sub-family members for lung adenocarcinoma. Medicine (Baltimore) 2021; 100:e25246. [PMID: 33879658 PMCID: PMC8078454 DOI: 10.1097/md.0000000000025246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 03/01/2021] [Indexed: 12/25/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is a lethal malignancy worldwide and a major public health concern. We explored the potential clinical significance for LUAD of ATP-binding cassette (ABC), sub-family C, consisting of ABCC1-6, 8-12, and cystic fibrosis transmembrane conductance regulator (CFTR).Five hundred LUAD patients from The Cancer Genome Atlas database were used for analysis, including differential expression and diagnostic and prognostic significance. Oncomine and MERAV databases were used to validate differential expression and diagnostic significance. A risk score model was constructed using prognosis-related ABCC members. Prognosis-related genes were further explored to correlate their expression with tumor stage progression. Interaction networks, including biological processes and metabolic pathways, were constructed using Cytoscape software and STRING website.ABCC1-3 consistently showed high expression in tumor tissues (all P ≤ 0.05). Most datasets indicated that ABCC5, 10, and 11 were highly expressed in tumor tissues whereas ABCC6, 9, and CFTR were highly expressed in nontumor tissues (all P ≤ 0.05). Diagnostic significance of ABCC3 and ABCC5 was consistently assessed and validated in three datasets (all area under the curve > 0.700) whereas ABCC6, 8, 10, 11, and CFTR were assessed in The Cancer Genome Atlas dataset and validated in one dataset (all area under the curve > 0.700). Prognostic analysis indicated that ABCC2, 6, and 8 mRNA expression was associated with survival of LUAD (all adjusted P ≤ .037). The risk score model constructed using ABCC2, 6, and 8 suggested prognostic significance for survival predictions. ABCC2 expression was associated with tumor stage, whereas ABCC6 and 8 were not. Interaction networks indicated that they were involved in establishment of localization, ion transport, plasma membrane, apical plasma membrane, adenylyl nucleotide binding, ABC transporters, ABC transporter disorders, ABC-family-protein-mediated transport, and bile secretion.Differentially expressed ABCC2 and ABCC5 might be diagnostic whereas ABCC2, 6, and 8 may be prognostic biomarkers for LUAD, possibly through ABC-family-mediated transporter disorders.
Collapse
|
27
|
Liu X, Song Z, Li Y, Yao Y, Fang M, Bai C, An P, Chen H, Chen Z, Tang B, Shen J, Gao X, Zhang M, Chen P, Zhang T, Jia H, Liu X, Hou Y, Yang H, Wang J, Wang F, Xu X, Min J, Nie C, Zeng Y. Integrated genetic analyses revealed novel human longevity loci and reduced risks of multiple diseases in a cohort study of 15,651 Chinese individuals. Aging Cell 2021; 20:e13323. [PMID: 33657282 PMCID: PMC7963337 DOI: 10.1111/acel.13323] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 01/16/2021] [Accepted: 01/23/2021] [Indexed: 12/14/2022] Open
Abstract
There is growing interest in studying the genetic contributions to longevity, but limited relevant genes have been identified. In this study, we performed a genetic association study of longevity in a total of 15,651 Chinese individuals. Novel longevity loci, BMPER (rs17169634; p = 7.91 × 10-15 ) and TMEM43/XPC (rs1043943; p = 3.59 × 10-8 ), were identified in a case-control analysis of 11,045 individuals. BRAF (rs1267601; p = 8.33 × 10-15 ) and BMPER (rs17169634; p = 1.45 × 10-10 ) were significantly associated with life expectancy in 12,664 individuals who had survival status records. Additional sex-stratified analyses identified sex-specific longevity genes. Notably, sex-differential associations were identified in two linkage disequilibrium blocks in the TOMM40/APOE region, indicating potential differences during meiosis between males and females. Moreover, polygenic risk scores and Mendelian randomization analyses revealed that longevity was genetically causally correlated with reduced risks of multiple diseases, such as type 2 diabetes, cardiovascular diseases, and arthritis. Finally, we incorporated genetic markers, disease status, and lifestyles to classify longevity or not-longevity groups and predict life span. Our predictive models showed good performance (AUC = 0.86 for longevity classification and explained 19.8% variance of life span) and presented a greater predictive efficiency in females than in males. Taken together, our findings not only shed light on the genetic contributions to longevity but also elucidate correlations between diseases and longevity.
Collapse
Affiliation(s)
- Xiaomin Liu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
- BGI Education Center University of Chinese Academy of Sciences Shenzhen China
| | - Zijun Song
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Yan Li
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yao Yao
- Center for the Study of Aging and Human Development Medical School of Duke University Durham USA
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
| | - Mingyan Fang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Chen Bai
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
- School of Labor and Human Resources Renmin University Beijing China
| | - Peng An
- Beijing Advanced Innovation Center for Food Nutrition and Human Health China Agricultural University Beijing China
| | - Huashuai Chen
- Business School of Xiangtan University Xiangtan China
| | - Zhihua Chen
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Biyao Tang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Juan Shen
- BGI Genomics BGI‐Shenzhen Shenzhen China
| | - Xiaotong Gao
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | | | - Pengyu Chen
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Tao Zhang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Huijue Jia
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Xiao Liu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yong Hou
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Huanming Yang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Jian Wang
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Fudi Wang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health China Agricultural University Beijing China
| | - Xun Xu
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
- Guangdong Provincial Key Laboratory of Genome Read and Write Shenzhen China
| | - Junxia Min
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine, Zhejiang University Hangzhou China
| | - Chao Nie
- BGI‐Shenzhen Shenzhen China
- China National Genebank Shenzhen China
| | - Yi Zeng
- Center for the Study of Aging and Human Development Medical School of Duke University Durham USA
- Center for Healthy Aging and Development Studies National School of Development, Raissun Institute for Advanced Studies, Peking University Beijing China
| |
Collapse
|
28
|
Ruffini N, Klingenberg S, Schweiger S, Gerber S. Common Factors in Neurodegeneration: A Meta-Study Revealing Shared Patterns on a Multi-Omics Scale. Cells 2020; 9:E2642. [PMID: 33302607 PMCID: PMC7764447 DOI: 10.3390/cells9122642] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are heterogeneous, progressive diseases with frequently overlapping symptoms characterized by a loss of neurons. Studies have suggested relations between neurodegenerative diseases for many years (e.g., regarding the aggregation of toxic proteins or triggering endogenous cell death pathways). We gathered publicly available genomic, transcriptomic, and proteomic data from 177 studies and more than one million patients to detect shared genetic patterns between the neurodegenerative diseases on three analyzed omics-layers. The results show a remarkably high number of shared differentially expressed genes between the transcriptomic and proteomic levels for all conditions, while showing a significant relation between genomic and proteomic data between AD and PD and AD and ALS. We identified a set of 139 genes being differentially expressed in several transcriptomic experiments of all four diseases. These 139 genes showed overrepresented gene ontology (GO) Terms involved in the development of neurodegeneration, such as response to heat and hypoxia, positive regulation of cytokines and angiogenesis, and RNA catabolic process. Furthermore, the four analyzed neurodegenerative diseases (NDDs) were clustered by their mean direction of regulation throughout all transcriptomic studies for this set of 139 genes, with the closest relation regarding this common gene set seen between AD and HD. GO-Term and pathway analysis of the proteomic overlap led to biological processes (BPs), related to protein folding and humoral immune response. Taken together, we could confirm the existence of many relations between Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis on transcriptomic and proteomic levels by analyzing the pathways and GO-Terms arising in these intersections. The significance of the connection and the striking relation of the results to processes leading to neurodegeneration between the transcriptomic and proteomic data for all four analyzed neurodegenerative diseases showed that exploring many studies simultaneously, including multiple omics-layers of different neurodegenerative diseases simultaneously, holds new relevant insights that do not emerge from analyzing these data separately. Furthermore, the results shed light on processes like the humoral immune response that have previously been described only for certain diseases. Our data therefore suggest human patients with neurodegenerative diseases should be addressed as complex biological systems by integrating multiple underlying data sources.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
- Leibniz Institute for Resilience Research, Leibniz Association, Wallstraße 7, 55122 Mainz, Germany
| | - Susanne Klingenberg
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susann Schweiger
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susanne Gerber
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| |
Collapse
|
29
|
Stroobants S, D'Hooge R, Damme M. Aged Tmem106b knockout mice display gait deficits in coincidence with Purkinje cell loss and only limited signs of non-motor dysfunction. Brain Pathol 2020; 31:223-238. [PMID: 33016371 PMCID: PMC8018119 DOI: 10.1111/bpa.12903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/14/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Genetic variants in TMEM106B are a major risk factor for several neurodegenerative diseases including frontotemporal degeneration, limbic‐predominant age‐related TDP‐43 encephalopathy, Parkinson's disease, late‐onset‐Alzheimer's disease and constitute a genetic determinant of differential aging. TMEM106B encodes an integral lysosomal membrane protein but its precise physiological function in the central nervous system remains enigmatic. Presently, we aimed to increase understanding of TMEM106B contribution to general brain function and aging. We analyzed an aged cohort of Tmem106b knockout‐, heterozygote and wild‐type mice in a behavioral test battery including assessments of motor function as well as, social, emotional and cognitive function. Aged Tmem106b knockout (KO) mice displayed diverse behavioral deficits including motor impairment, gait defects and reduced startle reactivity. In contrast, no prominent deficits were observed in social, emotional or cognitive behaviors. Histologically, we observed late‐onset loss of Purkinje cells followed by reactive gliosis in the cerebellum, which likely contributed to progressive decline in motor function and gait defects in particular. Reactive gliosis was not restricted to the cerebellum but observed in different areas of the brain including the brain stem and parts of the cerebral cortex. Surviving Purkinje cells showed vacuolated lysosomes in the axon initial segment, implicating TMEM106B‐dependent lysosomal trafficking defects as the underlying cause of axonal and more general neuronal dysfunction contributing to behavioral impairments. Our experiments help to elucidate how TMEM106B affects spatial neuronal homeostasis and exemplifies a critical role of TMEM106B in neuronal cells for survival.
