1
|
Babić Leko M, Španić Popovački E, Willumsen N, Nikolac Perković M, Pleić N, Zubčić K, Langer Horvat L, Vogrinc Ž, Boban M, Borovečki F, Zemunik T, de Silva R, Šimić G. Further validation of the association between MAPT haplotype-tagging polymorphisms and Alzheimer's disease: neuropsychological tests, cerebrospinal fluid biomarkers, and APOE genotype. Front Mol Neurosci 2024; 17:1456670. [PMID: 39386049 PMCID: PMC11461444 DOI: 10.3389/fnmol.2024.1456670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/28/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Genetic studies have shown that variants in the microtubule-associated protein tau (MAPT) gene, which encodes tau protein, can increase the risk for Alzheimer's disease (AD). Additionally, two haplotypes of the MAPT gene (H1 and H2) are associated with various neurodegenerative disorders, including AD. This study aimed to test the association of MAPT haplotypes (H1 and H2) and MAPT haplotype-tagging polymorphisms (rs1467967, rs242557, rs3785883, rs2471738, del-In9, rs7521) with AD. Methods The study included 964 individuals: 113 with AD, 53 with mild cognitive impairment (MCI), 54 with other dementias, and 744 healthy controls. Results The results showed that individuals carrying the A allele in the MAPT rs1467967 polymorphism, the GG genotype in the MAPT rs7521 polymorphism, and the G allele in the MAPT rs242557 polymorphism had worse performance on various neuropsychological tests. Carriers of the C allele in MAPT rs2471738 polymorphism and CC homozygotes also showed worse performance on neuropsychological tests and pathological levels of several cerebrospinal fluid (CSF) biomarkers. However, T allele carriers in the MAPT rs2471738 polymorphism were more represented among patients with dementia and apolipoprotein E (APOE) ɛ4 carriers. Carriers of the H2 MAPT haplotype had worse performance on various neuropsychological tests, consistent with our previous study, which associated the H2 MAPT haplotype with pathological levels of CSF AD biomarkers. Regarding the MAPT rs3785883 polymorphism, further research is needed since both the AA and GG genotypes were associated with pathological levels of CSF and plasma AD biomarkers. Discussion In conclusion, further genetic studies are needed to elucidate the role of MAPT haplotypes and MAPT haplotype-tagging polymorphisms in the development of AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Ena Španić Popovački
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Nanet Willumsen
- Reta Lila Weston Institute, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, United Kingdom
| | | | - Nikolina Pleić
- Department of Biology and Human Genetics, School of Medicine, University of Split, Split, Croatia
| | - Klara Zubčić
- Department of Molecular Diagnostics and Genetics, Dubrava University Hospital, Zagreb, Croatia
| | - Lea Langer Horvat
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Željka Vogrinc
- Laboratory for Neurobiochemistry, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Marina Boban
- Department of Neurology, University Hospital Centre Zagreb, Kišpatićeva, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Kišpatićeva, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Department for Functional Genomics, Centre for Translational and Clinical Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Tatijana Zemunik
- Department of Biology and Human Genetics, School of Medicine, University of Split, Split, Croatia
- Department of Nuclear Medicine, University Hospital Split, Split, Croatia
| | - Rohan de Silva
- Reta Lila Weston Institute, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Goran Šimić
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
2
|
Toral-Rios D, Pichardo-Rojas P, Ruiz-Sánchez E, Rosas-Carrasco Ó, Carvajal-García R, Gálvez-Coutiño DC, Martínez-Rodríguez NL, Rubio-Chávez AD, Alcántara-Flores M, López-Ramírez A, Martínez-Rosas AR, Ruiz-Chow ÁA, Alonso-Vanegas M, Campos-Peña V. Synergistic Effect between the APOE ε4 Allele with Genetic Variants of GSK3B and MAPT: Differential Profile between Refractory Epilepsy and Alzheimer Disease. Int J Mol Sci 2024; 25:10228. [PMID: 39337715 PMCID: PMC11432663 DOI: 10.3390/ijms251810228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Temporal Lobe Epilepsy (TLE) is a chronic neurological disorder characterized by recurrent focal seizures originating in the temporal lobe. Despite the variety of antiseizure drugs currently available to treat TLE, about 30% of cases continue to have seizures. The etiology of TLE is complex and multifactorial. Increasing evidence indicates that Alzheimer's disease (AD) and drug-resistant TLE present common pathological features that may induce hyperexcitability, especially aberrant hyperphosphorylation of tau protein. Genetic polymorphic variants located in genes of the microtubule-associated protein tau (MAPT) and glycogen synthase kinase-3β (GSK3B) have been associated with the risk of developing AD. The APOE ε4 allele is a major genetic risk factor for AD. Likewise, a gene-dose-dependent effect of ε4 seems to influence TLE. The present study aimed to investigate whether the APOE ɛ4 allele and genetic variants located in the MAPT and GSK3B genes are associated with the risk of developing AD and drug-resistant TLE in a cohort of the Mexican population. A significant association with the APOE ε4 allele was observed in patients with AD and TLE. Additional genetic interactions were identified between this allele and variants of the MAPT and GSK3B genes.
Collapse
Affiliation(s)
- Danira Toral-Rios
- Department of Psychiatry, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Pavel Pichardo-Rojas
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Elizabeth Ruiz-Sánchez
- Neurochemistry Laboratory, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| | - Óscar Rosas-Carrasco
- Geriatric Assessment Center, Department of Health, Iberoamerican University, Mexico City 01219, Mexico
| | | | - Dey Carol Gálvez-Coutiño
- Experimental Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| | - Nancy Lucero Martínez-Rodríguez
- Epidemiological Research Unit in Endocrinology and Nutrition, Children's Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| | - Ana Daniela Rubio-Chávez
- High Specialty Medical Unit (UMAE), Specialty Hospital, National Medical Center (CMN), XXI Century, Mexico City 06720, Mexico
| | - Myr Alcántara-Flores
- Department of Psychiatry, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| | - Arely López-Ramírez
- Experimental Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| | - Alma Rosa Martínez-Rosas
- Cognition and Behavior Unit, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| | - Ángel Alberto Ruiz-Chow
- Department of Psychiatry, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| | - Mario Alonso-Vanegas
- Director of the International Center for Epilepsy Surgery, HMG-Coyoacan Hospital, Mexico City 04380, Mexico
| | - Victoria Campos-Peña
- Experimental Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| |
Collapse
|
3
|
Ressler HW, Humphrey J, Vialle RA, Babrowicz B, Kandoi S, Raj T, Dickson DW, Ertekin-Taner N, Crary JF, Farrell K. MAPT haplotype-associated transcriptomic changes in progressive supranuclear palsy. Acta Neuropathol Commun 2024; 12:135. [PMID: 39154163 PMCID: PMC11330133 DOI: 10.1186/s40478-024-01839-3] [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: 06/07/2024] [Accepted: 07/28/2024] [Indexed: 08/19/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative movement and cognitive disorder characterized by abnormal accumulation of the microtubule-associated protein tau in the brain. Biochemically, inclusions in PSP are enriched for tau proteoforms with four microtubule-binding domain repeats (4R), an isoform that arises from alternative tau pre-mRNA splicing. While preferential aggregation and reduced degradation of 4R tau protein is thought to play a role in inclusion formation and toxicity, an alternative hypothesis is that altered expression of tau mRNA isoforms plays a causal role. This stems from the observation that PSP is associated with common variation in the tau gene (MAPT) at the 17q21.31 locus which contains low copy number repeats flanking a large recurrent genomic inversion. The complex genomic structural changes at the locus give rise to two dominant haplotypes, termed H1 and H2, that have the potential to markedly influence gene expression. Here, we explored haplotype-dependent differences in gene expression using a bulk RNA-seq dataset derived from human post-mortem brain tissue from PSP (n = 84) and controls (n = 77) using a rigorous computational pipeline, including alternative pre-mRNA splicing. We found 3579 differentially expressed genes in the temporal cortex and 10,011 in the cerebellum. We also found 7214 differential splicing events in the temporal cortex and 18,802 in the cerebellum. In the cerebellum, total tau mRNA levels and the proportion of transcripts encoding 4R tau were significantly increased in PSP compared to controls. In the temporal cortex, the proportion of reads that expressed 4R tau was increased in cases compared to controls. 4R tau mRNA levels were significantly associated with the H1 haplotype in the temporal cortex. Further, we observed a marked haplotype-dependent difference in KANSL1 expression that was strongly associated with H1 in both brain regions. These findings support the hypothesis that sporadic PSP is associated with haplotype-dependent increases in 4R tau mRNA that might play a causal role in this disorder.
Collapse
Affiliation(s)
- Hadley W Ressler
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place Box 1194, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack Humphrey
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo A Vialle
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bergan Babrowicz
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place Box 1194, New York, NY, 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shrishtee Kandoi
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Towfique Raj
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - John F Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place Box 1194, New York, NY, 10029, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Kurt Farrell
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place Box 1194, New York, NY, 10029, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Andersen J, Jeffrey B, Varikatt W, Rodriguez M, Lin MW, Brown DA. IgLON5-IgG: Innocent Bystander or Perpetrator? Int J Mol Sci 2024; 25:7956. [PMID: 39063198 PMCID: PMC11276813 DOI: 10.3390/ijms25147956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Anti-IgLON5 (IgLON5-IgG)-associated disease is a newly defined clinical entity. This literature review aims to evaluate its pathogenesis, which remains a pivotal question. Features that favour a primary neurodegenerative mechanism include the non-inflammatory tauopathy neuropathological signature and overrepresentation of microtubule-associated protein tau (MAPT) H1/H1 genotype as seen in other sporadic tauopathies. In contrast, the cell-surface localisation of IgLON5, capability of anti-IgLON5 antibodies to exert direct in vitro pathogenicity and disrupt IgLON5 interactions with its binding partners, human leukocyte antigen (HLA)-DRB1*10:01 and HLA-DQB1*05:01 allele preponderance with high affinity binding of IgLON5 peptides, and responsiveness to immunotherapy favour a primary autoimmune process. The presentation and course of anti-IgLON5-associated disease is heterogenous; hence, we hypothesise that a multitude of immune mechanisms are likely simultaneously operational in this disease cohort.
Collapse
Affiliation(s)
- Jane Andersen
- Department of Immunology, NSW Health Pathology-ICPMR, Westmead Hospital, Sydney, NSW 2145, Australia; (B.J.); (M.-W.L.); (D.A.B.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; (W.V.); (M.R.)
| | - Bronte Jeffrey
- Department of Immunology, NSW Health Pathology-ICPMR, Westmead Hospital, Sydney, NSW 2145, Australia; (B.J.); (M.-W.L.); (D.A.B.)
- Faculty of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Winny Varikatt
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; (W.V.); (M.R.)
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology-ICPMR, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Michael Rodriguez
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; (W.V.); (M.R.)
- Douglass Hanly Moir Pathology, Sydney, NSW 2000, Australia
| | - Ming-Wei Lin
- Department of Immunology, NSW Health Pathology-ICPMR, Westmead Hospital, Sydney, NSW 2145, Australia; (B.J.); (M.-W.L.); (D.A.B.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; (W.V.); (M.R.)
| | - David A. Brown
- Department of Immunology, NSW Health Pathology-ICPMR, Westmead Hospital, Sydney, NSW 2145, Australia; (B.J.); (M.-W.L.); (D.A.B.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; (W.V.); (M.R.)
| |
Collapse
|
5
|
Pedicone C, Weitzman SA, Renton AE, Goate AM. Unraveling the complex role of MAPT-containing H1 and H2 haplotypes in neurodegenerative diseases. Mol Neurodegener 2024; 19:43. [PMID: 38812061 PMCID: PMC11138017 DOI: 10.1186/s13024-024-00731-x] [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: 09/30/2023] [Accepted: 05/11/2024] [Indexed: 05/31/2024] Open
Abstract
A ~ 1 Mb inversion polymorphism exists within the 17q21.31 locus of the human genome as direct (H1) and inverted (H2) haplotype clades. This inversion region demonstrates high linkage disequilibrium, but the frequency of each haplotype differs across ancestries. While the H1 haplotype exists in all populations and shows a normal pattern of genetic variability and recombination, the H2 haplotype is enriched in European ancestry populations, is less frequent in African ancestry populations, and nearly absent in East Asian ancestry populations. H1 is a known risk factor for several neurodegenerative diseases, and has been associated with many other traits, suggesting its importance in cellular phenotypes of the brain and entire body. Conversely, H2 is protective for these diseases, but is associated with predisposition to recurrent microdeletion syndromes and neurodevelopmental disorders such as autism. Many single nucleotide variants and copy number variants define H1/H2 haplotypes and sub-haplotypes, but identifying the causal variant(s) for specific diseases and phenotypes is complex due to the extended linkage equilibrium. In this review, we assess the current knowledge of this inversion region regarding genomic structure, gene expression, cellular phenotypes, and disease association. We discuss recent discoveries and challenges, evaluate gaps in knowledge, and highlight the importance of understanding the effect of the 17q21.31 haplotypes to promote advances in precision medicine and drug discovery for several diseases.
Collapse
Affiliation(s)
- Chiara Pedicone
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah A Weitzman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alan E Renton
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Ellis MJ, Lekka C, Holden KL, Tulmin H, Seedat F, O'Brien DP, Dhayal S, Zeissler ML, Knudsen JG, Kessler BM, Morgan NG, Todd JA, Richardson SJ, Stefana MI. Identification of high-performing antibodies for the reliable detection of Tau proteoforms by Western blotting and immunohistochemistry. Acta Neuropathol 2024; 147:87. [PMID: 38761203 PMCID: PMC11102361 DOI: 10.1007/s00401-024-02729-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 05/20/2024]
Abstract
Antibodies are essential research tools whose performance directly impacts research conclusions and reproducibility. Owing to its central role in Alzheimer's disease and other dementias, hundreds of distinct antibody clones have been developed against the microtubule-associated protein Tau and its multiple proteoforms. Despite this breadth of offer, limited understanding of their performance and poor antibody selectivity have hindered research progress. Here, we validate a large panel of Tau antibodies by Western blot (79 reagents) and immunohistochemistry (35 reagents). We address the reagents' ability to detect the target proteoform, selectivity, the impact of protein phosphorylation on antibody binding and performance in human brain samples. While most antibodies detected Tau at high levels, many failed to detect it at lower, endogenous levels. By WB, non-selective binding to other proteins affected over half of the antibodies tested, with several cross-reacting with the related MAP2 protein, whereas the "oligomeric Tau" T22 antibody reacted with monomeric Tau by WB, thus calling into question its specificity to Tau oligomers. Despite the presumption that "total" Tau antibodies are agnostic to post-translational modifications, we found that phosphorylation partially inhibits binding for many such antibodies, including the popular Tau-5 clone. We further combine high-sensitivity reagents, mass-spectrometry proteomics and cDNA sequencing to demonstrate that presumptive Tau "knockout" human cells continue to express residual protein arising through exon skipping, providing evidence of previously unappreciated gene plasticity. Finally, probing of human brain samples with a large panel of antibodies revealed the presence of C-term-truncated versions of all main Tau brain isoforms in both control and tauopathy donors. Ultimately, we identify a validated panel of Tau antibodies that can be employed in Western blotting and/or immunohistochemistry to reliably detect even low levels of Tau expression with high selectivity. This work represents an extensive resource that will enable the re-interpretation of published data, improve reproducibility in Tau research, and overall accelerate scientific progress.