Collapse
Affiliation(s)
- Stijn Stroobants
- Laboratory of Biological Psychology, KU Leuven, Tiensestraat 102, Leuven, 3000, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, KU Leuven, Tiensestraat 102, Leuven, 3000, Belgium
| | - Markus Damme
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, 24098, Germany
| |
Collapse
|
30
|
Katsumata Y, Abner EL, Karanth S, Teylan MA, Mock CN, Cykowski MD, Lee EB, Boehme KL, Mukherjee S, Kauwe JSK, Kryscio RJ, Schmitt FA, Fardo DW, Nelson PT. Distinct clinicopathologic clusters of persons with TDP-43 proteinopathy. Acta Neuropathol 2020; 140:659-674. [PMID: 32797255 PMCID: PMC7572241 DOI: 10.1007/s00401-020-02211-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022]
Abstract
To better understand clinical and neuropathological features of TDP-43 proteinopathies, data were analyzed from autopsied research volunteers who were followed in the National Alzheimer's Coordinating Center (NACC) data set. All subjects (n = 495) had autopsy-proven TDP-43 proteinopathy as an inclusion criterion. Subjects underwent comprehensive longitudinal clinical evaluations yearly for 6.9 years before death on average. We tested whether an unsupervised clustering algorithm could detect coherent groups of TDP-43 immunopositive cases based on age at death and extensive neuropathologic data. Although many of the brains had mixed pathologies, four discernible clusters were identified. Key differentiating features were age at death and the severity of comorbid Alzheimer's disease neuropathologic changes (ADNC), particularly neuritic amyloid plaque densities. Cluster 1 contained mostly cases with a pathologic diagnosis of frontotemporal lobar degeneration (FTLD-TDP), consistent with enrichment of frontotemporal dementia clinical phenotypes including appetite/eating problems, disinhibition and primary progressive aphasia (PPA). Cluster 2 consisted of elderly limbic-predominant age-related TDP-43 encephalopathy (LATE-NC) subjects without severe neuritic amyloid plaques. Subjects in Cluster 2 had a relatively slow cognitive decline. Subjects in both Clusters 3 and 4 had severe ADNC + LATE-NC; however, Cluster 4 was distinguished by earlier disease onset, swifter disease course, more Lewy body pathology, less neocortical TDP-43 proteinopathy, and a suggestive trend in a subgroup analysis (n = 114) for increased C9orf72 risk SNP rs3849942 T allele (Fisher's exact test p value = 0.095). Overall, clusters enriched with neocortical TDP-43 proteinopathy (Clusters 1 and 2) tended to have lower levels of neuritic amyloid plaques, and those dying older (Clusters 2 and 3) had far less PPA or disinhibition, but more apathy. Indeed, 98% of subjects dying past age 85 years lacked clinical features of the frontotemporal dementia syndrome. Our study revealed discernible subtypes of LATE-NC and underscored the importance of age of death for differentiating FTLD-TDP and LATE-NC.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY, 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Department of Epidemiology, University of Kentucky, Lexington, KY, 40536, USA
| | - Shama Karanth
- Department of Epidemiology, University of Kentucky, Lexington, KY, 40536, USA
| | - Merilee A Teylan
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, 98105, USA
| | - Charles N Mock
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, 98105, USA
- Department of Epidemiology, University of Washington, Seattle, WA, 98105, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kevin L Boehme
- Department of Biology, Brigham Young University, Provo, UT, 84602, USA
| | | | - John S K Kauwe
- Brigham Young University-Hawaii, Laie, HI, 96762, USA
- Biology Department, Brigham Young University, Provo, UT, 84602, USA
| | - Richard J Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Department of Statistics, University of Kentucky, Lexington, KY, 40536, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY, 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Pathology, University of Kentucky, Rm 311 Sanders-Brown Center on Aging, 800 S. Limestone Avenue, Lexington, KY, 40536, USA.
| |
Collapse
|
31
|
Blue EE, Cheng A, Chen S, Yu CE. Association of Uncommon, Noncoding Variants in the APOE Region With Risk of Alzheimer Disease in Adults of European Ancestry. JAMA Netw Open 2020; 3:e2017666. [PMID: 33090224 PMCID: PMC7582128 DOI: 10.1001/jamanetworkopen.2020.17666] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPORTANCE The ε2 and ε4 alleles of the apolipoprotein E (APOE) gene are associated with Alzheimer disease (AD) risk. Although nearby genetic variants have also been shown to be associated with AD, including rs2075650 in the TOMM40 gene and rs4420638 near the APOC1 gene, it is unknown whether these associations are independent of the ε2 and ε4 alleles. OBJECTIVE To assess whether variants near APOE are associated with AD independently of the ε2/ε3/ε4 genotype. DESIGN, SETTING, AND PARTICIPANTS In this genetic association study of the Alzheimer's Disease Genetics Consortium imputed genotype at data, 14 415 variants near APOE (±500 kilobase) for 18 795 individuals with European ancestry were tested for association with AD using 4 logistic mixed models adjusting for sex, cohort, population structure, and relatedness. Model 1 had no APOE adjustment, and model 2 adjusted for the count of ε2 and ε4 alleles. Model 3 was restricted to ε3 homozygotes, and model 4 was restricted to ε4 homozygotes. Data were downloaded from May 31, 2018, to June 3, 2018, and analyzed from November 1, 2018, to June 24, 2020. MAIN OUTCOMES AND MEASURES Alzheimer disease affectation status was defined by clinicians using standard National Institute of Neurological and Communicative Disorders and Stroke and Alzheimer Disease and Related Disorders Association criteria. Association was evaluated using Score tests; results with P < .05 divided by the number of independent tests per model were considered statistically significant. RESULTS Among the 18 795 individuals in the study, 9704 were affected by AD and 9066 were control individuals; the median age at onset/evaluation was 76 (interquartile range, 70-82) years; and 11 167 were female (59.4%). Associations with AD were found for rs2075650 (odds ratio [OR], 2.59; 95% CI, 2.45-2.75; P = 3.19 × 10-228) and rs4420638 (OR, 2.77; 95% CI, 2.62-2.94; P = 2.99 × 10-254) without APOE adjustment. Although rs2075650 was nominally associated with AD among the ε4 homozygotes (OR, 1.33; 95% CI, 1.00-1.77; P = .047), the association between rs4420638 and AD was eliminated by APOE adjustment (model 2 OR, 1.06 [95% CI, 0.96-1.18; P = .24]; model 3 OR, 1.13 [95% CI, 0.95-1.34; P = .18]; model 4 OR, 0.90 [95% CI, 0.56-1.45; P = .66]). There was a significant association between rs192879175 and AD among ε3 homozygotes (OR, 0.50; 95% CI, 0.37-0.68; P = 8.30 × 10-6). CONCLUSIONS AND RELEVANCE The results of this genetic association study suggest that ε2/ε3/ε4 alleles are not the only variants in the APOE region that are associated with AD risk. Additional work with independent data is needed to replicate these results.
Collapse
Affiliation(s)
- Elizabeth E. Blue
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle
| | - Anqi Cheng
- Department of Biostatistics, University of Washington, Seattle
| | - Sunny Chen
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Chang-En Yu
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle
| |
Collapse
|
32
|
Nolan M, Scott C, Gamarallage MP, Lunn D, Carpenter K, McDonough E, Meyer D, Kaanumalle S, Santamaria-Pang A, Turner MR, Talbot K, Ansorge O. Quantitative patterns of motor cortex proteinopathy across ALS genotypes. Acta Neuropathol Commun 2020; 8:98. [PMID: 32616036 PMCID: PMC7331195 DOI: 10.1186/s40478-020-00961-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Degeneration of the primary motor cortex is a defining feature of amyotrophic lateral sclerosis (ALS), which is associated with the accumulation of microscopic protein aggregates in neurons and glia. However, little is known about the quantitative burden and pattern of motor cortex proteinopathies across ALS genotypes. We combined quantitative digital image analysis with multi-level generalized linear modelling in an independent cohort of 82 ALS cases to explore the relationship between genotype, total proteinopathy load and cellular vulnerability to aggregate formation. Primary motor cortex phosphorylated (p)TDP-43 burden and microglial activation were more severe in sporadic ALS-TDP disease than C9-ALS. Oligodendroglial pTDP-43 pathology was a defining feature of ALS-TDP in sporadic ALS, C9-ALS and ALS with OPTN, HNRNPA1 or TARDBP mutations. ALS-FUS and ALS-SOD1 showed less cortical proteinopathy in relation to spinal cord pathology than ALS-TDP, where pathology was more evenly spread across the motor cortex-spinal cord axis. Neuronal pTDP-43 aggregates were rare in GAD67+ and Parvalbumin+ inhibitory interneurons, consistent with predominant accumulation in excitatory neurons. Finally, we show that cortical microglia, but not astrocytes, contain pTDP-43. Our findings suggest divergent quantitative, genotype-specific vulnerability of the ALS primary motor cortex to proteinopathies, which may have implications for our understanding of disease pathogenesis and the development of genotype-specific therapies.
Collapse
|
33
|
Jicha GA, Nelson PT. Hippocampal Sclerosis, Argyrophilic Grain Disease, and Primary Age-Related Tauopathy. Continuum (Minneap Minn) 2020; 25:208-233. [PMID: 30707194 DOI: 10.1212/con.0000000000000697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Hippocampal sclerosis, argyrophilic grain disease, and primary age-related tauopathy are common Alzheimer disease mimics that currently lack clinical diagnostic criteria. Increased understanding of these pathologic entities is important for the neurologist who may encounter patients with an unusually slowly progressive degenerative dementia that may appear to meet criteria for Alzheimer disease but who progress to develop symptoms that are unusual for classic Alzheimer disease RECENT FINDINGS: Hippocampal sclerosis has traditionally been associated with hypoxic/ischemic injury and poorly controlled epilepsy, but it is now recognized that hippocampal sclerosis may also be associated with a unique degenerative disease of aging or may be an associated pathologic finding in many cases of frontotemporal lobar degeneration. Argyrophilic grain disease has been recognized as an enigma in the field of pathology for over 30 years, but recent discoveries suggest that it may overlap with other tau-related disorders within the spectrum of frontotemporal lobar degeneration. Primary age-related tauopathy has long been recognized as a distinct clinical entity that lies on the Alzheimer pathologic spectrum, with the presence of neurofibrillary tangles that lack the coexistent Alzheimer plaque development; thus, it is thought to represent a distinct pathologic entity. SUMMARY Despite advances in dementia diagnosis that suggest that we have identified and unlocked the mysteries of the major degenerative disease states responsible for cognitive decline and dementia in the elderly, diseases such as hippocampal sclerosis, argyrophilic grain disease, and primary age-related tauopathy demonstrate that we remain on the frontier of discovery and that our diagnostic repertoire of diseases responsible for such clinical symptoms remains in its infancy. Understanding such diagnostic confounds is important for the neurologist in assigning appropriate diagnoses and selecting appropriate therapeutic management strategies for patients with mild cognitive impairment and dementia.
Collapse
|
34
|
Smeland MF, McClenaghan C, Roessler HI, Savelberg S, Hansen GÅM, Hjellnes H, Arntzen KA, Müller KI, Dybesland AR, Harter T, Sala-Rabanal M, Emfinger CH, Huang Y, Singareddy SS, Gunn J, Wozniak DF, Kovacs A, Massink M, Tessadori F, Kamel SM, Bakkers J, Remedi MS, Van Ghelue M, Nichols CG, van Haaften G. ABCC9-related Intellectual disability Myopathy Syndrome is a K ATP channelopathy with loss-of-function mutations in ABCC9. Nat Commun 2019; 10:4457. [PMID: 31575858 PMCID: PMC6773855 DOI: 10.1038/s41467-019-12428-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/30/2019] [Indexed: 11/30/2022] Open
Abstract
Mutations in genes encoding KATP channel subunits have been reported for pancreatic disorders and Cantú syndrome. Here, we report a syndrome in six patients from two families with a consistent phenotype of mild intellectual disability, similar facies, myopathy, and cerebral white matter hyperintensities, with cardiac systolic dysfunction present in the two oldest patients. Patients are homozygous for a splice-site mutation in ABCC9 (c.1320 + 1 G > A), which encodes the sulfonylurea receptor 2 (SUR2) subunit of KATP channels. This mutation results in an in-frame deletion of exon 8, which results in non-functional KATP channels in recombinant assays. SUR2 loss-of-function causes fatigability and cardiac dysfunction in mice, and reduced activity, cardiac dysfunction and ventricular enlargement in zebrafish. We term this channelopathy resulting from loss-of-function of SUR2-containing KATP channels ABCC9-related Intellectual disability Myopathy Syndrome (AIMS). The phenotype differs from Cantú syndrome, which is caused by gain-of-function ABCC9 mutations, reflecting the opposing consequences of KATP loss- versus gain-of-function.