Collapse
Affiliation(s)
- Michael J Ellis
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Christiana Lekka
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - Katie L Holden
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Hanna Tulmin
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Faheem Seedat
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Women's Centre, University of Oxford, John Radcliffe Hospital, Level 3, Oxford, UK
| | - Darragh P O'Brien
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Shalinee Dhayal
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - Marie-Louise Zeissler
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - Jakob G Knudsen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Oxford, Radcliffe, UK
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Benedikt M Kessler
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Noel G Morgan
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Sarah J Richardson
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - M Irina Stefana
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.
| |
Collapse
|
7
|
Valentino RR, Scotton WJ, Roemer SF, Lashley T, Heckman MG, Shoai M, Martinez-Carrasco A, Tamvaka N, Walton RL, Baker MC, Macpherson HL, Real R, Soto-Beasley AI, Mok K, Revesz T, Christopher EA, DeTure M, Seeley WW, Lee EB, Frosch MP, Molina-Porcel L, Gefen T, Redding-Ochoa J, Ghetti B, Robinson AC, Kobylecki C, Rowe JB, Beach TG, Teich AF, Keith JL, Bodi I, Halliday GM, Gearing M, Arzberger T, Morris CM, White CL, Mechawar N, Boluda S, MacKenzie IR, McLean C, Cykowski MD, Wang SHJ, Graff C, Nagra RM, Kovacs GG, Giaccone G, Neumann M, Ang LC, Carvalho A, Morris HR, Rademakers R, Hardy JA, Dickson DW, Rohrer JD, Ross OA. MAPT H2 haplotype and risk of Pick's disease in the Pick's disease International Consortium: a genetic association study. Lancet Neurol 2024; 23:487-499. [PMID: 38631765 DOI: 10.1016/s1474-4422(24)00083-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Pick's disease is a rare and predominantly sporadic form of frontotemporal dementia that is classified as a primary tauopathy. Pick's disease is pathologically defined by the presence in the frontal and temporal lobes of Pick bodies, composed of hyperphosphorylated, three-repeat tau protein, encoded by the MAPT gene. MAPT has two distinct haplotypes, H1 and H2; the MAPT H1 haplotype is the major genetic risk factor for four-repeat tauopathies (eg, progressive supranuclear palsy and corticobasal degeneration), and the MAPT H2 haplotype is protective for these disorders. The primary aim of this study was to evaluate the association of MAPT H2 with Pick's disease risk, age at onset, and disease duration. METHODS In this genetic association study, we used data from the Pick's disease International Consortium, which we established to enable collection of data from individuals with pathologically confirmed Pick's disease worldwide. For this analysis, we collected brain samples from individuals with pathologically confirmed Pick's disease from 35 sites (brainbanks and hospitals) in North America, Europe, and Australia between Jan 1, 2020, and Jan 31, 2023. Neurologically healthy controls were recruited from the Mayo Clinic (FL, USA, or MN, USA between March 1, 1998, and Sept 1, 2019). For the primary analysis, individuals were directly genotyped for the MAPT H1-H2 haplotype-defining variant rs8070723. In a secondary analysis, we genotyped and constructed the six-variant-defined (rs1467967-rs242557-rs3785883-rs2471738-rs8070723-rs7521) MAPT H1 subhaplotypes. Associations of MAPT variants and MAPT haplotypes with Pick's disease risk, age at onset, and disease duration were examined using logistic and linear regression models; odds ratios (ORs) and β coefficients were estimated and correspond to each additional minor allele or each additional copy of the given haplotype. FINDINGS We obtained brain samples from 338 people with pathologically confirmed Pick's disease (205 [61%] male and 133 [39%] female; 338 [100%] White) and 1312 neurologically healthy controls (611 [47%] male and 701 [53%] female; 1312 [100%] White). The MAPT H2 haplotype was associated with increased risk of Pick's disease compared with the H1 haplotype (OR 1·35 [95% CI 1·12 to 1·64], p=0·0021). MAPT H2 was not associated with age at onset (β -0·54 [95% CI -1·94 to 0·87], p=0·45) or disease duration (β 0·05 [-0·06 to 0·16], p=0·35). Although not significant after correcting for multiple testing, associations were observed at p less than 0·05: with risk of Pick's disease for the H1f subhaplotype (OR 0·11 [0·01 to 0·99], p=0·049); with age at onset for H1b (β 2·66 [0·63 to 4·70], p=0·011), H1i (β -3·66 [-6·83 to -0·48], p=0·025), and H1u (β -5·25 [-10·42 to -0·07], p=0·048); and with disease duration for H1x (β -0·57 [-1·07 to -0·07], p=0·026). INTERPRETATION The Pick's disease International Consortium provides an opportunity to do large studies to enhance our understanding of the pathobiology of Pick's disease. This study shows that, in contrast to the decreased risk of four-repeat tauopathies, the MAPT H2 haplotype is associated with an increased risk of Pick's disease in people of European ancestry. This finding could inform development of isoform-related therapeutics for tauopathies. FUNDING Wellcome Trust, Rotha Abraham Trust, Brain Research UK, the Dolby Fund, Dementia Research Institute (Medical Research Council), US National Institutes of Health, and the Mayo Clinic Foundation.
Collapse
Affiliation(s)
| | - William J Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK.
| | - Shanu F Roemer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, University College London, Queen Square Institute of Neurology London, UK; Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology London, UK
| | - Michael G Heckman
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL, USA
| | - Maryam Shoai
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology London, UK
| | - Alejandro Martinez-Carrasco
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology London, UK
| | - Nicole Tamvaka
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Ronald L Walton
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Matthew C Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Hannah L Macpherson
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology London, UK
| | | | - Kin Mok
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology London, UK; UK Dementia Research Institute at UCL, London, UK; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders, University College London, Queen Square Institute of Neurology London, UK; Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology London, UK
| | | | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew P Frosch
- Neuropathology Service, C S Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura Molina-Porcel
- Neurological Tissue Bank, Biobanc-Hospital Clínic-Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Alzheimer's Disease and other Cognitive Disorders Unit, Neurology Department, Hospital Clinic, Barcelona, Spain; Barcelona Clinical Research Foundation-August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew C Robinson
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Christopher Kobylecki
- Department of Neurology, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, UK
| | - James B Rowe
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK; Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| | - Thomas G Beach
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Andrew F Teich
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Julia L Keith
- Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Istvan Bodi
- Clinical Neuropathology Department, King's College Hospital NHS Foundation Trust, London, UK; London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Glenda M Halliday
- University of Sydney Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences, Camperdown, NSW, Australia
| | - Marla Gearing
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Goizueta Alzheimer's Disease Center Brain Bank, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas Arzberger
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christopher M Morris
- Newcastle Brain Tissue Resource, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Charles L White
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Naguib Mechawar
- Douglas Hospital Research Centre, McGill University, Montreal, QC, Canada
| | - Susana Boluda
- Laboratoire de Neuropathologie Escourolle, Hôpital de la Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France; Alzheimer Prion Team, L'Institut du Cerveau, Paris, France
| | - Ian R MacKenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Catriona McLean
- Department of Anatomical Pathology Alfred Heath, Melbourne, VIC, Australia; Victorian Brain Bank, The Florey Institute of Neuroscience of Mental Health, Parkville, VIC, Australia
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Weill Cornell Medicine, Houston, TX, USA
| | - Shih-Hsiu J Wang
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Caroline Graff
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Unit for Hereditary Dementias, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Rashed M Nagra
- Human Brain and Spinal Fluid Resource Center, Brentwood Biomedical Research Institute, Los Angeles, CA, USA
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Giorgio Giaccone
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Manuela Neumann
- Molecular Neuropathology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany; Department of Neuropathology, University Hospital of Tübingen, Tübingen, Germany
| | - Lee-Cyn Ang
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology London, UK
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Vlaams Instituut voor Biotechnologie-Universiteit Antwerpen, Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - John A Hardy
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology London, UK; Reta Lila Weston Institute, University College London, Queen Square Institute of Neurology London, UK; UK Dementia Research Institute at UCL, London, UK; Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
| | | | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
8
|
Rogers BB, Anderson AG, Lauzon SN, Davis MN, Hauser RM, Roberts SC, Rodriguez-Nunez I, Trausch-Lowther K, Barinaga EA, Hall PI, Knuesel MT, Taylor JW, Mackiewicz M, Roberts BS, Cooper SJ, Rizzardi LF, Myers RM, Cochran JN. Neuronal MAPT expression is mediated by long-range interactions with cis-regulatory elements. Am J Hum Genet 2024; 111:259-279. [PMID: 38232730 PMCID: PMC10870142 DOI: 10.1016/j.ajhg.2023.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
Tauopathies are a group of neurodegenerative diseases defined by abnormal aggregates of tau, a microtubule-associated protein encoded by MAPT. MAPT expression is near absent in neural progenitor cells (NPCs) and increases during differentiation. This temporally dynamic expression pattern suggests that MAPT expression could be controlled by transcription factors and cis-regulatory elements specific to differentiated cell types. Given the relevance of MAPT expression to neurodegeneration pathogenesis, identification of such elements is relevant to understanding disease risk and pathogenesis. Here, we performed chromatin conformation assays (HiC & Capture-C), single-nucleus multiomics (RNA-seq+ATAC-seq), bulk ATAC-seq, and ChIP-seq for H3K27ac and CTCF in NPCs and differentiated neurons to nominate candidate cis-regulatory elements (cCREs). We assayed these cCREs using luciferase assays and CRISPR interference (CRISPRi) experiments to measure their effects on MAPT expression. Finally, we integrated cCRE annotations into an analysis of genetic variation in neurodegeneration-affected individuals and control subjects. We identified both proximal and distal regulatory elements for MAPT and confirmed the regulatory function for several regions, including three regions centromeric to MAPT beyond the H1/H2 haplotype inversion breakpoint. We also found that rare and predicted damaging genetic variation in nominated CREs was nominally depleted in dementia-affected individuals relative to control subjects, consistent with the hypothesis that variants that disrupt MAPT enhancer activity, and thereby reduced MAPT expression, may be protective against neurodegenerative disease. Overall, this study provides compelling evidence for pursuing detailed knowledge of CREs for genes of interest to permit better understanding of disease risk.
Collapse
Affiliation(s)
- Brianne B Rogers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Shelby N Lauzon
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - M Natalie Davis
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Rebecca M Hauser
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Sydney C Roberts
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | | | - Erin A Barinaga
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Paige I Hall
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Jared W Taylor
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Mark Mackiewicz
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Brian S Roberts
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Sara J Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA.
| | | |
Collapse
|
9
|
Robbins M. Therapies for Tau-associated neurodegenerative disorders: targeting molecules, synapses, and cells. Neural Regen Res 2023; 18:2633-2637. [PMID: 37449601 PMCID: PMC10358644 DOI: 10.4103/1673-5374.373670] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/14/2023] [Accepted: 03/15/2023] [Indexed: 07/18/2023] Open
Abstract
Advances in experimental and computational technologies continue to grow rapidly to provide novel avenues for the treatment of neurodegenerative disorders. Despite this, there remain only a handful of drugs that have shown success in late-stage clinical trials for Tau-associated neurodegenerative disorders. The most commonly prescribed treatments are symptomatic treatments such as cholinesterase inhibitors and N-methyl-D-aspartate receptor blockers that were approved for use in Alzheimer's disease. As diagnostic screening can detect disorders at earlier time points, the field needs pre-symptomatic treatments that can prevent, or significantly delay the progression of these disorders (Koychev et al., 2019). These approaches may be different from late-stage treatments that may help to ameliorate symptoms and slow progression once symptoms have become more advanced should early diagnostic screening fail. This mini-review will highlight five key avenues of academic and industrial research for identifying therapeutic strategies to treat Tau-associated neurodegenerative disorders. These avenues include investigating (1) the broad class of chemicals termed "small molecules"; (2) adaptive immunity through both passive and active antibody treatments; (3) innate immunity with an emphasis on microglial modulation; (4) synaptic compartments with the view that Tau-associated neurodegenerative disorders are synaptopathies. Although this mini-review will focus on Alzheimer's disease due to its prevalence, it will also argue the need to target other tauopathies, as through understanding Alzheimer's disease as a Tau-associated neurodegenerative disorder, we may be able to generalize treatment options. For this reason, added detail linking back specifically to Tau protein as a direct therapeutic target will be added to each topic.
Collapse
Affiliation(s)
- Miranda Robbins
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Ave, Trumpington, Cambridge, UK; University of Cambridge, Department of Zoology, Cambridge, UK
| |
Collapse
|
10
|
Gasca-Salas C, Trompeta C, López-Aguirre M, Rodríguez Rojas R, Clarimon J, Dols-Icardo O, El Bounasri S, Guida P, Mata-Marín D, Hernández-Fernández F, Marras C, García-Cañamaque L, Plaza de Las Heras I, Obeso I, Vela L, Fernández-Rodríguez B. Brain hypometabolism in non-demented microtubule-associated protein tau H1 carriers with Parkinson's disease. J Neuroimaging 2023; 33:953-959. [PMID: 37726927 DOI: 10.1111/jon.13156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND AND PURPOSE The microtubule-associated protein tau (MAPT) H1 homozygosity (H1/H1 haplotype) is a genetic risk factor for neurodegenerative diseases, such as Parkinson's disease (PD). MAPT H1 homozygosity has been associated with conversion to PD; however, results are conflicting since some studies did not find a strong influence. Cortical hypometabolism is associated with cognitive impairment in PD. In this study, we aimed to evaluate the metabolic pattern in nondemented PD patients MAPT H1/H1 carriers in comparison with MAPT H1/H2 haplotype. In addition, we evaluated domain-specific cognitive differences according to MAPT haplotype. METHODS We compared a group of 26 H1/H1 and 20 H1/H2 carriers with late-onset PD. Participants underwent a comprehensive neuropsychological cognitive evaluation and a [18F]-Fluorodeoxyglucose PET-MR scan. RESULTS MAPT H1/H1 carriers showed worse performance in the digit span forward test of attention compared to MAPT H1/H2 carriers. In the [18F]-Fluorodeoxyglucose PET comparisons, MAPT H1/H1 displayed hypometabolism in the frontal cortex, parahippocampal, and cingulate gyrus, as well as in the caudate and globus pallidus. CONCLUSION PD patients MAPT H1/H1 carriers without dementia exhibit relative hypometabolism in several cortical areas as well as in the basal ganglia, and worse performance in attention than MAPT H1/H2 carriers. Longitudinal studies should assess if lower scores in attention and dysfunction in these areas are predictors of dementia in MAPT H1/H1 homozygotes.