Collapse
Affiliation(s)
- Marie F Smeland
- Department of Medical Genetics, University Hospital of North Norway, 9019, Tromsø, Norway.
| | - Conor McClenaghan
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
| | - Helen I Roessler
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands
| | - Sanne Savelberg
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands
| | | | - Helene Hjellnes
- Department of Medical Genetics, University Hospital of North Norway, 9019, Tromsø, Norway
| | - Kjell Arne Arntzen
- Department of Neurology, University Hospital of North Norway, 9019, Tromsø, Norway
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019, Tromsø, Norway
- The National Neuromuscular Centre of Norway, University Hospital of North Norway, 9019, Tromsø, Norway
| | - Kai Ivar Müller
- Department of Neurology, University Hospital of North Norway, 9019, Tromsø, Norway
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019, Tromsø, Norway
| | - Andreas Rosenberger Dybesland
- The National Neuromuscular Centre of Norway, University Hospital of North Norway, 9019, Tromsø, Norway
- Department of Physiotherapy, University Hospital of North Norway, 9019, Tromsø, Norway
| | - Theresa Harter
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
| | - Monica Sala-Rabanal
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University, St Louis, MO, 63110, USA
| | - Chris H Emfinger
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
| | - Yan Huang
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Soma S Singareddy
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
| | - Jamie Gunn
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Attila Kovacs
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Maarten Massink
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands
| | - Federico Tessadori
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands
- Hubrecht Institute-KNAW and UMC Utrecht, 3584 CT, Utrecht, the Netherlands
| | - Sarah M Kamel
- Hubrecht Institute-KNAW and UMC Utrecht, 3584 CT, Utrecht, the Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and UMC Utrecht, 3584 CT, Utrecht, the Netherlands
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands
| | - Maria S Remedi
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University, St Louis, MO, 63110, USA
| | - Marijke Van Ghelue
- Department of Medical Genetics, University Hospital of North Norway, 9019, Tromsø, Norway
- Department of Medical Genetics, the Arctic University of Norway, 9019, Tromsø, Norway
| | - Colin G Nichols
- Department of Cell Biology and Physiology, and Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St Louis, MO, 63110, USA
| | - Gijs van Haaften
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
35
|
Hokkanen SRK, Kero M, Kaivola K, Hunter S, Keage HAD, Kiviharju A, Raunio A, Tienari PJ, Paetau A, Matthews FE, Fleming J, Graff C, Polvikoski TM, Myllykangas L, Brayne C. Putative risk alleles for LATE-NC with hippocampal sclerosis in population-representative autopsy cohorts. Brain Pathol 2019; 30:364-372. [PMID: 31376286 PMCID: PMC7065086 DOI: 10.1111/bpa.12773] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Limbic‐predominant age‐related TAR‐DNA‐binding protein‐43 (TDP‐43) encephalopathy with hippocampal sclerosis pathology (LATE‐NC + HS) is a neurodegenerative disorder characterized by severe hippocampal CA1 neuron loss and TDP‐43‐pathology, leading to cognitive dysfunction and dementia. Polymorphisms in GRN, TMEM106B and ABCC9 are proposed as LATE‐NC + HS risk factors in brain bank collections. To replicate these results in independent population‐representative cohorts, hippocampal sections from brains donated to three such studies (Cambridge City over 75‐Cohort [CC75C], Cognitive Function and Ageing Study [CFAS], and Vantaa 85+ Study) were stained with hematoxylin–eosin (n = 744) and anti‐pTDP‐43 (n = 713), and evaluated for LATE‐NC + HS and TDP‐43 pathology. Single nucleotide polymorphism genotypes in GRN rs5848, TMEM106B rs1990622 and ABCC9 rs704178 were determined. LATE‐NC + HS (n = 58) was significantly associated with the GRN rs5848 genotype (χ2(2) = 20.61, P < 0.001) and T‐allele (χ2(1) = 21.04, P < 0.001), and TMEM106B rs1990622 genotype (Fisher's exact test, P < 0.001) and A‐allele (χ2(1) = 25.75, P < 0.001). No differences in ABCC9 rs704178 genotype or allele frequency were found between LATE‐NC + HS and non‐LATE‐NC + HS neuropathology cases. Dentate gyrus TDP‐43 pathology associated with GRN and TMEM106B variations, but the association with TMEM106B nullified when LATE‐NC + HS cases were excluded. Our results indicate that GRN and TMEM106B are associated with severe loss of CA1 neurons in the aging brain, while ABCC9 was not confirmed as a genetic risk factor for LATE‐NC + HS. The association between TMEM106B and LATE‐NC + HS may be independent of dentate TDP‐43 pathology.
Collapse
Affiliation(s)
| | - Mia Kero
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Karri Kaivola
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Sally Hunter
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Hannah A D Keage
- Cognitive Ageing and Impairment Neurosciences Laboratory, School of Psychology, Social Work and Social Policy, University of South Australia, Adelaide, Australia
| | - Anna Kiviharju
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Anna Raunio
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Pentti J Tienari
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Fiona E Matthews
- Institute for Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Jane Fleming
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Caroline Graff
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, J10:20, Visionsgatan 4, Solna, 171 64, Sweden.,Theme Aging, Genetics Unit, Karolinska University Hospital-Solna, QA22, Stockholm, Sweden
| | - Tuomo M Polvikoski
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Liisa Myllykangas
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
36
|
Tau and TDP-43 proteinopathies: kindred pathologic cascades and genetic pleiotropy. J Transl Med 2019; 99:993-1007. [PMID: 30742063 PMCID: PMC6609463 DOI: 10.1038/s41374-019-0196-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
We review the literature on Tau and TDP-43 proteinopathies in aged human brains and the relevant underlying pathogenetic cascades. Complex interacting pathways are implicated in Alzheimer's disease and related dementias (ADRD), wherein multiple proteins tend to misfold in a manner that is "reactive," but, subsequently, each proteinopathy may contribute strongly to the clinical symptoms. Tau proteinopathy exists in brains of individuals across a broad spectrum of primary underlying conditions-e.g., developmental, traumatic, and inflammatory/infectious diseases. TDP-43 proteinopathy is also expressed in a wide range of clinical disorders. Although TDP-43 proteinopathy was first described in the central nervous system of patients with amyotrophic lateral sclerosis (ALS) and in subtypes of frontotemporal dementia (FTD/FTLD), TDP-43 proteinopathy is also present in chronic traumatic encephalopathy, cognitively impaired persons in advanced age with hippocampal sclerosis, Huntington's disease, and other diseases. We list known Tau and TDP-43 proteinopathies. There is also evidence of cellular co-localization between Tau and TDP-43 misfolded proteins, suggesting common pathways or protein interactions facilitating misfolding in one protein by the other. Multiple pleiotropic gene variants can alter risk for Tau or TDP-43 pathologies, and certain gene variants (e.g., APOE ε4, Huntingtin triplet repeats) are associated with increases of both Tau and TDP-43 proteinopathies. Studies of genetic risk factors have provided insights into multiple nodes of the pathologic cascades involved in Tau and TDP-43 proteinopathies. Variants from a specific gene can be either a low-penetrant risk factor for a group of diseases, or alternatively, a different variant of the same gene may be a disease-driving allele that is associated with a relatively aggressive and early-onset version of a clinically and pathologically specific disease type. Overall, a complex but enlightening paradigm has emerged, wherein both Tau and TDP-43 proteinopathies are linked to numerous overlapping upstream influences, and both are associated with multiple downstream pathologically- and clinically-defined deleterious effects.
Collapse
|
37
|
Nelson PT, Dickson DW, Trojanowski JQ, Jack CR, Boyle PA, Arfanakis K, Rademakers R, Alafuzoff I, Attems J, Brayne C, Coyle-Gilchrist ITS, Chui HC, Fardo DW, Flanagan ME, Halliday G, Hokkanen SRK, Hunter S, Jicha GA, Katsumata Y, Kawas CH, Keene CD, Kovacs GG, Kukull WA, Levey AI, Makkinejad N, Montine TJ, Murayama S, Murray ME, Nag S, Rissman RA, Seeley WW, Sperling RA, White III CL, Yu L, Schneider JA. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain 2019; 142:1503-1527. [PMID: 31039256 PMCID: PMC6536849 DOI: 10.1093/brain/awz099] [Citation(s) in RCA: 851] [Impact Index Per Article: 170.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/10/2019] [Accepted: 02/25/2019] [Indexed: 12/18/2022] Open
Abstract
We describe a recently recognized disease entity, limbic-predominant age-related TDP-43 encephalopathy (LATE). LATE neuropathological change (LATE-NC) is defined by a stereotypical TDP-43 proteinopathy in older adults, with or without coexisting hippocampal sclerosis pathology. LATE-NC is a common TDP-43 proteinopathy, associated with an amnestic dementia syndrome that mimicked Alzheimer's-type dementia in retrospective autopsy studies. LATE is distinguished from frontotemporal lobar degeneration with TDP-43 pathology based on its epidemiology (LATE generally affects older subjects), and relatively restricted neuroanatomical distribution of TDP-43 proteinopathy. In community-based autopsy cohorts, ∼25% of brains had sufficient burden of LATE-NC to be associated with discernible cognitive impairment. Many subjects with LATE-NC have comorbid brain pathologies, often including amyloid-β plaques and tauopathy. Given that the 'oldest-old' are at greatest risk for LATE-NC, and subjects of advanced age constitute a rapidly growing demographic group in many countries, LATE has an expanding but under-recognized impact on public health. For these reasons, a working group was convened to develop diagnostic criteria for LATE, aiming both to stimulate research and to promote awareness of this pathway to dementia. We report consensus-based recommendations including guidelines for diagnosis and staging of LATE-NC. For routine autopsy workup of LATE-NC, an anatomically-based preliminary staging scheme is proposed with TDP-43 immunohistochemistry on tissue from three brain areas, reflecting a hierarchical pattern of brain involvement: amygdala, hippocampus, and middle frontal gyrus. LATE-NC appears to affect the medial temporal lobe structures preferentially, but other areas also are impacted. Neuroimaging studies demonstrated that subjects with LATE-NC also had atrophy in the medial temporal lobes, frontal cortex, and other brain regions. Genetic studies have thus far indicated five genes with risk alleles for LATE-NC: GRN, TMEM106B, ABCC9, KCNMB2, and APOE. The discovery of these genetic risk variants indicate that LATE shares pathogenetic mechanisms with both frontotemporal lobar degeneration and Alzheimer's disease, but also suggests disease-specific underlying mechanisms. Large gaps remain in our understanding of LATE. For advances in prevention, diagnosis, and treatment, there is an urgent need for research focused on LATE, including in vitro and animal models. An obstacle to clinical progress is lack of diagnostic tools, such as biofluid or neuroimaging biomarkers, for ante-mortem detection of LATE. Development of a disease biomarker would augment observational studies seeking to further define the risk factors, natural history, and clinical features of LATE, as well as eventual subject recruitment for targeted therapies in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Konstantinos Arfanakis
- Rush University Medical Center, Chicago, IL, USA
- Illinois Institute of Technology, Chicago, IL, USA
| | | | | | | | | | | | - Helena C Chui
- University of Southern California, Los Angeles, CA, USA
| | | | | | - Glenda Halliday
- The University of Sydney Brain and Mind Centre and Central Clinical School Faculty of Medicine and Health, Sydney, Australia
| | | | | | | | | | | | | | - Gabor G Kovacs
- Institute of Neurology Medical University of Vienna, Vienna, Austria
| | | | | | | | | | - Shigeo Murayama
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | | | - Sukriti Nag
- Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | - Lei Yu
- Rush University Medical Center, Chicago, IL, USA
| | | |
Collapse
|
38
|
Abstract
After more than 10 years of accumulated efforts, genome-wide association studies (GWAS) have led to many findings, most of which have been deposited into the GWAS Catalog. Between GWAS's inception and March 2017, the GWAS Catalog has collected 2429 studies, 1818 phenotypes, and 28,462 associated SNPs. We reclassified the psychology-related phenotypes into 217 reclassified phenotypes, which accounted for 514 studies and 7052 SNPs. In total, 1223 of the SNPs reached genome-wide significance. Of these, 147 were replicated for the same psychological trait in different studies. Another 305 SNPs were replicated within one original study. The SNPs rs2075650 and rs4420638 were linked to the most replications within a single reclassified phenotype or very similar reclassified phenotypes; both were associated with Alzheimer's disease (AD). Schizophrenia was associated with 74 within-phenotype SNPs reported in independents studies. Alzheimer's disease and schizophrenia were both linked to some physical phenotypes, including cholesterol and body mass index, through common GWAS signals. Alzheimer's disease also shared risk SNPs with age-related phenotypes such as age-related macular degeneration and longevity. Smoking-related SNPs were linked to lung cancer and respiratory function. Alcohol-related SNPs were associated with cardiovascular and digestive system phenotypes and disorders. Two separate studies also identified a shared risk SNP for bipolar disorder and educational attainment. This review revealed a list of reproducible SNPs worthy of future functional investigation. Additionally, by identifying SNPs associated with multiple phenotypes, we illustrated the importance of studying the relationships among phenotypes to resolve the nature of their causal links. The insights within this review will hopefully pave the way for future evidence-based genetic studies.
Collapse
|
39
|
Morita M, Ikeshima-Kataoka H, Kreft M, Vardjan N, Zorec R, Noda M. Metabolic Plasticity of Astrocytes and Aging of the Brain. Int J Mol Sci 2019; 20:ijms20040941. [PMID: 30795555 PMCID: PMC6413111 DOI: 10.3390/ijms20040941] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 01/03/2023] Open
Abstract
As part of the blood-brain-barrier, astrocytes are ideally positioned between cerebral vasculature and neuronal synapses to mediate nutrient uptake from the systemic circulation. In addition, astrocytes have a robust enzymatic capacity of glycolysis, glycogenesis and lipid metabolism, managing nutrient support in the brain parenchyma for neuronal consumption. Here, we review the plasticity of astrocyte energy metabolism under physiologic and pathologic conditions, highlighting age-dependent brain dysfunctions. In astrocytes, glycolysis and glycogenesis are regulated by noradrenaline and insulin, respectively, while mitochondrial ATP production and fatty acid oxidation are influenced by the thyroid hormone. These regulations are essential for maintaining normal brain activities, and impairments of these processes may lead to neurodegeneration and cognitive decline. Metabolic plasticity is also associated with (re)activation of astrocytes, a process associated with pathologic events. It is likely that the recently described neurodegenerative and neuroprotective subpopulations of reactive astrocytes metabolize distinct energy substrates, and that this preference is supposed to explain some of their impacts on pathologic processes. Importantly, physiologic and pathologic properties of astrocytic metabolic plasticity bear translational potential in defining new potential diagnostic biomarkers and novel therapeutic targets to mitigate neurodegeneration and age-related brain dysfunctions.
Collapse
Affiliation(s)
- Mitsuhiro Morita
- Department of Biology, Graduate School of Sciences, Kobe University, 657-8501 Kobe, Japan.
| | - Hiroko Ikeshima-Kataoka
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| | - Marko Kreft
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
- Department of Biology, Biotechnical Faculty University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
40
|
TDP-43 proteinopathy in aging: Associations with risk-associated gene variants and with brain parenchymal thyroid hormone levels. Neurobiol Dis 2019; 125:67-76. [PMID: 30682540 DOI: 10.1016/j.nbd.2019.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/13/2019] [Accepted: 01/19/2019] [Indexed: 02/08/2023] Open
Abstract
TDP-43 proteinopathy is very prevalent among the elderly (affecting at least 25% of individuals over 85 years of age) and is associated with substantial cognitive impairment. Risk factors implicated in age-related TDP-43 proteinopathy include commonly inherited gene variants, comorbid Alzheimer's disease pathology, and thyroid hormone dysfunction. To test parameters that are associated with aging-related TDP-43 pathology, we performed exploratory analyses of pathologic, genetic, and biochemical data derived from research volunteers in the University of Kentucky Alzheimer's Disease Center autopsy cohort (n = 136 subjects). Digital pathologic methods were used to discriminate and quantify both neuritic and intracytoplasmic TDP-43 pathology in the hippocampal formation. Overall, 46.4% of the cases were positive for TDP-43 intracellular inclusions, which is consistent with results in other prior community-based cohorts. The pathologies were correlated with hippocampal sclerosis of aging (HS-Aging) linked genotypes. We also assayed brain parenchymal thyroid hormone (triiodothyronine [T3] and thyroxine [T4]) levels. In cases with SLCO1A2/IAPP or ABCC9 risk associated genotypes, the T3/T4 ratio tended to be reduced (p = .051 using 2-tailed statistical test), and in cases with low T3/T4 ratios (bottom quintile), there was a higher likelihood of HS-Aging pathology (p = .025 using 2-tailed statistical test). This is intriguing because the SLCO1A2/IAPP and ABCC9 risk associated genotypes have been associated with altered expression of the astrocytic thyroid hormone receptor (protein product of the nearby gene SLCO1C1). These data indicate that dysregulation of thyroid hormone signaling may play a role in age-related TDP-43 proteinopathy.