Collapse
Affiliation(s)
- Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- University CEU-San Pablo, Madrid, Spain
| | - Clara Trompeta
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Health Sciences, University of Alcala de Henares Alcalá de Henares, Madrid, Spain
| | - Miguel López-Aguirre
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- PhD Program in Physics, Complutense University of Madrid, Madrid, Spain
| | - Rafael Rodríguez Rojas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Jordi Clarimon
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Shaimaa El Bounasri
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pasqualina Guida
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience, Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - David Mata-Marín
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience, Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Frida Hernández-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Department of Nursing and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Connie Marras
- The Edmond J Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Lina García-Cañamaque
- Nuclear Medicine Department, PET-MRI Centre, HM Puerta del Sur University Hospital, HM Hospitales, Madrid, Spain
| | - Isabel Plaza de Las Heras
- Nuclear Medicine Department, PET-MRI Centre, HM Puerta del Sur University Hospital, HM Hospitales, Madrid, Spain
| | - Ignacio Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Lydia Vela
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Department of Neurology, Hospital U Fundación Alcorcón, Calle Budapest, Alcorcón, Spain
| | - Beatriz Fernández-Rodríguez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience, Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| |
Collapse
|
11
|
Samudra N, Lane-Donovan C, VandeVrede L, Boxer AL. Tau pathology in neurodegenerative disease: disease mechanisms and therapeutic avenues. J Clin Invest 2023; 133:e168553. [PMID: 37317972 PMCID: PMC10266783 DOI: 10.1172/jci168553] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023] Open
Abstract
Tauopathies are disorders associated with tau protein dysfunction and insoluble tau accumulation in the brain at autopsy. Multiple lines of evidence from human disease, as well as nonclinical translational models, suggest that tau has a central pathologic role in these disorders, historically thought to be primarily related to tau gain of toxic function. However, a number of tau-targeting therapies with various mechanisms of action have shown little promise in clinical trials in different tauopathies. We review what is known about tau biology, genetics, and therapeutic mechanisms that have been tested in clinical trials to date. We discuss possible reasons for failures of these therapies, such as use of imperfect nonclinical models that do not predict human effects for drug development; heterogeneity of human tau pathologies which may lead to variable responses to therapy; and ineffective therapeutic mechanisms, such as targeting of the wrong tau species or protein epitope. Innovative approaches to human clinical trials can help address some of the difficulties that have plagued our field's development of tau-targeting therapies thus far. Despite limited clinical success to date, as we continue to refine our understanding of tau's pathogenic mechanism(s) in different neurodegenerative diseases, we remain optimistic that tau-targeting therapies will eventually play a central role in the treatment of tauopathies.
Collapse
|
12
|
Conte M, De Feo MS, Sidrak MMA, Corica F, Gorica J, Granese GM, Filippi L, De Vincentis G, Frantellizzi V. Imaging of Tauopathies with PET Ligands: State of the Art and Future Outlook. Diagnostics (Basel) 2023; 13:diagnostics13101682. [PMID: 37238166 DOI: 10.3390/diagnostics13101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: Tauopathies are a group of diseases characterized by the deposition of abnormal tau protein. They are distinguished into 3R, 4R, and 3R/4R tauopathies and also include Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Positron emission tomography (PET) imaging represents a pivotal instrument to guide clinicians. This systematic review aims to summarize the current and novel PET tracers. (2) Methods: Literature research was conducted on Pubmed, Scopus, Medline, Central, and the Web of Science using the query "pet ligands" and "tauopathies". Articles published from January 2018 to 9 February, 2023, were searched. Only studies on the development of novel PET radiotracers for imaging in tauopathies or comparative studies between existing PET tracers were included. (3) Results: A total of 126 articles were found, as follows: 96 were identified from PubMed, 27 from Scopus, one on Central, two on Medline, and zero on the Web of Science. Twenty-four duplicated works were excluded, and 63 articles did not satisfy the inclusion criteria. The remaining 40 articles were included for quality assessment. (4) Conclusions: PET imaging represents a valid instrument capable of helping clinicians in diagnosis, but it is not always perfect in differential diagnosis, even if further investigations on humans for novel promising ligands are needed.
Collapse
Affiliation(s)
- Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Silvia De Feo
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marko Magdi Abdou Sidrak
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Ferdinando Corica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Joana Gorica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Giorgia Maria Granese
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Luca Filippi
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, 00410 Latina, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
13
|
Valentino RR, Scotton WJ, Roemer SF, Lashley T, Heckman MG, Shoai M, Martinez-Carrasco A, Tamvaka N, Walton RL, Baker MC, Macpherson HL, Real R, Soto-Beasley AI, Mok K, Revesz T, Warner TT, Jaunmuktane Z, Boeve BF, Christopher EA, DeTure M, Duara R, Graff-Radford NR, Josephs KA, Knopman DS, Koga S, Murray ME, Lyons KE, Pahwa R, Parisi JE, Petersen RC, Whitwell J, Grinberg LT, Miller B, Schlereth A, Seeley WW, Spina S, Grossman M, Irwin DJ, Lee EB, Suh E, Trojanowski JQ, Van Deerlin VM, Wolk DA, Connors TR, Dooley PM, Frosch MP, Oakley DH, Aldecoa I, Balasa M, Gelpi E, Borrego-Écija S, de Eugenio Huélamo RM, Gascon-Bayarri J, Sánchez-Valle R, Sanz-Cartagena P, Piñol-Ripoll G, Molina-Porcel L, Bigio EH, Flanagan ME, Gefen T, Rogalski EJ, Weintraub S, Redding-Ochoa J, Chang K, Troncoso JC, Prokop S, Newell KL, Ghetti B, Jones M, Richardson A, Robinson AC, Roncaroli F, Snowden J, Allinson K, Green O, Rowe JB, Singh P, Beach TG, Serrano GE, Flowers XE, Goldman JE, Heaps AC, Leskinen SP, Teich AF, Black SE, Keith JL, Masellis M, Bodi I, King A, Sarraj SA, Troakes C, Halliday GM, Hodges JR, Kril JJ, Kwok JB, Piguet O, Gearing M, Arzberger T, Roeber S, Attems J, Morris CM, Thomas AJ, Evers BM, White CL, Mechawar N, Sieben AA, Cras PP, De Vil BB, De Deyn PPP, Duyckaerts C, Le Ber I, Seihean D, Turbant-Leclere S, MacKenzie IR, McLean C, Cykowski MD, Ervin JF, Wang SHJ, Graff C, Nennesmo I, Nagra RM, Riehl J, Kovacs GG, Giaccone G, Nacmias B, Neumann M, Ang LC, Finger EC, Blauwendraat C, Nalls MA, Singleton AB, Vitale D, Cunha C, Carvalho A, Wszolek ZK, Morris HR, Rademakers R, Hardy JA, Dickson DW, Rohrer JD, Ross OA. Creating the Pick's disease International Consortium: Association study of MAPT H2 haplotype with risk of Pick's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.17.23288471. [PMID: 37163045 PMCID: PMC10168402 DOI: 10.1101/2023.04.17.23288471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background Pick's disease (PiD) is a rare and predominantly sporadic form of frontotemporal dementia that is classified as a primary tauopathy. PiD is pathologically defined by argyrophilic inclusion Pick bodies and ballooned neurons in the frontal and temporal brain lobes. PiD is characterised by the presence of Pick bodies which are formed from aggregated, hyperphosphorylated, 3-repeat tau proteins, encoded by the MAPT gene. The MAPT H2 haplotype has consistently been associated with a decreased disease risk of the 4-repeat tauopathies of progressive supranuclear palsy and corticobasal degeneration, however its role in susceptibility to PiD is unclear. The primary aim of this study was to evaluate the association between MAPT H2 and risk of PiD. Methods We established the Pick's disease International Consortium (PIC) and collected 338 (60.7% male) pathologically confirmed PiD brains from 39 sites worldwide. 1,312 neurologically healthy clinical controls were recruited from Mayo Clinic Jacksonville, FL (N=881) or Rochester, MN (N=431). For the primary analysis, subjects were directly genotyped for MAPT H1-H2 haplotype-defining variant rs8070723. In secondary analysis, we genotyped and constructed the six-variant MAPT H1 subhaplotypes (rs1467967, rs242557, rs3785883, rs2471738, rs8070723, and rs7521). Findings Our primary analysis found that the MAPT H2 haplotype was associated with increased risk of PiD (OR: 1.35, 95% CI: 1.12-1.64 P=0.002). In secondary analysis involving H1 subhaplotypes, a protective association with PiD was observed for the H1f haplotype (0.0% vs. 1.2%, P=0.049), with a similar trend noted for H1b (OR: 0.76, 95% CI: 0.58-1.00, P=0.051). The 4-repeat tauopathy risk haplotype MAPT H1c was not associated with PiD susceptibility (OR: 0.93, 95% CI: 0.70-1.25, P=0.65). Interpretation The PIC represents the first opportunity to perform relatively large-scale studies to enhance our understanding of the pathobiology of PiD. This study demonstrates that in contrast to its protective role in 4R tauopathies, the MAPT H2 haplotype is associated with an increased risk of PiD. This finding is critical in directing isoform-related therapeutics for tauopathies.
Collapse
Affiliation(s)
| | - William J Scotton
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
| | - Shanu F Roemer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, University College London, Queen Square Institute of Neurology London, UK
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
| | - Michael G Heckman
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Maryam Shoai
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
| | - Alejandro Martinez-Carrasco
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, UK
| | - Nicole Tamvaka
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ronald L Walton
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Matthew C Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Hannah L Macpherson
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
| | - Raquel Real
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, UK
| | | | - Kin Mok
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders, University College London, Queen Square Institute of Neurology London, UK
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
| | - Thomas T Warner
- Queen Square Brain Bank for Neurological Disorders, University College London, Queen Square Institute of Neurology London, UK
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, UK
| | - Zane Jaunmuktane
- Queen Square Brain Bank for Neurological Disorders, University College London, Queen Square Institute of Neurology London, UK
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, UK
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease and Memory Disorders, Mount Sinai Medical Center Miami Beach, FL
| | | | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kelly E Lyons
- University of Kansas Medical Center, Parkinson’s Disease & Movement Disorder Division, Kansas City, KS. 66160
| | - Rajesh Pahwa
- University of Kansas Medical Center, Parkinson’s Disease & Movement Disorder Division, Kansas City, KS. 66160
| | - Joseph E Parisi
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Bruce Miller
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Athena Schlereth
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Murray Grossman
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - EunRan Suh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David A Wolk
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theresa R Connors
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Patrick M Dooley
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Matthew P Frosch
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Derek H Oakley
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Iban Aldecoa
- Pathology, BDC, Hospital Clinic de Barcelona, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Neurological Tissue Bank, Biobanc-Hospital Clínic-FRCB-IDIBAPS, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer’s Disease and other Cognitive Disorders Unit, Neurology Department, Hospital Clinic, Barcelona, Spain
- Barcelona Clinical Research Foundation-August Pi i Sunyer Biomedical Research Institute (FRCB-IDIBAPS), Barcelona, Spain
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sergi Borrego-Écija
- University of Barcelona, Barcelona, Spain
- Alzheimer’s Disease and other Cognitive Disorders Unit, Neurology Department, Hospital Clinic, Barcelona, Spain
- Barcelona Clinical Research Foundation-August Pi i Sunyer Biomedical Research Institute (FRCB-IDIBAPS), Barcelona, Spain
| | | | - Jordi Gascon-Bayarri
- Servei de Neurologia, Hospital Universitari de Bellvitge. Institut d’Investigació Biomèdica de Bellvitge (Idibell). L’Hospitalet de Llobregat, Spain
| | - Raquel Sánchez-Valle
- University of Barcelona, Barcelona, Spain
- Alzheimer’s Disease and other Cognitive Disorders Unit, Neurology Department, Hospital Clinic, Barcelona, Spain
- Barcelona Clinical Research Foundation-August Pi i Sunyer Biomedical Research Institute (FRCB-IDIBAPS), Barcelona, Spain
| | | | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius (Cognitive Disorders Unit), Clinical Neuroscience Research, IRBLleida, Santa Maria University Hospital, Lleida, Spain
| | - Laura Molina-Porcel
- Neurological Tissue Bank, Biobanc-Hospital Clínic-FRCB-IDIBAPS, Barcelona, Spain
- Alzheimer’s Disease and other Cognitive Disorders Unit, Neurology Department, Hospital Clinic, Barcelona, Spain
- Barcelona Clinical Research Foundation-August Pi i Sunyer Biomedical Research Institute (FRCB-IDIBAPS), Barcelona, Spain
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily J Rogalski
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Koping Chang
- Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Stefan Prokop
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Kathy L Newell
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Matthew Jones
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, UK
- Division of Neuroscience, School of Biological Sciences, University of Manchester, UK
| | - Anna Richardson
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, UK
- Division of Neuroscience, School of Biological Sciences, University of Manchester, UK
| | - Andrew C Robinson
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, M6 8HD, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre (MAHSC), Manchester, UK
| | - Federico Roncaroli
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, M6 8HD, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre (MAHSC), Manchester, UK
| | - Julie Snowden
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, UK
- Division of Neuroscience, School of Biological Sciences, University of Manchester, UK
| | - Kieren Allinson
- Histopathology Box 235 Cambridge University Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ
| | - Oliver Green
- Histopathology Box 235 Cambridge University Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ
| | - James B Rowe
- Cambridge University Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| | - Poonam Singh
- Histopathology Box 235 Cambridge University Hospital NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ
| | - Thomas G Beach
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Geidy E Serrano
- Civin Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Xena E Flowers
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Allison C Heaps
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Sandra P Leskinen
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Andrew F Teich
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Sandra E Black
- Department of Medicine, Division of Neurology, Sunnybrook Health Sciences Centre and University of Toronto, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute
| | - Julia L Keith
- Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, and Laboratory Medicine and Pathobiology, University of Toronto
| | - Mario Masellis
- Department of Medicine, Division of Neurology, Sunnybrook Health Sciences Centre and University of Toronto, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute
| | - Istvan Bodi
- Clinical Neuropathology Department, King’s College Hospital NHS Foundation Trust, London, UK
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Andrew King
- Clinical Neuropathology Department, King’s College Hospital NHS Foundation Trust, London, UK
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Safa-Al Sarraj
- Clinical Neuropathology Department, King’s College Hospital NHS Foundation Trust, London, UK
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Claire Troakes
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Glenda M Halliday
- University of Sydney Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences
| | - John R Hodges
- University of Sydney Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences
| | - Jillian J Kril
- University of Sydney Faculty of Medicine and Health School of Medical Sciences
| | - John B Kwok
- University of Sydney Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences
| | - Olivier Piguet
- University of Sydney Brain and Mind Centre and Faculty of Science School of Psychology
| | - Marla Gearing
- Dept. of Pathology and Laboratory Medicine, Dept. of Neurology, and Goizueta Alzheimer’s Disease Center Brain Bank; Emory University School of Medicine, Atlanta, GA USA
| | - Thomas Arzberger
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University Munich, Germany
| | - Sigrun Roeber
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Germany
| | - Johannes Attems
- Newcastle Brain Tissue Resource, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Christopher M Morris
- Newcastle Brain Tissue Resource, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Alan J Thomas
- Newcastle Brain Tissue Resource, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Bret M. Evers
- University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Charles L White
- University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Anne A Sieben
- Laboratory of Neurology, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- IBB-NeuroBiobank BB190113, Born Bunge Institute, Antwerp, Belgium
- Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
- Department of Neurology, Ghent University Hospital, Ghent University, Belgium
| | - Patrick P Cras
- Laboratory of Neurology, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- IBB-NeuroBiobank BB190113, Born Bunge Institute, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital - UZA, Antwerp, Belgium
| | - Bart B De Vil
- Laboratory of Neurology, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- IBB-NeuroBiobank BB190113, Born Bunge Institute, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital - UZA, Antwerp, Belgium
| | - Peter Paul P.P. De Deyn
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Universiteitsplein 1, 2610 Antwerpen, Belgium
| | - Charles Duyckaerts
- Laboratoire de Neuropathologie Escourolle, Hôpital de la Salpêtrière, AP-HP, & Alzheimer Prion Team, ICM, 47 Bd de l’Hôpital, 75651 CEDEX 13 Paris, France
| | - Isabelle Le Ber
- Inserm U1127, CNRS UMR 7225, Sorbonne Université, Paris Brain Institute (ICM), Hôpital Pitié-Salpêtrière, Paris, France
- Centre de référence des démences rares ou précoces, Hôpital Pitié-Salpêtrière, Paris, France
| | - Danielle Seihean
- Laboratoire de Neuropathologie Escourolle, Hôpital de la Salpêtrière, AP-HP, & ICM, 47 Bd de l’Hôpital, 75651 CEDEX 13 Paris, France
| | - Sabrina Turbant-Leclere
- Inserm U1127, CNRS UMR 7225, Sorbonne Université, Paris Brain Institute (ICM) Hôpital Pitié-Salpêtrière, Paris, France
| | - Ian R MacKenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada V6T 2B5
| | - Catriona McLean
- Department of Anatomical Pathology Alfred Heath, Melbourne, Victoria, 3004, Australia
- Victorian Brain Bank, The Florey Institute of Neuroscience of Mental Health, Parkville, Victoria, 3052, Australia
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute and Weill Cornell Medicine, Houston, TX
| | - John F Ervin
- Department of Neurology, Duke University Medical Center, Durham, USA
| | - Shih-Hsiu J Wang
- Department of Neurology, Duke University Medical Center, Durham, USA
| | - Caroline Graff
- Division for Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Inger Nennesmo
- Dept of laboratory Medicine Huddinge Karolinska Institutet, Stockholm Sweden
- Dept of Pathology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Rashed M Nagra
- Human Brain and Spinal Fluid Resource Center, Brentwood Biomedical Research Institute, Los Angeles, CA, United States
| | | | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease (CRND) and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | | | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Manuela Neumann
- Molecular Neuropathology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, University Hospital of Tübingen, Tübingen, Germany
| | - Lee-Cyn Ang
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON, Canada
- Schulich School of Medicine and Dentistry, Western University, London. ON, Canada
| | - Elizabeth C Finger
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Washington, DC, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Dan Vitale
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Washington, DC, USA
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Huw R Morris
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, UK
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- VIBUAntwerp Center for Molecular Neurology, University of Antwerp, Antwerp 2610, Belgium
| | - John A Hardy
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Reta Lila Weston Institute, University College London, Queen Square Institute of Neurology, London, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
14
|
Senkevich K, Bandres-Ciga S, Cisterna-García A, Yu E, Bustos BI, Krohn L, Lubbe SJ, Botía JA, Gan-Or Z. Genome-wide association study stratified by MAPT haplotypes identifies potential novel loci in Parkinson's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.14.23288478. [PMID: 37292720 PMCID: PMC10246147 DOI: 10.1101/2023.04.14.23288478] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective To identify genetic factors that may modify the effects of the MAPT locus in Parkinson's disease (PD). Methods We used data from the International Parkinson's Disease Genomics Consortium (IPDGC) and the UK biobank (UKBB). We stratified the IPDGC cohort for carriers of the H1/H1 genotype (PD patients n=8,492 and controls n=6,765) and carriers of the H2 haplotype (with either H1/H2 or H2/H2 genotypes, patients n=4,779 and controls n=4,849) to perform genome-wide association studies (GWASs). Then, we performed replication analyses in the UKBB data. To study the association of rare variants in the new nominated genes, we performed burden analyses in two cohorts (Accelerating Medicines Partnership - Parkinson Disease and UKBB) with a total sample size PD patients n=2,943 and controls n=18,486. Results We identified a novel locus associated with PD among MAPT H1/H1 carriers near EMP1 (rs56312722, OR=0.88, 95%CI= 0.84-0.92, p= 1.80E-08), and a novel locus associated with PD among MAPT H2 carriers near VANGL1 (rs11590278, OR=1.69 95%CI=1.40-2.03, p=2.72E-08). Similar analysis of the UKBB data did not replicate these results and rs11590278 near VANGL1 did have similar effect size and direction in carriers of H2 haplotype, albeit not statistically significant (OR= 1.32, 95%CI= 0.94-1.86, p=0.17). Rare EMP1 variants with high CADD scores were associated with PD in the MAPT H2 stratified analysis (p=9.46E-05), mainly driven by the p.V11G variant. Interpretation We identified several loci potentially associated with PD stratified by MAPT haplotype and larger replication studies are required to confirm these associations.
Collapse
Affiliation(s)
- Konstantin Senkevich
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and neurosurgery, McGill University, Montréal, QC, Canada, Canada
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Washington DC, USA
| | - Alejandro Cisterna-García
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
| | - Eric Yu
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Bernabe I. Bustos
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lynne Krohn
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Steven J. Lubbe
- Ken and Ruth Davee Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Juan A. Botía
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
| | | | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and neurosurgery, McGill University, Montréal, QC, Canada, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| |
Collapse
|
15
|
Rogers BB, Anderson AG, Lauzon SN, Davis MN, Hauser RM, Roberts SC, Rodriguez-Nunez I, Trausch-Lowther K, Barinaga EA, Taylor JW, Mackiewicz M, Roberts BS, Cooper SJ, Rizzardi LF, Myers RM, Cochran JN. MAPT expression is mediated by long-range interactions with cis-regulatory elements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531520. [PMID: 37090552 PMCID: PMC10120716 DOI: 10.1101/2023.03.07.531520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Background Tauopathies are a group of neurodegenerative diseases driven by abnormal aggregates of tau, a microtubule associated protein encoded by the MAPT gene. MAPT expression is absent in neural progenitor cells (NPCs) and increases during differentiation. This temporally dynamic expression pattern suggests that MAPT expression is controlled by transcription factors and cis-regulatory elements specific to differentiated cell types. Given the relevance of MAPT expression to neurodegeneration pathogenesis, identification of such elements is relevant to understanding genetic risk factors. Methods We performed HiC, chromatin conformation capture (Capture-C), single-nucleus multiomics (RNA-seq+ATAC-seq), bulk ATAC-seq, and ChIP-seq for H3K27Ac and CTCF in NPCs and neurons differentiated from human iPSC cultures. We nominated candidate cis-regulatory elements (cCREs) for MAPT in human NPCs, differentiated neurons, and pure cultures of inhibitory and excitatory neurons. We then assayed these cCREs using luciferase assays and CRISPR interference (CRISPRi) experiments to measure their effects on MAPT expression. Finally, we integrated cCRE annotations into an analysis of genetic variation in AD cases and controls. Results Using orthogonal genomics approaches, we nominated 94 cCREs for MAPT, including the identification of cCREs specifically active in differentiated neurons. Eleven regions enhanced reporter gene transcription in luciferase assays. Using CRISPRi, 5 of the 94 regions tested were identified as necessary for MAPT expression as measured by RT-qPCR and RNA-seq. Rare and predicted damaging genetic variation in both nominated and confirmed CREs was depleted in AD cases relative to controls (OR = 0.40, p = 0.004), consistent with the hypothesis that variants that disrupt MAPT enhancer activity, and thereby reduce MAPT expression, may be protective against neurodegenerative disease. Conclusions We identified both proximal and distal regulatory elements for MAPT and confirmed the regulatory function for several regions, including three regions centromeric to MAPT beyond the well-described H1/H2 haplotype inversion breakpoint. This study provides compelling evidence for pursuing detailed knowledge of CREs for genes of interest to permit better understanding of disease risk.
Collapse
Affiliation(s)
- Brianne B. Rogers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | | | | | | | - Jared W. Taylor
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Mark Mackiewicz
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - Sara J. Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | | | | |
Collapse
|
16
|
Olfati N, Ghodsi H, Bayram E, Litvan I. Why Therapeutic Trials Fail in Primary Tauopathies. Mov Disord 2023; 38:545-550. [PMID: 36670054 PMCID: PMC10398638 DOI: 10.1002/mds.29322] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/08/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Affiliation(s)
- Nahid Olfati
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Hamidreza Ghodsi
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Ece Bayram
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Irene Litvan
- Parkinson and Other Movement Disorders Center, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Tauber CV, Schwarz SC, Rösler TW, Arzberger T, Gentleman S, Windl O, Krumbiegel M, Reis A, Ruf VC, Herms J, Höglinger GU. Different MAPT haplotypes influence expression of total MAPT in postmortem brain tissue. Acta Neuropathol Commun 2023; 11:40. [PMID: 36906636 PMCID: PMC10008602 DOI: 10.1186/s40478-023-01534-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/21/2023] [Indexed: 03/13/2023] Open
Abstract
The MAPT gene, encoding the microtubule-associated protein tau on chromosome 17q21.31, is result of an inversion polymorphism, leading to two allelic variants (H1 and H2). Homozygosity for the more common haplotype H1 is associated with an increased risk for several tauopathies, but also for the synucleinopathy Parkinson's disease (PD). In the present study, we aimed to clarify whether the MAPT haplotype influences expression of MAPT and SNCA, encoding the protein α-synuclein (α-syn), on mRNA and protein levels in postmortem brains of PD patients and controls. We also investigated mRNA expression of several other MAPT haplotype-encoded genes. Postmortem tissues from cortex of fusiform gyrus (ctx-fg) and of the cerebellar hemisphere (ctx-cbl) of neuropathologically confirmed PD patients (n = 95) and age- and sex-matched controls (n = 81) were MAPT haplotype genotyped to identify cases homozygous for either H1 or H2. Relative expression of genes was quantified using real-time qPCR; soluble and insoluble protein levels of tau and α-syn were determined by Western blotting. Homozygosity for H1 versus H2 was associated with increased total MAPT mRNA expression in ctx-fg regardless of disease state. Inversely, H2 homozygosity was associated with markedly increased expression of the corresponding antisense MAPT-AS1 in ctx-cbl. PD patients had higher levels of insoluble 0N3R and 1N4R tau isoforms regardless of the MAPT genotype. The increased presence of insoluble α-syn in PD patients in ctx-fg validated the selected postmortem brain tissue. Our findings in this small, but well controlled cohort of PD and controls support a putative biological relevance of tau in PD. However, we did not identify any link between the disease-predisposing H1/H1 associated overexpression of MAPT with PD status. Further studies are required to gain a deeper understanding of the potential regulatory role of MAPT-AS1 and its association to the disease-protective H2/H2 condition in the context of PD.
Collapse
Affiliation(s)
- Christina V Tauber
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, School of Medicine, Technical University Munich, Munich, Germany.,Department of Obstetrics and Gynecology, Ludiwgs-Maximilians University of Munich, Munich, Germany
| | - Sigrid C Schwarz
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas W Rösler
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, School of Medicine, Technical University Munich, Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany.,Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Steve Gentleman
- Parkinson's UK Brain Bank, Department of Brain Sciences, Imperial College London, London, UK.,Neuropathology Unit, Department of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Otto Windl
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Mandy Krumbiegel
- Institute of Human Genetics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Viktoria C Ruf
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Günter U Höglinger
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany. .,Department of Neurology, Ludwig-Maximilians University of Munich, Munich, Germany.
| |
Collapse
|
18
|
Tseng FS, Foo JQX, Mai AS, Tan EK. The genetic basis of multiple system atrophy. J Transl Med 2023; 21:104. [PMID: 36765380 PMCID: PMC9912584 DOI: 10.1186/s12967-023-03905-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/19/2023] [Indexed: 02/12/2023] Open
Abstract
Multiple system atrophy (MSA) is a heterogenous, uniformly fatal neurodegenerative ɑ-synucleinopathy. Patients present with varying degrees of dysautonomia, parkinsonism, cerebellar dysfunction, and corticospinal degeneration. The underlying pathophysiology is postulated to arise from aberrant ɑ-synuclein deposition, mitochondrial dysfunction, oxidative stress and neuroinflammation. Although MSA is regarded as a primarily sporadic disease, there is a possible genetic component that is poorly understood. This review summarizes current literature on genetic risk factors and potential pathogenic genes and loci linked to both sporadic and familial MSA, and underlines the biological mechanisms that support the role of genetics in MSA. We discuss a broad range of genes that have been associated with MSA including genes related to Parkinson's disease (PD), oxidative stress, inflammation, and tandem gene repeat expansions, among several others. Furthermore, we highlight various genetic polymorphisms that modulate MSA risk, including complex gene-gene and gene-environment interactions, which influence the disease phenotype and have clinical significance in both presentation and prognosis. Deciphering the exact mechanism of how MSA can result from genetic aberrations in both experimental and clinical models will facilitate the identification of novel pathophysiologic clues, and pave the way for translational research into the development of disease-modifying therapeutic targets.