Collapse
|
41
|
Trieu T, Sajjadi SA, Kawas CH, Nelson PT, Corrada MM. Risk factors of hippocampal sclerosis in the oldest old: The 90+ Study. Neurology 2018; 91:e1788-e1798. [PMID: 30315072 DOI: 10.1212/wnl.0000000000006455] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 07/31/2018] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE To examine the risk factors and comorbidities of hippocampal sclerosis (HS) in the oldest-old. METHODS A total of 134 participants with dementia from The 90+ Study with longitudinal evaluations and autopsy were included in this investigation. Participants were divided into 2 groups, one with and one without HS pathology, and differences in clinical and pathologic characteristics were compared. RESULTS Persons with HS tended to have a longer duration of dementia compared to participants without HS (mean 4.0 years vs 6.7 years, odds ratio [OR] 1.26; 95% confidence interval [CI] 1.11-1.42; p < 0.001). HS was more likely in participants with a history of autoimmune diseases (rheumatoid arthritis or thyroid disease, OR 3.15; 95% CI 1.30-7.62; p = 0.011), high thyroid-stimulating hormone (OR 4.94; 95% CI 1.40-17.46; p = 0.013), or high thyroid antibodies (OR 3.45; 95% CI 1.09-10.88; p = 0.035). Lewy body disease (LBD) pathology was also associated with an increased likelihood of HS (OR 5.70; 95% CI 1.22-26.4; p = 0.027). CONCLUSION We identified autoimmune conditions (rheumatoid arthritis and thyroid disease) as potential risk factors for HS in our cohort. LBD was the only pathology that was associated with increased odds of HS and those harboring HS pathology had a longer duration of dementia. This suggests multiple pathways of HS pathology among the oldest-old.
Collapse
Affiliation(s)
- Thomas Trieu
- From the College of Medicine (T.T.), The Pennsylvania State University, State College; Departments of Neurology (S.A.S., C.H.K., M.M.C.), Neurobiology and Behavior (C.H.K.), and Epidemiology (M.M.C.), University of California, Irvine; and Department of Pathology, University of Kentucky Sanders-Brown Center on Aging (P.T.N.), University of Kentucky, Lexington
| | - Seyed Ahmad Sajjadi
- From the College of Medicine (T.T.), The Pennsylvania State University, State College; Departments of Neurology (S.A.S., C.H.K., M.M.C.), Neurobiology and Behavior (C.H.K.), and Epidemiology (M.M.C.), University of California, Irvine; and Department of Pathology, University of Kentucky Sanders-Brown Center on Aging (P.T.N.), University of Kentucky, Lexington
| | - Claudia H Kawas
- From the College of Medicine (T.T.), The Pennsylvania State University, State College; Departments of Neurology (S.A.S., C.H.K., M.M.C.), Neurobiology and Behavior (C.H.K.), and Epidemiology (M.M.C.), University of California, Irvine; and Department of Pathology, University of Kentucky Sanders-Brown Center on Aging (P.T.N.), University of Kentucky, Lexington
| | - Peter T Nelson
- From the College of Medicine (T.T.), The Pennsylvania State University, State College; Departments of Neurology (S.A.S., C.H.K., M.M.C.), Neurobiology and Behavior (C.H.K.), and Epidemiology (M.M.C.), University of California, Irvine; and Department of Pathology, University of Kentucky Sanders-Brown Center on Aging (P.T.N.), University of Kentucky, Lexington
| | - María M Corrada
- From the College of Medicine (T.T.), The Pennsylvania State University, State College; Departments of Neurology (S.A.S., C.H.K., M.M.C.), Neurobiology and Behavior (C.H.K.), and Epidemiology (M.M.C.), University of California, Irvine; and Department of Pathology, University of Kentucky Sanders-Brown Center on Aging (P.T.N.), University of Kentucky, Lexington.
| |
Collapse
|
42
|
Vagaitseva KV, Bocharova AV, Marusin AV, Kolesnikova EA, Makeeva OA, Stepanov VA. Development of Multiplex Genotyping Method of Polymorphic Markers of Genes Associated with Cognitive Abilities. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418060121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Vidal-Taboada JM, Pugliese M, Salvadó M, Gámez J, Mahy N, Rodríguez MJ. K ATP Channel Expression and Genetic Polymorphisms Associated with Progression and Survival in Amyotrophic Lateral Sclerosis. Mol Neurobiol 2018; 55:7962-7972. [PMID: 29492846 DOI: 10.1007/s12035-018-0970-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/16/2018] [Indexed: 02/06/2023]
Abstract
The ATP-sensitive potassium (KATP) channel directly regulates the microglia-mediated inflammatory response following CNS injury. To determine the putative role of the KATP channel in amyotrophic lateral sclerosis (ALS) pathology, we investigated whether ALS induces changes in KATP channel expression in the spinal cord and motor cortex. We also characterized new functional variants of human ABCC8, ABCC9, KCNJ8, and KCNJ11 genes encoding for the KATP channel and analyzed their association with ALS risk, rate of progression, and survival in a Spanish ALS cohort. The expression of ABCC8 and KCNJ8 genes was enhanced in the spinal cord of ALS samples, and KCNJ11 increased in motor cortex of ALS samples, as determined by real-time polymerase chain reaction. We then sequenced the exons and regulatory regions of KATP channel genes from a subset of 28 ALS patients and identified 50 new genetic variants. For the case-control association analysis, we genotyped five selected polymorphisms with predicted functional relevance in 185 Spanish ALS (134 spinal ALS and 51 bulbar ALS) patients and 493 controls. We found that bulbar ALS patients presenting the G/G genotype of the rs4148646 variant of ABCC8 and the T/T genotype of the rs5219 variant of KCNJ11 survived longer than other ALS patients presenting other genotypes. Also, the C/C genotype of the rs4148642 variant of ABCC8 and the T/C genotype of the rs148416760 variant of ABCC9 modified the progression rate in spinal ALS patients. Our results suggest that the KATP channel plays a role in the pathophysiological mechanisms of ALS.
Collapse
Affiliation(s)
- José M Vidal-Taboada
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Marco Pugliese
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Maria Salvadó
- ALS Unit, Department of Neurology, Hospital Universitari Vall d'Hebron - VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Josep Gámez
- ALS Unit, Department of Neurology, Hospital Universitari Vall d'Hebron - VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nicole Mahy
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain
| | - Manuel J Rodríguez
- Department of Biomedical Sciences, Institut de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de Barcelona, Barcelona, Spain. .,Unitat de Bioquímica i Biologia Molecular, Department of Biomedicina, Facultat de Medicina, UB, c/ Casanova 143, E-08036, Barcelona, Spain.