Collapse
Affiliation(s)
- Fan Shuen Tseng
- grid.163555.10000 0000 9486 5048Division of Medicine, Singapore General Hospital, Singapore, Singapore
| | - Joel Qi Xuan Foo
- grid.276809.20000 0004 0636 696XDepartment of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Aaron Shengting Mai
- grid.4280.e0000 0001 2180 6431Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, 169856, Singapore. .,Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
19
|
Kawles A, Minogue G, Zouridakis A, Keszycki R, Gill N, Nassif C, Coventry C, Zhang H, Rogalski E, Flanagan ME, Castellani R, Bigio EH, Mesulam MM, Geula C, Gefen T. Differential vulnerability of the dentate gyrus to tauopathies in dementias. Acta Neuropathol Commun 2023; 11:1. [PMID: 36597124 PMCID: PMC9811688 DOI: 10.1186/s40478-022-01485-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Abstract
The dentate gyrus (DG), a key hippocampal subregion in memory processing, generally resists phosphorylated tau accumulation in the amnestic dementia of the Alzheimer's type due to Alzheimer's disease (DAT-AD), but less is known about the susceptibility of the DG to other tauopathies. Here, we report stereologic densities of total DG neurons and tau inclusions in thirty-two brains of human participants with autopsy-confirmed tauopathies with distinct isoform profiles-3R Pick's disease (PiD, N = 8), 4R corticobasal degeneration (CBD, N = 8), 4R progressive supranuclear palsy (PSP, N = 8), and 3/4R AD (N = 8). All participants were diagnosed during life with primary progressive aphasia (PPA), an aphasic clinical dementia syndrome characterized by progressive deterioration of language abilities with spared non-language cognitive abilities in early stages, except for five patients with DAT-AD as a comparison group. 51% of total participants were female. All specimens were stained immunohistochemically with AT8 to visualize tau pathology, and PPA cases were stained for Nissl substance to visualize neurons. Unbiased stereological analysis was performed in granule and hilar DG cells, and inclusion-to-neuron ratios were calculated. In the PPA group, PiD had highest mean total (granule + hilar) densities of DG tau pathology (p < 0.001), followed by CBD, AD, then PSP. PPA-AD cases showed more inclusions in hilar cells compared to granule cells, while the opposite was true in PiD and CBD. Inclusion-to-neuron ratios revealed, on average, 33% of all DG neurons in PiD cases contained a tau inclusion, compared to ~ 7% in CBD, 2% in AD, and 0.4% in PSP. There was no significant difference between DAT-AD and PPA-AD pathologic tau burden, suggesting that differences in DG burden are not specific to clinical phenotype. We conclude that the DG is differentially vulnerable to pathologic tau accumulation, raising intriguing questions about the structural integrity and functional significance of hippocampal circuits in neurodegenerative dementias.
Collapse
Affiliation(s)
- Allegra Kawles
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Grace Minogue
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Antonia Zouridakis
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Rachel Keszycki
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Nathan Gill
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Caren Nassif
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Margaret E. Flanagan
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Rudolph Castellani
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Eileen H. Bigio
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - M. Marsel Mesulam
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| |
Collapse
|
20
|
Długosz-Pokorska A, Perlikowska R, Janecki T, Janecka A. New Uracil Analog with Exocyclic Methylidene Group Can Reverse Resistance to Taxol in MCF-7 Cancer Cells. Biologics 2023; 17:69-83. [PMID: 37213261 PMCID: PMC10198386 DOI: 10.2147/btt.s405080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/10/2023] [Indexed: 05/23/2023]
Abstract
Introduction Taxol (Tx), a microtubule-stabilizing drug, has been widely used as a chemotherapeutic in several types of cancer. However, the development of resistance limited its application. One of the strategies used to prevent the emergence of drug resistance is combination treatment, involving at least two drugs. The aim of the current study was to assess if a new uracil analog, 3-p-bromophenyl-1-ethyl-5-methylidenedihydrouracil (U-359) can prevent the development of Tx resistance in breast cancer cells. Methods The cytotoxicity of the new drug was tested in MCF-7 (hormone receptor (ER, PR) positive cell-line) and MCF-10A cell lines using MTT method. For the detection of apoptosis and necrosis, the Wright and Giemsa staining was used. Gene expression was measured by real-time PCR, and changes in the protein levels were evaluated by ELISA and bioluminescent method. Results We investigated the effect of Tx and U-359 on cancer MCF-7 and normal MCF-10A cells alone and in combination. Tx co-administered with U-359 inhibited proliferation of MCF-7 cells to 7% while the level of ATPase drastically decreased to 14%, compared with effects produced by Tx alone. The apoptosis process was induced through the mitochondrial pathway. These effects were not seen in MCF-10A cells, showing the wide safety margin. The obtained results have shown that U-359 produced a synergistic effect with Tx probably by reducing Tx resistance in MCF-7 cells. To elucidate the possible mechanism of resistance, expression of tubulin III (TUBIII), responsible for microtubule stabilization and tau and Nlp proteins, responsible for microtubule dynamics, was assessed. Conclusion Combination of Tx with U-359 reduced overexpression of TUBIII and Nlp. Thus, U-359 may stand for a potential reversal agent for the treatment of MDR in cancer cells.
Collapse
Affiliation(s)
- Angelika Długosz-Pokorska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
- Correspondence: Angelika Długosz-Pokorska, Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, Lodz, 92-215, Poland, Tel +48 42 2725706, Fax +48 42 678 42 77, Email
| | - Renata Perlikowska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Tomasz Janecki
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
21
|
Campoy E, Puig M, Yakymenko I, Lerga-Jaso J, Cáceres M. Genomic architecture and functional effects of potential human inversion supergenes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210209. [PMID: 35694745 PMCID: PMC9189494 DOI: 10.1098/rstb.2021.0209] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Supergenes are involved in adaptation in multiple organisms, but they are little known in humans. Genomic inversions are the most common mechanism of supergene generation and maintenance. Here, we review the information about two large inversions that are the best examples of potential human supergenes. In addition, we do an integrative analysis of the newest data to understand better their functional effects and underlying genetic changes. We have found that the highly divergent haplotypes of the 17q21.31 inversion of approximately 1.5 Mb have multiple phenotypic associations, with consistent effects in brain-related traits, red and white blood cells, lung function, male and female characteristics and disease risk. By combining gene expression and nucleotide variation data, we also analysed the molecular differences between haplotypes, including gene duplications, amino acid substitutions and regulatory changes, and identify CRHR1, KANLS1 and MAPT as good candidates to be responsible for these phenotypes. The situation is more complex for the 8p23.1 inversion, where there is no clear genetic differentiation. However, the inversion is associated with several related phenotypes and gene expression differences that could be linked to haplotypes specific of one orientation. Our work, therefore, contributes to the characterization of both exceptional variants and illustrates the important role of inversions. This article is part of the theme issue 'Genomic architecture of supergenes: causes and evolutionary consequences'.
Collapse
Affiliation(s)
- Elena Campoy
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Marta Puig
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Illya Yakymenko
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Jon Lerga-Jaso
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Mario Cáceres
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
22
|
Gallo D, Ruiz A, Sánchez-Juan P. Genetic architecture of primary tauopathies. Neuroscience 2022; 518:27-37. [PMID: 35609758 DOI: 10.1016/j.neuroscience.2022.05.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/29/2022] [Accepted: 05/17/2022] [Indexed: 11/26/2022]
Abstract
Primary Tauopathies are a group of diseases defined by the accumulation of Tau, in which the alteration of this protein is the primary driver of the neurodegenerative process. In addition to the classical syndromes (Pick's disease (PiD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and argyrophilic grain disease (AGD)), new entities, like primary age-related Tauopathy (PART), have been recently described. Except for the classical Richardson's syndrome phenotype in PSP, the correlation between the clinical picture of the primary Tauopathies and underlying pathology is poor. This fact has challenged genetic studies. However, thanks to multicenter collaborations, several genome-wide association studies are helping us unravel the genetic structure of these diseases. The most relevant risk factor revealed by these studies is the Tau gene (MAPT), which, in addition to mutations causing rare familial forms, plays a fundamental role in sporadic cases of PSP and CBD in which there is a strong predominance of the H1 and H1c haplotypes. But outside of MAPT, several other genes have been robustly associated with PSP. These findings, pointing towards multifactorial causation, imply the participation of several pathways involving the myelin sheath integrity, the endoplasmic reticulum unfolded protein response, microglia, intracellular vesicle trafficking, or the ubiquitin-proteasome system. Additionally, GWAS show a high degree of genetic overlap across different Tauopathies. This is especially salient between PSP and CBD, but also GWAS studying the recently described PART phenotype shows genetic overlap with genes that promote Tau pathology and with others associated with Alzheimer's disease.
Collapse
|
23
|
Fray S, Achouri-Rassas AA, Hadj Fredj S, Messaoud T, Belal S. Association between H2 haplotype of microtubule associated protein tau gene (deletion / insertion) with Alzheimer Disease in Tunisian patients. Neurol Res 2022; 44:814-818. [PMID: 35348036 DOI: 10.1080/01616412.2022.2056338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
It is widely recognized that Alzheimer's disease (AD) is the main cause of dementia in the elderly. AD is typically characterized by the extraneuronal plaque made up essentially of the amyloid β peptide and intraneuronal tangles of hyperphosphorylated microtubule-associated Tau protein. This study investigates the possible interaction between AD and the deletion/insertion polymorphism in intron 9 of the Tau gene haplotype and APOE state in a Tunisian AD cases population (n = 85) and control (n = 91). The H2/H2 genotype was higher in the AD group as compared to the controls (22.4% vs. 7.8%). The frequency of H2 allele is higher in the patients group, and the difference of allele frequency is statistically significant between the two groups (χ2 = 12.220, p < 0.05). H2 allele is correlated with the female gender within the patient group (χ2 = 7.649, p = 0.006) Tau H2 haplotype can be identified as a risk factor of AD in the studied Tunisian population and was associated to female gender. There is no significant correlation between the frequency of Tau gene ins/del polymorphism and cognitive profile distribution in the patient group (p > 0.05).
Collapse
Affiliation(s)
- Saloua Fray
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia.,Department of Neurology, Charles Nicolle Hospital, Tunis, Tunisia.,Children Hospital, Biochemistry and Molecular Biology Laboratory, Tunis, Tunisia
| | - Afef Achouri Achouri-Rassas
- Department of Neurology, Charles Nicolle Hospital, Tunis, Tunisia.,Neurosciences Project, Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Sondes Hadj Fredj
- Children Hospital, Biochemistry and Molecular Biology Laboratory, Tunis, Tunisia
| | - Taieb Messaoud
- Children Hospital, Biochemistry and Molecular Biology Laboratory, Tunis, Tunisia
| | - Samir Belal
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia.,Department of Neurology, National Institute Mongi Ben Hmida of Neurology, Tunis, Tunisia.,Molecular Neurobiology and Neuropathology Research laboratory, Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
24
|
Rabaneda-Bueno R, Mena-Montes B, Torres-Castro S, Torres-Carrillo N, Torres-Carrillo NM. Advances in Genetics and Epigenetic Alterations in Alzheimer's Disease: A Notion for Therapeutic Treatment. Genes (Basel) 2021; 12:1959. [PMID: 34946908 PMCID: PMC8700838 DOI: 10.3390/genes12121959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a disabling neurodegenerative disorder that leads to long-term functional and cognitive impairment and greatly reduces life expectancy. Early genetic studies focused on tracking variations in genome-wide DNA sequences discovered several polymorphisms and novel susceptibility genes associated with AD. However, despite the numerous risk factors already identified, there is still no fully satisfactory explanation for the mechanisms underlying the onset of the disease. Also, as with other complex human diseases, the causes of low heritability are unclear. Epigenetic mechanisms, in which changes in gene expression do not depend on changes in genotype, have attracted considerable attention in recent years and are key to understanding the processes that influence age-related changes and various neurological diseases. With the recent use of massive sequencing techniques, methods for studying epigenome variations in AD have also evolved tremendously, allowing the discovery of differentially expressed disease traits under different conditions and experimental settings. This is important for understanding disease development and for unlocking new potential AD therapies. In this work, we outline the genomic and epigenomic components involved in the initiation and development of AD and identify potentially effective therapeutic targets for disease control.
Collapse
Affiliation(s)
- Rubén Rabaneda-Bueno
- Biology Centre of the Czech Academy of Sciences, Institute of Hydrobiology, 37005 České Budějovice, Czech Republic
- School of Biological Sciences, James Clerk Maxwell Building, The King’s Buildings Campus, University of Edinburgh, Edinburgh EH9 3FD, UK
| | - Beatriz Mena-Montes
- Laboratorio de Biología del Envejecimiento, Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City 10200, Mexico;
| | - Sara Torres-Castro
- Departamento de Epidemiología Demográfica y Determinantes Sociales, Instituto Nacional de Geriatría, Mexico City 10200, Mexico;
| | - Norma Torres-Carrillo
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.T.-C.); (N.M.T.-C.)
| | - Nora Magdalena Torres-Carrillo
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.T.-C.); (N.M.T.-C.)
| |
Collapse
|
25
|
Stamelou M, Respondek G, Giagkou N, Whitwell JL, Kovacs GG, Höglinger GU. Evolving concepts in progressive supranuclear palsy and other 4-repeat tauopathies. Nat Rev Neurol 2021; 17:601-620. [PMID: 34426686 DOI: 10.1038/s41582-021-00541-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Tauopathies are classified according to whether tau deposits predominantly contain tau isoforms with three or four repeats of the microtubule-binding domain. Those in which four-repeat (4R) tau predominates are known as 4R-tauopathies, and include progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, globular glial tauopathies and conditions associated with specific MAPT mutations. In these diseases, 4R-tau deposits are found in various cell types and anatomical regions of the brain and the conditions share pathological, pathophysiological and clinical characteristics. Despite being considered 'prototype' tauopathies and, therefore, ideal for studying neuroprotective agents, 4R-tauopathies are still severe and untreatable diseases for which no validated biomarkers exist. However, advances in research have addressed the issues of phenotypic overlap, early clinical diagnosis, pathophysiology and identification of biomarkers, setting a road map towards development of treatments. New clinical criteria have been developed and large cohorts with early disease are being followed up in prospective studies. New clinical trial readouts are emerging and biomarker research is focused on molecular pathways that have been identified. Lessons learned from failed trials of neuroprotective drugs are being used to design new trials. In this Review, we present an overview of the latest research in 4R-tauopathies, with a focus on progressive supranuclear palsy, and discuss how current evidence dictates ongoing and future research goals.
Collapse
Affiliation(s)
- Maria Stamelou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece. .,European University of Cyprus, Nicosia, Cyprus. .,Philipps University, Marburg, Germany.
| | - Gesine Respondek
- Department of Neurology, Hanover Medical School, Hanover, Germany
| | - Nikolaos Giagkou
- Parkinson's Disease and Movement Disorders Dept, HYGEIA Hospital, Athens, Greece
| | | | - Gabor G Kovacs
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Toronto, Ontario, Canada.,Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Günter U Höglinger
- Department of Neurology, Hanover Medical School, Hanover, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
26
|
Leveille E, Ross OA, Gan-Or Z. Tau and MAPT genetics in tauopathies and synucleinopathies. Parkinsonism Relat Disord 2021; 90:142-154. [PMID: 34593302 PMCID: PMC9310195 DOI: 10.1016/j.parkreldis.2021.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/25/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
MAPT encodes the microtubule-associated protein tau, which is the main component of neurofibrillary tangles (NFTs) and found in other protein aggregates. These aggregates are among the pathological hallmarks of primary tauopathies such as frontotemporal dementia (FTD). Abnormal tau can also be observed in secondary tauopathies such as Alzheimer's disease (AD) and synucleinopathies such as Parkinson's disease (PD). On top of pathological findings, genetic data also links MAPT to these disorders. MAPT variations are a cause or risk factors for many tauopathies and synucleinopathies and are associated with certain clinical and pathological features in affected individuals. In addition to clinical, pathological, and genetic overlap, evidence also suggests that tau and alpha-synuclein may interact on the molecular level, and thus might collaborate in the neurodegenerative process. Understanding the role of MAPT variations in tauopathies and synucleinopathies is therefore essential to elucidate the role of tau in the pathogenesis and phenotype of those disorders, and ultimately to develop targeted therapies. In this review, we describe the role of MAPT genetic variations in tauopathies and synucleinopathies, several genotype-phenotype and pathological features, and discuss their implications for the classification and treatment of those disorders.
Collapse
Affiliation(s)
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-hospital), McGill University, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada.
| |
Collapse
|
27
|
Strauß T, Marvian-Tayaranian A, Sadikoglou E, Dhingra A, Wegner F, Trümbach D, Wurst W, Heutink P, Schwarz SC, Höglinger GU. iPS Cell-Based Model for MAPT Haplotype as a Risk Factor for Human Tauopathies Identifies No Major Differences in TAU Expression. Front Cell Dev Biol 2021; 9:726866. [PMID: 34532319 PMCID: PMC8438159 DOI: 10.3389/fcell.2021.726866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
The H1 haplotype of the microtubule-associated protein tau (MAPT) gene is a common genetic risk factor for some neurodegenerative diseases such as progressive supranuclear palsy, corticobasal degeneration, and Parkinson's disease. The molecular mechanism causing the increased risk for the named diseases, however, remains unclear. In this paper, we present a valuable tool of eight small molecule neural precursor cell lines (smNPC) homozygous for the MAPT haplotypes (four H1/H1 and four H2/H2 cell lines), which can be used to identify MAPT-dependent phenotypes. The employed differentiation protocol is fast due to overexpression of NEUROGENIN-2 and therefore suitable for high-throughput approaches. A basic characterization of all human cell lines was performed, and their TAU and α-SYNUCLEIN profiles were compared during a differentiation time of 30 days. We could identify higher levels of conformationally altered TAU in cell lines carrying the H2 haplotype. Additionally, we found increased expression levels of α-SYNUCLEIN in H1/H1 cells. With this resource, we aim to fill a gap in neurodegenerative disease modeling with induced pluripotent stem cells (iPSC) for sporadic tauopathies.
Collapse
Affiliation(s)
- Tabea Strauß
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technical University Munich, Munich, Germany
| | - Amir Marvian-Tayaranian
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technical University Munich, Munich, Germany
| | - Eldem Sadikoglou
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ashutosh Dhingra
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Florian Wegner
- Department of Neurology, Hanover Medical School, Hanover, Germany
- Center for Systems Neuroscience, Hanover, Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Oberschleißheim, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, Oberschleißheim, Germany
- TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Sigrid C. Schwarz
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technical University Munich, Munich, Germany
- Geriatric Clinic Haag, Haag in Oberbayern, Germany
| | - Günter U. Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technical University Munich, Munich, Germany
- Department of Neurology, Hanover Medical School, Hanover, Germany
- Center for Systems Neuroscience, Hanover, Germany
| |
Collapse
|
28
|
Kim SH, Farrell K, Cosentino S, Vonsattel JPG, Faust PL, Cortes EP, Bennet DA, Louis ED, Crary JF. Tau Isoform Profile in Essential Tremor Diverges From Other Tauopathies. J Neuropathol Exp Neurol 2021; 80:835-843. [PMID: 34363663 DOI: 10.1093/jnen/nlab073] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Patients with essential tremor (ET) frequently develop concurrent dementia, which is often assumed to represent co-morbid Alzheimer disease (AD). Autopsy studies have identified a spectrum of tau pathologies in ET and tau isoforms have not been examined in ET. We performed immunoblotting using autopsy cerebral cortical tissue from patients with ET (n = 13), progressive supranuclear palsy ([PSP], n = 10), Pick disease ([PiD], n = 2), and AD (n = 7). Total tau in ET samples was similar to that in PSP and PiD but was significantly lower than that in AD. Abnormal tau levels measured using the AT8 phospho-tau specific (S202/T205/S208) monoclonal antibody in ET were similar to those in PSP but were lower than in PiD and AD. In aggregates, tau with 3 microtubule-binding domain repeats (3R) was significantly higher in AD than ET, while tau with 4 repeats (4R) was significantly higher in PSP. Strikingly, the total tau without N-terminal inserts in ET was significantly lower than in PSP, PiD, and AD, but total tau with other N-terminal inserts was not. Monomeric tau with one insert in ET was similar to that in PSP and PiD was lower than in AD. Thus, ET brains exhibit an expression profile of tau protein isoforms that diverges from that of other tauopathies.
Collapse
Affiliation(s)
- Soong Ho Kim
- From the Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, JFC).,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC)
| | - Kurt Farrell
- From the Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC)
| | - Stephanie Cosentino
- Cognitive Neuroscience Division, Department of Neurology, Columbia University Medical Center, New York, New York, USA (SC); G.H. Sergievsky Center, Columbia University Medical Center, New York, New York, USA (SC, JPV).,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA (SC, JPV)
| | - Jean-Paul G Vonsattel
- Cognitive Neuroscience Division, Department of Neurology, Columbia University Medical Center, New York, New York, USA (SC); G.H. Sergievsky Center, Columbia University Medical Center, New York, New York, USA (SC, JPV).,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA (SC, JPV).,Department of Pathology & Cell Biology, Columbia University Medical Center and the New York Presbyterian Hospital, New York, New York, USA (JPV, PLF)
| | - Phyllis L Faust
- Department of Pathology & Cell Biology, Columbia University Medical Center and the New York Presbyterian Hospital, New York, New York, USA (JPV, PLF)
| | - Etty P Cortes
- From the Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA (EC, JFC)
| | - David A Bennet
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA (DAB)
| | - Elan D Louis
- Department of Neurology, University of Texas Southwestern, Dallas, Texas, USA (EDL)
| | - John F Crary
- From the Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, JFC).,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA (SHK, KF, EC, JFC).,Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA (EC, JFC)
| |
Collapse
|
29
|
Wallon D, Boluda S, Rovelet-Lecrux A, Thierry M, Lagarde J, Miguel L, Lecourtois M, Bonnevalle A, Sarazin M, Bottlaender M, Mula M, Marty S, Nakamura N, Schramm C, Sellal F, Jonveaux T, Heitz C, Le Ber I, Epelbaum S, Magnin E, Zarea A, Rousseau S, Quenez O, Hannequin D, Clavaguera F, Campion D, Duyckaerts C, Nicolas G. Clinical and neuropathological diversity of tauopathy in MAPT duplication carriers. Acta Neuropathol 2021; 142:259-278. [PMID: 34095977 DOI: 10.1007/s00401-021-02320-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 11/30/2022]
Abstract
Microduplications of the 17q21.31 chromosomal region encompassing the MAPT gene, which encodes the Tau protein, were identified in patients with a progressive disorder initially characterized by severe memory impairment with or without behavioral changes that can clinically mimic Alzheimer disease. The unique neuropathological report showed a primary tauopathy, which could not be unanimously classified in a given known subtype, showing both 4R- and 3R-tau inclusions, mainly within temporal cortical subregions and basal ganglia, without amyloid deposits. Recently, two subjects harboring the same duplication were reported with an atypical extrapyramidal syndrome and gait disorder. To decipher the phenotypic spectrum associated with MAPT duplications, we studied ten carriers from nine families, including two novel unrelated probands, gathering clinical (n = 10), cerebrospinal fluid (n = 6), MRI (n = 8), dopamine transporter scan (n = 4), functional (n = 5), amyloid (n = 3) and Tau-tracer (n = 2) PET imaging data as well as neuropathological examination (n = 4). Ages at onset ranged from 37 to 57 years, with prominent episodic memory impairment in 8/10 patients, associated with behavioral changes in four, while two patients showed atypical extrapyramidal syndrome with gait disorder at presentation, including one with associated cognitive deficits. Amyloid imaging was negative but Tau imaging showed significant deposits mainly in both mesiotemporal cortex. Dopaminergic denervation was found in 4/4 patients, including three without extrapyramidal symptoms. Neuropathological examination exclusively showed Tau-immunoreactive lesions. Distribution, aspect and 4R/3R tau aggregates composition suggested a spectrum from predominantly 3R, mainly cortical deposits well correlating with cognitive and behavioral changes, to predominantly 4R deposits, mainly in the basal ganglia and midbrain, in patients with prominent extrapyramidal syndrome. Finally, we performed in vitro seeding experiments in HEK-biosensor cells. Morphological features of aggregates induced by homogenates of three MAPT duplication carriers showed dense/granular ratios graduating between those induced by homogenates of a Pick disease and a progressive supranuclear palsy cases. These results suggest that MAPT duplication causes a primary tauopathy associated with diverse clinical and neuropathological features.
Collapse
Affiliation(s)
- David Wallon
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Neurology and CNR-MAJ, F-76000, Rouen, France.
| | - Susana Boluda
- Sorbonne Université, INSERM, CNRS U1127, Institut du Cerveau, ICM, Paris, France
- AP-HP, Hôpital de La Pitié-Salpêtrière, Laboratoire de Neuropathologie R. Escourolle, Paris, France
| | - Anne Rovelet-Lecrux
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France
| | - Manon Thierry
- Sorbonne Université, INSERM, CNRS U1127, Institut du Cerveau, ICM, Paris, France
- AP-HP, Hôpital de La Pitié-Salpêtrière, Laboratoire de Neuropathologie R. Escourolle, Paris, France
| | - Julien Lagarde
- Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
- Université de Paris, 75006, Paris, France
- Université Paris-Saclay, BioMaps, Service Hospitalier Frederic Joliot, CEA, CNRS, Inserm, F-91401, Orsay, France
| | - Laetitia Miguel
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France
| | - Magalie Lecourtois
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France
| | - Antoine Bonnevalle
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Neurology and CNR-MAJ, F-76000, Rouen, France
| | - Marie Sarazin
- Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte Anne, F-75014, Paris, France
- Université de Paris, 75006, Paris, France
- Université Paris-Saclay, BioMaps, Service Hospitalier Frederic Joliot, CEA, CNRS, Inserm, F-91401, Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, BioMaps, Service Hospitalier Frederic Joliot, CEA, CNRS, Inserm, F-91401, Orsay, France
- UNIACT, Neurospin, CEA, 91191, Gif-sur-Yvette, France
| | - Mathieu Mula
- AP-HP, Hôpital de La Pitié-Salpêtrière, Laboratoire de Neuropathologie R. Escourolle, Paris, France
| | - Serge Marty
- Sorbonne Université, INSERM, CNRS U1127, Institut du Cerveau, ICM, Paris, France
| | - Natsuko Nakamura
- Sorbonne Université, INSERM, CNRS U1127, Institut du Cerveau, ICM, Paris, France
| | - Catherine Schramm
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France
| | - François Sellal
- Department of Neurology, Hôpitaux Civils de Colmar and INSERM U-1118, School of Medicine, Strasbourg University, Strasbourg, France
| | - Thérèse Jonveaux
- CMRR Department of Neurology, Nancy University Hospital, Laboratoire Lorraine de Psychologie et de Neurosciences de la Dynamique des Comportements 2LPN EA7489 Lorraine University, Nancy, France
| | - Camille Heitz
- Neurology Department, Hôpital Universitaire de Nîmes, Nîmes, France
| | - Isabelle Le Ber
- Sorbonne Universités, UPMC Univ Paris 06, Inserm U1127, CNRS UMR 7225, Institut du Cerveau et la Moelle Epinière (ICM), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
- Reference Centre for Rare or Early Dementias, IM2A, Département de Neurologie, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Stéphane Epelbaum
- Centre Mémoire Ressources et Recherche (CMRR), Centre Expert Parkinson (CEP), Service de Neurologie, CHRU Besançon, 25000, Besançon, France
- Neurosciences Intégratives et Cliniques UR481, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Eloi Magnin
- Centre Mémoire Ressources et Recherche (CMRR), Centre Expert Parkinson (CEP), Service de Neurologie, CHRU Besançon, 25000, Besançon, France
- Neurosciences Intégratives et Cliniques UR481, Univ. Bourgogne Franche-Comté, 25000, Besançon, France
| | - Aline Zarea
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Neurology and CNR-MAJ, F-76000, Rouen, France
| | - Stéphane Rousseau
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France
| | - Olivier Quenez
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France
| | - Didier Hannequin
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Neurology and CNR-MAJ, F-76000, Rouen, France
| | - Florence Clavaguera
- Sorbonne Université, INSERM, CNRS U1127, Institut du Cerveau, ICM, Paris, France
| | - Dominique Campion
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France
| | - Charles Duyckaerts
- Sorbonne Université, INSERM, CNRS U1127, Institut du Cerveau, ICM, Paris, France
- AP-HP, Hôpital de La Pitié-Salpêtrière, Laboratoire de Neuropathologie R. Escourolle, Paris, France
| | - Gaël Nicolas
- Normandie Univ, UNIROUEN, Inserm U1245, CHU Rouen, Department of Genetics and CNR-MAJ, F-76000, Rouen, France.
| |
Collapse
|
30
|
Robert A, Schöll M, Vogels T. Tau Seeding Mouse Models with Patient Brain-Derived Aggregates. Int J Mol Sci 2021; 22:6132. [PMID: 34200180 PMCID: PMC8201271 DOI: 10.3390/ijms22116132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/23/2022] Open
Abstract
Tauopathies are a heterogeneous class of neurodegenerative diseases characterized by intracellular inclusions of aggregated tau proteins. Tau aggregates in different tauopathies have distinct structural features and can be found in different cell types. Transgenic animal models overexpressing human tau have been used for over two decades in the research of tau pathology. However, these models poorly recapitulate the heterogeneity of tauopathies found in human brains. Recent findings demonstrate that injection of purified tau aggregates from the brains of human tauopathy patients recapitulates both the structural features and cell-type specificity of the tau pathology of the donor tauopathy. These models may therefore have unique translational value in the study of functional consequences of tau pathology, tau-based diagnostics, and tau targeting therapeutics. This review provides an update of the literature relating to seeding-based tauopathy and their potential applications.