| |
Collapse
|
44
|
Guo X, Qiu W, Garcia-Milian R, Lin X, Zhang Y, Cao Y, Tan Y, Wang Z, Shi J, Wang J, Liu D, Song L, Xu Y, Wang X, Liu N, Sun T, Zheng J, Luo J, Zhang H, Xu J, Kang L, Ma C, Wang K, Luo X. Genome-wide significant, replicated and functional risk variants for Alzheimer's disease. J Neural Transm (Vienna) 2017; 124:1455-1471. [PMID: 28770390 PMCID: PMC5654670 DOI: 10.1007/s00702-017-1773-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023]
Abstract
Genome-wide association studies (GWASs) have reported numerous associations between risk variants and Alzheimer's disease (AD). However, these associations do not necessarily indicate a causal relationship. If the risk variants can be demonstrated to be biologically functional, the possibility of a causal relationship would be increased. In this article, we reviewed all of the published GWASs to extract the genome-wide significant (p < 5×10-8) and replicated associations between risk variants and AD or AD-biomarkers. The regulatory effects of these risk variants on the expression of a novel class of non-coding RNAs (piRNAs) and protein-coding RNAs (mRNAs), the alteration of proteins caused by these variants, the associations between AD and these variants in our own sample, the expression of piRNAs, mRNAs and proteins in human brains targeted by these variants, the expression correlations between the risk genes and APOE, the pathways and networks that the risk genes belonged to, and the possible long non-coding RNAs (LncRNAs) that might regulate the risk genes were analyzed, to investigate the potential biological functions of the risk variants and explore the potential mechanisms underlying the SNP-AD associations. We found replicated and significant associations for AD or AD-biomarkers, surprisingly, only at 17 SNPs located in 11 genes/snRNAs/LncRNAs in eight genomic regions. Most of these 17 SNPs enriched some AD-related pathways or networks, and were potentially functional in regulating piRNAs and mRNAs; some SNPs were associated with AD in our sample, and some SNPs altered protein structures. Most of the protein-coding genes regulated by the risk SNPs were expressed in human brain and correlated with APOE expression. We conclude that these variants were most robust risk markers for AD, and their contributions to AD risk was likely to be causal. As expected, APOE and the lipoprotein metabolism pathway possess the highest weight among these contributions.
Collapse
Affiliation(s)
- Xiaoyun Guo
- Shanghai Mental Health Center, Shanghai 200030, China
- Department of Psychiatry, Yale University School of Medicine, New
Haven, CT 06510, USA
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of
Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences,
School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Rolando Garcia-Milian
- Curriculum & Research Support Department, Cushing/Whitney
Medical Library, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xiandong Lin
- Department of Pathology, Fujian Provincial Cancer Hospital, the
Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin 300222, China
| | - Yuping Cao
- Department of Psychiatry, Second Xiangya Hospital, Central South
University, Changsha 410012, China
| | - Yunlong Tan
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| | - Zhiren Wang
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| | - Jing Shi
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| | - Jijun Wang
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Dengtang Liu
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Lisheng Song
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Yifeng Xu
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Xiaoping Wang
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao
Tong University, Shanghai 200080, China
| | - Na Liu
- Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029,
China
| | - Tao Sun
- Huashan Hospital, Fudan University School of Medicine, Shanghai
200040, China
| | - Jianming Zheng
- Huashan Hospital, Fudan University School of Medicine, Shanghai
200040, China
| | - Justine Luo
- Department of Psychiatry, Yale University School of Medicine, New
Haven, CT 06510, USA
| | - Huihao Zhang
- The First Affiliated Hospital, Fujian Medical University, Fuzhou
350001, China
| | - Jianying Xu
- Zhuhai Municipal Maternal and Children’s Health Hospital,
Zhuhai, Guangdong 519000, China
| | - Longli Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention
Research on High Altitude Diseases of Tibet Autonomous Region, Xizang Minzu
University School of Medicine, Xiangyang, Shaanxi 712082, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of
Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences,
School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Kesheng Wang
- Department of Biostatistics and Epidemiology, College of Public
Health, East Tennessee State University, Johnson City, TN 37614, USA
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New
Haven, CT 06510, USA
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| |
Collapse
|
45
|
Buroker NE, Ning XH, Zhou ZN, Li K, Cen WJ, Wu XF, Zhu WZ, Scott CR, Chen SH. SNPs, linkage disequilibrium, and chronic mountain sickness in Tibetan Chinese. HYPOXIA 2017; 5:67-74. [PMID: 28770234 PMCID: PMC5529112 DOI: 10.2147/hp.s117967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic mountain sickness (CMS) is estimated at 1.2% in Tibetans living at the Qinghai-Tibetan Plateau. Eighteen single-nucleotide polymorphisms (SNPs) from nine nuclear genes that have an association with CMS in Tibetans have been analyzed by using pairwise linkage disequilibrium (LD). The SNPs included are the angiotensin-converting enzyme (rs4340), the angiotensinogen (rs699), and the angiotensin II type 1 receptor (AGTR1) (rs5186) from the renin-angiotensin system. A low-density lipoprotein apolipoprotein B (rs693) SNP was also included. From the hypoxia-inducible factor oxygen signaling pathway, the endothetal Per-Arnt-Sim domain protein 1 (EPAS1) and the egl nine homolog 1 (ENGL1) (rs480902) SNPs were included in the study. SNPs from the vascular endothelial growth factor (VEGF) signaling pathway included are the v-akt murine thymoma viral oncogene homolog 3 (rs4590656 and rs2291409), the endothelial cell nitric oxide synthase 3 (rs1007311 and rs1799983), and the (VEGFA) (rs699947, rs34357231, rs79469752, rs13207351, rs28357093, rs1570360, rs2010963, and rs3025039). An increase in LD occurred in 40 pairwise comparisons, whereas a decrease in LD was found in 55 pairwise comparisons between the controls and CMS patients. These changes were found to occur within and between signaling pathways, which suggests that there is an interaction between SNP alleles from different areas of the genome that affect CMS.
Collapse
Affiliation(s)
| | - Xue-Han Ning
- Department of Pediatrics, University of Washington.,Division of Cardiology, Seattle Children's Hospital Research Foundation, Seattle, WA, USA
| | - Zhao-Nian Zhou
- Laboratory of Hypoxia Physiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kui Li
- Lhasa People Hospital, Lhasa, Tibet
| | | | - Xiu-Feng Wu
- Laboratory of Hypoxia Physiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei-Zhong Zhu
- Center for Cardiovascular Biology and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | | | - Shi-Han Chen
- Department of Pediatrics, University of Washington
| |
Collapse
|
46
|
Qiu W, Guo X, Lin X, Yang Q, Zhang W, Zhang Y, Zuo L, Zhu Y, Li CSR, Ma C, Luo X. Transcriptome-wide piRNA profiling in human brains of Alzheimer's disease. Neurobiol Aging 2017; 57:170-177. [PMID: 28654860 DOI: 10.1016/j.neurobiolaging.2017.05.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/21/2017] [Accepted: 05/26/2017] [Indexed: 01/03/2023]
Abstract
Discovered in the brains of multiple animal species, piRNAs may contribute to the pathogenesis of neuropsychiatric illnesses. The present study aimed to identify brain piRNAs across transcriptome that are associated with Alzheimer's disease (AD). Prefrontal cortical tissues of 6 AD cases and 6 controls were examined for piRNA expression levels using an Arraystar HG19 piRNA array (containing 23,677 piRNAs) and genotyped for 17 genome-wide significant and replicated risk SNPs. We examined whether piRNAs are expressed differently between AD cases and controls and explored the potential regulatory effects of risk SNPs on piRNA expression levels. We identified a total of 9453 piRNAs in human brains, with 103 nominally (p < 0.05) differentially (>1.5 fold) expressed in AD cases versus controls and most of the 103 piRNAs nominally correlated with genome-wide significant risk SNPs. We conclude that piRNAs are abundant in human brains and may represent risk biomarkers of AD.