Collapse
Affiliation(s)
- Aiko Robert
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London WC1N 3BG, UK; (A.R.); (M.S.)
| | - Michael Schöll
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London WC1N 3BG, UK; (A.R.); (M.S.)
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, 413 45 Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Thomas Vogels
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London WC1N 3BG, UK; (A.R.); (M.S.)
- Department of Psychiatry and Neurochemistry, University of Gothenburg, 413 45 Gothenburg, Sweden
- Sylics (Synaptologics B.V.), 3721 MA Bilthoven, The Netherlands
| |
Collapse
|
31
|
Kouri N, Murray ME, Reddy JS, Serie DJ, Soto-Beasley A, Allen M, Carrasquillo MM, Wang X, Castanedes MC, Baker MC, Rademakers R, Uitti RJ, Graff-Radford NR, Wszolek ZK, Schellenberg GD, Crook JE, Ertekin-Taner N, Ross OA, Dickson DW. Latent trait modeling of tau neuropathology in progressive supranuclear palsy. Acta Neuropathol 2021; 141:667-680. [PMID: 33635380 PMCID: PMC8043857 DOI: 10.1007/s00401-021-02289-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 11/01/2022]
Abstract
Progressive supranuclear palsy (PSP) is the second most common neurodegenerative Parkinsonian disorder after Parkinson's disease, and is characterized as a primary tauopathy. Leveraging the considerable clinical and neuropathologic heterogeneity associated with PSP, we measured tau neuropathology as quantitative traits to perform a genome-wide association study (GWAS) within PSP to identify genes and biological pathways that underlie the PSP disease process. In 882 PSP cases, semi-quantitative scores for phosphorylated tau-immunoreactive coiled bodies (CBs), neurofibrillary tangles (NFTs), tufted astrocytes (TAs), and tau threads were documented from 18 brain regions, and converted to latent trait (LT) variables using the R ltm package. LT analysis utilizes a multivariate regression model that links categorical responses to unobserved covariates allowing for a reduction of dimensionality, generating a single, continuous variable to account for the multiple lesions and brain regions assessed. We first tested for association with PSP LTs and the top PSP GWAS susceptibility loci. Significant SNP/LT associations were identified at rs242557 (MAPT H1c sub-haplotype) with hindbrain CBs and rs1768208 (MOBP) with forebrain tau threads. Digital microscopy was employed to quantify phosphorylated tau burden in midbrain tectum and red nucleus in 795 PSP cases and tau burdens were used as quantitative phenotypes in GWAS. Top associations were identified at rs1768208 with midbrain tectum and red nucleus tau burden. Additionally, we performed a PSP LT GWAS on an initial cohort, a follow-up SNP panel (37 SNPs, P < 10-5) in an extended cohort, and a combined analysis. Top SNP/LT associations were identified at SNPs in or near SPTBN5/EHD4, SEC13/ATP2B2, EPHB1/PPP2R3A, TBC1D8, IFNGR1/OLIG3, ST6GAL1, HK1, CALB1, and SGCZ. Finally, testing for SNP/transcript associations using whole transcriptome and whole genome data identified significant expression quantitative trait loci at rs3088159/SPTBN5/EHD4 and rs154239/GHRL. Modeling tau neuropathology heterogeneity using LTs as quantitative phenotypes in a GWAS may provide substantial insight into biological pathways involved in PSP by affecting regional tau burden.
Collapse
Affiliation(s)
- Naomi Kouri
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Joseph S Reddy
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Daniel J Serie
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Alexandra Soto-Beasley
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Minerva M Carrasquillo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Xue Wang
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Matthew C Baker
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium
| | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julia E Crook
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
32
|
Arienti F, Lazzeri G, Vizziello M, Monfrini E, Bresolin N, Saetti MC, Picillo M, Franco G, Di Fonzo A. Unravelling Genetic Factors Underlying Corticobasal Syndrome: A Systematic Review. Cells 2021; 10:171. [PMID: 33467748 PMCID: PMC7830591 DOI: 10.3390/cells10010171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Corticobasal syndrome (CBS) is an atypical parkinsonian presentation characterized by heterogeneous clinical features and different underlying neuropathology. Most CBS cases are sporadic; nevertheless, reports of families and isolated individuals with genetically determined CBS have been reported. In this systematic review, we analyze the demographical, clinical, radiological, and anatomopathological features of genetically confirmed cases of CBS. A systematic search was performed using the PubMed, EMBASE, and Cochrane Library databases, included all publications in English from 1 January 1999 through 1 August 2020. We found forty publications with fifty-eight eligible cases. A second search for publications dealing with genetic risk factors for CBS led to the review of eight additional articles. GRN was the most common gene involved in CBS, representing 28 out of 58 cases, followed by MAPT, C9ORF72, and PRNP. A set of symptoms was shown to be significantly more common in GRN-CBS patients, including visuospatial impairment, behavioral changes, aphasia, and language alterations. In addition, specific demographical, clinical, biochemical, and radiological features may suggest mutations in other genes. We suggest a diagnostic algorithm to help in identifying potential genetic cases of CBS in order to improve the diagnostic accuracy and to better understand the still poorly defined underlying pathogenetic process.
Collapse
Affiliation(s)
- Federica Arienti
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Giulia Lazzeri
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Maria Vizziello
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Edoardo Monfrini
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Nereo Bresolin
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy; (N.B.); (G.F.)
| | - Maria Cristina Saetti
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, Neuroscience Section, University of Milan, 20122 Milan, Italy; (F.A.); (G.L.); (M.V.); (E.M.); (M.C.S.)
| | - Marina Picillo
- Center for Neurodegenerative Diseases, Department of Medicine, Surgery and Dentistry, Neuroscience Section, University of Salerno, 84084 Salerno, Italy;
| | - Giulia Franco
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy; (N.B.); (G.F.)
| | - Alessio Di Fonzo
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Neurology Unit, 20122 Milan, Italy; (N.B.); (G.F.)
| |
Collapse
|
33
|
Wen Y, Zhou Y, Jiao B, Shen L. Genetics of Progressive Supranuclear Palsy: A Review. JOURNAL OF PARKINSON'S DISEASE 2021; 11:93-105. [PMID: 33104043 PMCID: PMC7990399 DOI: 10.3233/jpd-202302] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 09/26/2020] [Indexed: 02/06/2023]
Abstract
Progressive supranuclear palsy (PSP) is an atypical parkinsonism with prominent 4R-tau neuropathology, and the classical clinical phenotype is characterized by vertical supranuclear gaze palsy, unprovoked falls, akinetic-rigid syndrome and cognitive decline. Though PSP is generally regarded as sporadic, there is increasing evidence suggesting that a series of common and rare genetic variants impact on sporadic and familial forms of PSP. To date, more than 10 genes have been reported to show a potential association with PSP. Among these genes, the microtubule-associated protein tau (MAPT) is the risk locus with the strongest effect size on sporadic PSP in the case-control genome-wide association studies (GWAS). Additionally, MAPT mutations are the most common cause of familial PSP while the leucine-rich repeat kinase 2 (LRRK2) is a rare monogenic cause of PSP, and several other gene mutations may mimic the PSP phenotype, like the dynactin subunit 1 (DCTN1). In total, 15 MAPT mutations have been identified in cases with PSP, and the mean age at onset is much earlier than in cases carrying LRRK2 or DCTN1 mutations. GWAS have further identified several risk loci of PSP, proposing molecular pathways related to PSP. The present review focused on genetic studies on PSP and summarized genetic factors of PSP, which may help to elucidate the underlying pathogenesis and provide new perspectives for therapeutic strategies.
Collapse
Affiliation(s)
- Yafei Wen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yafang Zhou
- Department of Geriatrics Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, PR China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, PR China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, PR China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, PR China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan, PR China
| |
Collapse
|
34
|
Abdolmohammadi B, Dupre A, Evers L, Mez J. Genetics of Chronic Traumatic Encephalopathy. Semin Neurol 2020; 40:420-429. [DOI: 10.1055/s-0040-1713631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractAlthough chronic traumatic encephalopathy (CTE) garners substantial attention in the media and there have been marked scientific advances in the last few years, much remains unclear about the role of genetic risk in CTE. Two athletes with comparable contact-sport exposure may have varying amounts of CTE neuropathology, suggesting that other factors, including genetics, may contribute to CTE risk and severity. In this review, we explore reasons why genetics may be important for CTE, concepts in genetic study design for CTE (including choosing controls, endophenotypes, gene by environment interaction, and epigenetics), implicated genes in CTE (including APOE, MAPT, and TMEM106B), and whether predictive genetic testing for CTE should be considered.
Collapse
Affiliation(s)
- Bobak Abdolmohammadi
- Boston University Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA
- Boston University Chronic Traumatic Encephalopathy Center, Boston University School of Medicine, Boston, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| | - Alicia Dupre
- Boston University Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA
- Boston University Chronic Traumatic Encephalopathy Center, Boston University School of Medicine, Boston, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| | - Laney Evers
- Boston University Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA
- Boston University Chronic Traumatic Encephalopathy Center, Boston University School of Medicine, Boston, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| | - Jesse Mez
- Boston University Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA
- Boston University Chronic Traumatic Encephalopathy Center, Boston University School of Medicine, Boston, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| |
Collapse
|
35
|
Robinson JL, Yan N, Caswell C, Xie SX, Suh E, Van Deerlin VM, Gibbons G, Irwin DJ, Grossman M, Lee EB, Lee VMY, Miller B, Trojanowski JQ. Primary Tau Pathology, Not Copathology, Correlates With Clinical Symptoms in PSP and CBD. J Neuropathol Exp Neurol 2020; 79:296-304. [PMID: 31999351 DOI: 10.1093/jnen/nlz141] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/25/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Distinct neuronal and glial tau pathologies define corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP). Additional Alzheimer disease, TDP-43, and Lewy body copathologies are also common. The interplay of these pathologies with clinical symptoms remains unclear as individuals can present with corticobasal syndrome, frontotemporal dementia, PSP, or atypical Parkinsonism and may have additional secondary impairments. We report clinical, pathological, and genetic interactions in a cohort of CBD and PSP cases. Neurofibrillary tangles and plaques were common. Apolipoprotein E (APOE)ε4 carriers had more plaques while PSP APOEε2 carriers had fewer plaques. TDP-43 copathology was present and age-associated in 14% of PSP, and age-independent in 33% of CBD. Lewy body copathology varied from 9% to 15% and was not age-associated. The primary FTD-Tau burden-a sum of the neuronal, astrocytic and oligodendrocytic tau-was not age-, APOE-, or MAPT-related. In PSP, FTD-Tau, independent of copathology, associated with executive, language, motor, and visuospatial impairments, while PSP with Parkinsonism had a lower FTD-Tau burden, but this was not the case in CBD. Taken together, our results indicate that the primary tauopathy burden is the strongest correlate of clinical PSP, while copathologies are principally determined by age and genetic risk factors.
Collapse
Affiliation(s)
- John L Robinson
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine
| | - Ning Yan
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine.,Philadelphia, Pennsylvania; University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Carrie Caswell
- Penn Center for Neurodegenerative Disease Research.,Department of Biostatistics and Epidemiology, and Informatics
| | - Sharon X Xie
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine.,Department of Biostatistics and Epidemiology, and Informatics
| | - EunRan Suh
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine
| | - Vivianna M Van Deerlin
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine
| | - Garrett Gibbons
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine
| | - David J Irwin
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine.,Penn Frontotemporal Degeneration Center.,Department of Neurology, University of California San Francisco, San Francisco, California
| | - Murray Grossman
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Penn Frontotemporal Degeneration Center.,Department of Neurology, University of California San Francisco, San Francisco, California
| | - Edward B Lee
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine
| | - Virginia M-Y Lee
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine.,Department of Neurology, University of California San Francisco, San Francisco, California
| | - Bruce Miller
- Department of Neurology, University of California San Francisco, San Francisco, California
| | - John Q Trojanowski
- From the Penn Alzheimer's Disease Core Center.,Penn Center for Neurodegenerative Disease Research.,Department of Pathology and Laboratory Medicine.,Department of Neurology, University of California San Francisco, San Francisco, California
| |
Collapse
|
36
|
Shen XN, Miao D, Li JQ, Tan CC, Cao XP, Tan L, Yu JT. MAPT rs242557 variant is associated with hippocampus tau uptake on 18F-AV-1451 PET in non-demented elders. Aging (Albany NY) 2020; 11:874-884. [PMID: 30708351 PMCID: PMC6382414 DOI: 10.18632/aging.101783] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/15/2019] [Indexed: 11/29/2022]
Abstract
The microtubule-associated protein tau gene (MAPT) rs242557 variant is associated with multiple tauopathies and dementia. This study investigated whether it was correlated with brain tau-PET uptake in non-demented elders. Ninety non-demented elders were identified from the Alzheimer's Disease Neuroimaging Initiative cohort. We compared standardized uptake value ratios (SUVRs) of tau-PET tracer 18F-AV-1451 between rs242557 variant carriers and non-carriers in 25 regions of interest (ROIs). The minor allele A was associated with increased hippocampus 18F-AV-1451 uptake in non-demented elders (left: β = 0.111, Bonferroni corrected p = 0.035; right: β = 0.103, Bonferroni corrected p = 0.031). Aβ-positive participants (left: β = 0.206, Bonferroni corrected p = 0.029; right: β = 0.198, Bonferroni corrected p = 0.035) and APOE ε4 non-carriers (left: β = 0.140, Bonferroni corrected p = 0.006; right: β = 0.134, Bonferroni corrected p = 0.004) exhibited approximately the same findings in hippocampus. Considering no obvious associations in other regions, we confirmed the significant correlation of MAPT rs242557 risk variant with increased hippocampus tau deposition in non-demented elders. With higher magnitude signals in the hippocampus that is more likely to be uniquely affected in AD, the tau PET ligand 18F-AV-1451 seemed to possess a specific binding property for AD-like tau pathology.