Collapse
Affiliation(s)
- Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoyun Guo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Shanghai Mental Health Center, Shanghai, China
| | - Xiandong Lin
- Department of Pathology, Fujian Provincial Cancer Hospital, the Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Qian Yang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wanying Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin, China
| | - Lingjun Zuo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yong Zhu
- Department of Environmental Health Sciences, Yale University School of Public Health, New Haven, CT, USA
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China.
| |
Collapse
|
47
|
Cykowski MD, Powell SZ, Schulz PE, Takei H, Rivera AL, Jackson RE, Roman G, Jicha GA, Nelson PT. Hippocampal Sclerosis in Older Patients: Practical Examples and Guidance With a Focus on Cerebral Age-Related TDP-43 With Sclerosis. Arch Pathol Lab Med 2017; 141:1113-1126. [PMID: 28467211 DOI: 10.5858/arpa.2016-0469-sa] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CONTEXT - Autopsy studies of the older population (≥65 years of age), and particularly of the "oldest-old" (≥85 years of age), have identified a significant proportion (∼20%) of cognitively impaired patients in which hippocampal sclerosis is the major substrate of an amnestic syndrome. Hippocampal sclerosis may also be comorbid with frontotemporal lobar degeneration, Alzheimer disease, and Lewy body disease. Until recently, the terms hippocampal sclerosis of aging or hippocampal sclerosis dementia were applied in this context. Recent discoveries have prompted a conceptual expansion of hippocampal sclerosis of aging because (1) cellular inclusions of TAR DNA-binding protein 43 kDa (TDP-43) are frequent; (2) TDP-43 pathology may be found outside hippocampus; and (3) brain arteriolosclerosis is a common, possibly pathogenic, component. OBJECTIVE - To aid pathologists with recent recommendations for diagnoses of common neuropathologies in older persons, particularly hippocampal sclerosis, and highlight the recent shift in diagnostic terminology from HS-aging to cerebral age-related TDP-43 with sclerosis (CARTS). DATA SOURCES - Peer-reviewed literature and 5 autopsy examples that illustrate common age-related neuropathologies, including CARTS, and emphasize the importance of distinguishing CARTS from late-onset frontotemporal lobar degeneration with TDP-43 pathology and from advanced Alzheimer disease with TDP-43 pathology. CONCLUSIONS - In advanced old age, the substrates of cognitive impairment are often multifactorial. This article demonstrates common and frequently comorbid neuropathologic substrates of cognitive impairment in the older population, including CARTS, to aid those practicing in this area of pathology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peter T Nelson
- From the Departments of Pathology and Genomic Medicine (Drs Cykowski, Powell, Rivera, and Takei), Internal Medicine (Dr Jackson), and Neurology (Dr Roman), Houston Methodist Hospital, Houston, Texas; the Department of Neurology, University of Texas Health Science Center at Houston (Dr Schulz); the Department of Pathology, Division of Neuropathology (Dr Nelson) and Sanders-Brown Center on Aging (Drs Jicha and Nelson), University of Kentucky, Lexington
| |
Collapse
|
48
|
Katsumata Y, Nelson PT, Ellingson SR, Fardo DW. Gene-based association study of genes linked to hippocampal sclerosis of aging neuropathology: GRN, TMEM106B, ABCC9, and KCNMB2. Neurobiol Aging 2017; 53:193.e17-193.e25. [PMID: 28131462 PMCID: PMC5385271 DOI: 10.1016/j.neurobiolaging.2017.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/29/2016] [Accepted: 01/03/2017] [Indexed: 12/12/2022]
Abstract
Hippocampal sclerosis of aging (HS-Aging) is a common neurodegenerative condition associated with dementia. To learn more about genetic risk of HS-Aging pathology, we tested gene-based associations of the GRN, TMEM106B, ABCC9, and KCNMB2 genes, which were reported to be associated with HS-Aging pathology in previous studies. Genetic data were obtained from the Alzheimer's Disease Genetics Consortium, linked to autopsy-derived neuropathological outcomes from the National Alzheimer's Coordinating Center. Of the 3251 subjects included in the study, 271 (8.3%) were identified as an HS-Aging case. The significant gene-based association between the ABCC9 gene and HS-Aging appeared to be driven by a region in which a significant haplotype-based association was found. We tested this haplotype as an expression quantitative trait locus using 2 different public-access brain gene expression databases. The HS-Aging pathology protective ABCC9 haplotype was associated with decreased ABCC9 expression, indicating a possible toxic gain of function.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Sally R Ellingson
- Division of Biomedical Informatics, University of Kentucky, Lexington, KY, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
49
|
Fardo DW, Katsumata Y, Kauwe JSK, Deming Y, Harari O, Cruchaga C, Nelson PT. CSF protein changes associated with hippocampal sclerosis risk gene variants highlight impact of GRN/PGRN. Exp Gerontol 2017; 90:83-89. [PMID: 28189700 DOI: 10.1016/j.exger.2017.01.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 12/31/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hippocampal sclerosis of aging (HS-Aging) is a common cause of dementia in older adults. We tested the variability in cerebrospinal fluid (CSF) proteins associated with previously identified HS-Aging risk single nucleotide polymorphisms (SNPs). METHODS Alzheimer's Disease Neuroimaging Initiative cohort (ADNI; n=237) data, combining both multiplexed proteomics CSF and genotype data, were used to assess the association between CSF analytes and risk SNPs in four genes (SNPs): GRN (rs5848), TMEM106B (rs1990622), ABCC9 (rs704180), and KCNMB2 (rs9637454). For controls, non-HS-Aging SNPs in APOE (rs429358/rs7412) and MAPT (rs8070723) were also analyzed against Aβ1-42 and total tau CSF analytes. RESULTS The GRN risk SNP (rs5848) status correlated with variation in CSF proteins, with the risk allele (T) associated with increased levels of AXL Receptor Tyrosine Kinase (AXL), TNF-Related Apoptosis-Inducing Ligand Receptor 3 (TRAIL-R3), Vascular Cell Adhesion Molecule-1 (VCAM-1) and clusterin (CLU) (all p<0.05 after Bonferroni correction). The TRAIL-R3 correlation was significant in meta-analysis with an additional dataset (p=5.05×10-5). Further, the rs5848 SNP status was associated with increased CSF tau protein - a marker of neurodegeneration (p=0.015). These data are remarkable since this GRN SNP has been found to be a risk factor for multiple types of dementia-related brain pathologies.
Collapse
Affiliation(s)
- David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA.
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | | | - Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Pathology, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
50
|
Nho K, Saykin AJ, Nelson PT. Hippocampal Sclerosis of Aging, a Common Alzheimer's Disease 'Mimic': Risk Genotypes are Associated with Brain Atrophy Outside the Temporal Lobe. J Alzheimers Dis 2017; 52:373-83. [PMID: 27003218 DOI: 10.3233/jad-160077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hippocampal sclerosis of aging (HS-Aging) is a common brain disease in older adults with a clinical course that is similar to Alzheimer's disease. Four single-nucleotide polymorphisms (SNPs) have previously shown association with HS-Aging. The present study investigated structural brain changes associated with these SNPs using surface-based analysis. Participants from the Alzheimer's Disease Neuroimaging Initiative cohort (ADNI; n = 1,239), with both MRI scans and genotype data, were used to assess the association between brain atrophy and previously identified HS-Aging risk SNPs in the following genes: GRN, TMEM106B, ABCC9, and KCNMB2 (minor allele frequency for each is >30%). A fifth SNP (near the ABCC9 gene) was evaluated in post-hoc analysis. The GRN risk SNP (rs5848_T) was associated with a pattern of atrophy in the dorsomedial frontal lobes bilaterally, remarkable since GRN is a risk factor for frontotemporal dementia. The ABCC9 risk SNP (rs704180_A) was associated with multifocal atrophy whereas a SNP (rs7488080_A) nearby (∼50 kb upstream) ABCC9 was associated with atrophy in the right entorhinal cortex. Neither TMEM106B (rs1990622_T), KCNMB2 (rs9637454_A), nor any of the non-risk alleles were associated with brain atrophy. When all four previously identified HS-Aging risk SNPs were summed into a polygenic risk score, there was a pattern of associated multifocal brain atrophy in a predominately frontal pattern. We conclude that common SNPs previously linked to HS-Aging pathology were associated with a distinct pattern of anterior cortical atrophy. Genetic variation associated with HS-Aging pathology may represent a non-Alzheimer's disease contribution to atrophy outside of the hippocampus in older adults.
Collapse
Affiliation(s)
- Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Peter T Nelson
- University of Kentucky, Sanders-Brown Center on Aging and Pathology Department, University of Kentucky, Lexington, KY, USA
| |
Collapse
|