Collapse
Affiliation(s)
- Xue-Ning Shen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Neurology, Fudan University, Shanghai, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Dan Miao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jie-Qiong Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Institute of Neurology, Fudan University, Shanghai, China.,Alzheimer's Disease Neuroimaging Initiative
| |
Collapse
|
37
|
Toral-Rios D, Pichardo-Rojas PS, Alonso-Vanegas M, Campos-Peña V. GSK3β and Tau Protein in Alzheimer's Disease and Epilepsy. Front Cell Neurosci 2020; 14:19. [PMID: 32256316 PMCID: PMC7089874 DOI: 10.3389/fncel.2020.00019] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/23/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia present in older adults; its etiology involves genetic and environmental factors. In recent years, epidemiological studies have shown a correlation between AD and chronic epilepsy since a considerable number of patients with AD may present seizures later on. Although the pathophysiology of seizures in AD is not completely understood, it could represent the result of several molecular mechanisms linked to amyloid beta-peptide (Aβ) accumulation and the hyperphosphorylation of tau protein, which may induce an imbalance in the release and recapture of excitatory and inhibitory neurotransmitters, structural alterations of the neuronal cytoskeleton, synaptic loss, and neuroinflammation. These changes could favor the recurrent development of hypersynchronous discharges and epileptogenesis, which, in a chronic state, favor the neurodegenerative process and influence the cognitive decline observed in AD. Supporting this correlation, histopathological studies in the brain tissue of temporal lobe epilepsy (TLE) patients have revealed the presence of Aβ deposits and the accumulation of tau protein in the neurofibrillary tangles (NFTs), accompanied by an increase of glycogen synthase kinase-3 beta (GSK3β) activity that may lead to an imminent alteration in posttranslational modifications of some microtubule-associated proteins (MAPs), mainly tau. The present review is focused on understanding the pathological aspects of GSK3β and tau in the development of TLE and AD.
Collapse
Affiliation(s)
- Danira Toral-Rios
- Departamento de Fisiología Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Pavel S Pichardo-Rojas
- Facultad de Ciencias de la Salud, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - Mario Alonso-Vanegas
- Centro Internacional de Cirug#x000ED;a de Epilepsia, Instituto Nacional de Neurología y Neurocirugía, HMG, Hospital Coyoacán, Mexico City, Mexico
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| |
Collapse
|
38
|
Heckman MG, Brennan RR, Labbé C, Soto AI, Koga S, DeTure MA, Murray ME, Petersen RC, Boeve BF, van Gerpen JA, Uitti RJ, Wszolek ZK, Rademakers R, Dickson DW, Ross OA. Association of MAPT Subhaplotypes With Risk of Progressive Supranuclear Palsy and Severity of Tau Pathology. JAMA Neurol 2020; 76:710-717. [PMID: 30882841 DOI: 10.1001/jamaneurol.2019.0250] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Importance The association between the microtubule-associated protein tau (MAPT) H1 haplotype and the risk of progressive supranuclear palsy (PSP) has been well documented. However, the specific H1 subhaplotypes that drive the association have not been evaluated in large studies, nor have they been studied in relation to neuropathologic severity of disease. Objective To comprehensively evaluate the associations of MAPT haplotypes with the risk of PSP and the severity of tau pathology using a large series of neuropathologically confirmed PSP cases. Design, Setting, and Participants A case-control study was used to investigate the associations between MAPT haplotypes and the risk of PSP, and a case series was conducted for examination of associations of MAPT haplotypes with the severity of tau pathology. All 802 neuropathologically confirmed PSP cases were obtained from a neurodegenerative disorders brain bank between January 1, 1998, and December 31, 2013, and 1312 clinical controls were obtained from the neurology department of the Mayo Clinic. Statistical analysis was performed from February 17 to December 12, 2018. Main Outcomes and Measures Presence of PSP in case-control analysis and semiquantitative tau pathology scores for neurofibrillary tangles, neuropil threads, tufted astrocytes, and oligodendroglial coiled bodies in PSP cases. Results For 802 patients with PSP (376 women and 426 men), the median age at death was 75 years (range, 52-98 years). For 1312 controls (701 women and 611 men), the median age at blood collection was 69 years (range, 45-92 years). After adjustment for multiple testing, known associations with risk of PSP were observed for the H2 and H1c haplotypes. Novel associations with PSP were observed for 3 H1 subhaplotypes, including H1d (odds ratio, 1.86; 95% CI, 1.43-2.42; P = 2 × 10-6), H1g (odds ratio, 3.64; 95% CI, 2.04-6.50; P = 2 × 10-6), and H1o (odds ratio, 2.60; 95% CI, 1.63-4.16; P = 2 × 10-5). Although not significant after multiple testing adjustment, 3 of these PSP risk haplotypes (H2, H1c, and H1d) were also nominally associated with measures of severity of tau pathology in PSP cases. Nominally significant associations with severity of tau pathology were also noted for the H1e and H1q haplotypes. Conclusions and Relevance This study has identified novel associations with risk of PSP for 3 MAPT H1 subhaplotypes. In addition, potential weaker associations between several haplotypes (including several PSP risk haplotypes) and severity of tau pathology were observed. These findings expand the current understanding of the role of MAPT haplotypic variation in susceptibility to and neuropathologic severity of PSP.
Collapse
Affiliation(s)
- Michael G Heckman
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida
| | | | - Catherine Labbé
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | | | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | | | | | | | | | | | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | | | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida.,Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
39
|
Miguel L, Frebourg T, Campion D, Lecourtois M. Moderate Overexpression of Tau in Drosophila Exacerbates Amyloid-β-Induced Neuronal Phenotypes and Correlates with Tau Oligomerization. J Alzheimers Dis 2020; 74:637-647. [PMID: 32065789 DOI: 10.3233/jad-190906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is neuropathologically defined by two key hallmarks: extracellular senile plaques composed primarily of amyloid-β (Aβ) peptide and intraneuronal neurofibrillary tangles, containing abnormally hyperphosphorylated tau protein. The tau protein is encoded by the MAPT gene. Recently, the H1 and H2 haplotypes of the MAPT gene were associated with AD risk. The minor MAPT H2 haplotype has been linked with a decreased risk of developing late-onset AD (LOAD). MAPT haplotypes show different levels of MAPT/Tau expression with H1 being ∼1.5-fold more expressed than H2, suggesting that MAPT expression level could be related to LOAD risk. In this study, we investigated whether this moderate difference in MAPT/Tau expression could influence Aβ-induced toxicity in vivo. We show that modest overexpression of tau protein in Drosophila exacerbates neuronal phenotypes in AβPP/BACE1 flies. The exacerbation of neuronal defects correlates with the accumulation of insoluble dTau oligomers, suggesting that the moderate difference in level of tau expression observed between H1 and H2 haplotypes could influence Aβ toxicity through the production of oligomeric tau insoluble species.
Collapse
Affiliation(s)
- Laetitia Miguel
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Thierry Frebourg
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Dominique Campion
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Centre Hospitalier du Rouvray, Sotteville-Lès-Rouen, France
| | - Magalie Lecourtois
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| |
Collapse
|
40
|
Fernández-Nogales M, Lucas JJ. Altered Levels and Isoforms of Tau and Nuclear Membrane Invaginations in Huntington's Disease. Front Cell Neurosci 2020; 13:574. [PMID: 32009905 PMCID: PMC6978886 DOI: 10.3389/fncel.2019.00574] [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: 09/13/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
Since the early reports of neurofibrillary Tau pathology in brains of some Huntington’s disease (HD) patients, mounting evidence of multiple alterations of Tau in HD brain tissue has emerged in recent years. Such Tau alterations range from increased total levels, imbalance of isoforms generated by alternative splicing (increased 4R-/3R-Tau ratio) or by post-translational modifications such as hyperphosphorylation or truncation. Besides, the detection in HD brains of a new Tau histopathological hallmark known as Tau nuclear rods (TNRs) or Tau-positive nuclear indentations (TNIs) led to propose HD as a secondary Tauopathy. After their discovery in HD brains, TNIs have also been reported in hippocampal neurons of early Braak stage AD cases and in frontal and temporal cortical neurons of FTD-MAPT cases due to the intronic IVS10+16 mutation in the Tau gene (MAPT) which results in an increased 4R-/3R-Tau ratio similar to that observed in HD. TNIs are likely pathogenic for contributing to the disturbed nucleocytoplasmic transport observed in HD. A key question is whether correction of any of the mentioned Tau alterations might have positive therapeutic implications for HD. The beneficial effect of decreasing Tau expression in HD mouse models clearly implicates Tau in HD pathogenesis. Such beneficial effect might be exerted by diminishing the excess total levels of Tau or specifically by diminishing the excess 4R-Tau, as well as any of their downstream effects. In any case, since gene silencing drugs are under development to attenuate both Huntingtin (HTT) expression for HD and MAPT expression for FTD-MAPT, it is conceivable that the combined therapy in HD patients might be more effective than HTT silencing alone.
Collapse
Affiliation(s)
| | - José J Lucas
- Centro de Biología Molecular Severo Ochoa (CBMSO)(CSIC-UAM), Madrid, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
41
|
Sánchez-Juan P, Moreno S, de Rojas I, Hernández I, Valero S, Alegret M, Montrreal L, García González P, Lage C, López-García S, Rodrííguez-Rodríguez E, Orellana A, Tárraga L, Boada M, Ruiz A. The MAPT H1 Haplotype Is a Risk Factor for Alzheimer's Disease in APOE ε4 Non-carriers. Front Aging Neurosci 2019; 11:327. [PMID: 31866851 PMCID: PMC6905227 DOI: 10.3389/fnagi.2019.00327] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/12/2019] [Indexed: 01/12/2023] Open
Abstract
An ancestral inversion of 900 kb on chromosome 17q21, which includes the microtubule-associated protein tau (MAPT) gene, defines two haplotype clades in Caucasians (H1 and H2). The H1 haplotype has been linked inconsistently with AD. In a previous study, we showed that an SNP tagging this haplotype (rs1800547) was associated with AD risk in a large population from the Dementia Genetics Spanish Consortium (DEGESCO) including 4435 cases and 6147 controls. The association was mainly driven by individuals that were non-carriers of the APOE ε4 allele. Our aim was to replicate our previous findings in an independent sample of 4124 AD cases and 3290 controls from Spain (GR@ACE project) and to analyze the effect of the H1 sub-haplotype structure on the risk of AD. The H1 haplotype was associated with AD risk (OR = 1.12; p = 0.0025). Stratification analysis showed that this association was mainly driven by the APOE ε4 non-carriers (OR = 1.15; p = 0.0022). Pooled analysis of both Spanish datasets (n = 17,996) showed that the highest AD risk related to the MAPT H1/H2 haplotype was in those individuals that were the oldest [third tertile (>77 years)] and did not carry APOE ε4 allele (p = 0.001). We did not find a significant association between H1 sub-haplotypes and AD. H1c was nominally associated but lost statistical significance after adjusting by population sub-structure. Our results are consistent with the hypothesis that genetic variants linked to the MAPT H1/H2 are tracking a genuine risk allele for AD. The fact that this association is stronger in APOE ε4 non-carriers partially explains previous controversial results and might be related to a slower alternative causal pathway less dependent on brain amyloid load.
Collapse
Affiliation(s)
- Pascual Sánchez-Juan
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Service of Neurology, University Hospital Marqués de Valdecilla, IDIVAL, University of Cantabria, Santander, Spain
| | - Sonia Moreno
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Itziar de Rojas
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Isabel Hernández
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Sergi Valero
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Montse Alegret
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Laura Montrreal
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Pablo García González
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Carmen Lage
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Service of Neurology, University Hospital Marqués de Valdecilla, IDIVAL, University of Cantabria, Santander, Spain
| | - Sara López-García
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Service of Neurology, University Hospital Marqués de Valdecilla, IDIVAL, University of Cantabria, Santander, Spain
| | - Eloy Rodrííguez-Rodríguez
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Service of Neurology, University Hospital Marqués de Valdecilla, IDIVAL, University of Cantabria, Santander, Spain
| | - Adelina Orellana
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Lluís Tárraga
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Mercè Boada
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Agustín Ruiz
- Center for Networked Biomedical Research on Neurodegenerative Diseases, National Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
42
|
Shoeibi A, Olfati N, Litvan I. Frontrunner in Translation: Progressive Supranuclear Palsy. Front Neurol 2019; 10:1125. [PMID: 31695675 PMCID: PMC6817677 DOI: 10.3389/fneur.2019.01125] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/08/2019] [Indexed: 12/26/2022] Open
Abstract
Progressive supranuclear palsy (PSP) is a four-repeat tau proteinopathy. Abnormal tau deposition is not unique for PSP and is the basic pathologic finding in some other neurodegenerative disorders such as Alzheimer's disease (AD), age-related tauopathy, frontotemporal degeneration, corticobasal degeneration, and chronic traumatic encephalopathy. While AD research has mostly been focused on amyloid beta pathology until recently, PSP as a prototype of a primary tauopathy with high clinical-pathologic correlation and a rapid course is a crucial candidate for tau therapeutic research. Several novel approaches to slow disease progression are being developed. It is expected that the benefits of translational research in this disease will extend beyond the PSP population. This article reviews advances in the diagnosis, epidemiology, pathology, hypothesized etiopathogenesis, and biomarkers and disease-modifying therapeutic approaches of PSP that is leading it to become a frontrunner in translation.
Collapse
Affiliation(s)
- Ali Shoeibi
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nahid Olfati
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Irene Litvan
- UC San Diego Department of Neurosciences, Parkinson and Other Movement Disorder Center, La Jolla, CA, United States
| |
Collapse
|
43
|
Singh B, Covelo A, Martell-Martínez H, Nanclares C, Sherman MA, Okematti E, Meints J, Teravskis PJ, Gallardo C, Savonenko AV, Benneyworth MA, Lesné SE, Liao D, Araque A, Lee MK. Tau is required for progressive synaptic and memory deficits in a transgenic mouse model of α-synucleinopathy. Acta Neuropathol 2019; 138:551-574. [PMID: 31168644 PMCID: PMC6778173 DOI: 10.1007/s00401-019-02032-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/27/2019] [Accepted: 05/27/2019] [Indexed: 01/01/2023]
Abstract
Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are clinically and neuropathologically highly related α-synucleinopathies that collectively constitute the second leading cause of neurodegenerative dementias. Genetic and neuropathological studies directly implicate α-synuclein (αS) abnormalities in PDD and DLB pathogenesis. However, it is currently unknown how αS abnormalities contribute to memory loss, particularly since forebrain neuronal loss in PDD and DLB is less severe than in Alzheimer's disease. Previously, we found that familial Parkinson's disease-linked human mutant A53T αS causes aberrant localization of the microtubule-associated protein tau to postsynaptic spines in neurons, leading to postsynaptic deficits. Thus, we directly tested if the synaptic and memory deficits in a mouse model of α-synucleinopathy (TgA53T) are mediated by tau. TgA53T mice exhibit progressive memory deficits associated with postsynaptic deficits in the absence of obvious neuropathological and neurodegenerative changes in the hippocampus. Significantly, removal of endogenous mouse tau expression in TgA53T mice (TgA53T/mTau-/-), achieved by mating TgA53T mice to mouse tau-knockout mice, completely ameliorates cognitive dysfunction and concurrent synaptic deficits without affecting αS expression or accumulation of selected toxic αS oligomers. Among the known tau-dependent effects, memory deficits in TgA53T mice were associated with hippocampal circuit remodeling linked to chronic network hyperexcitability. This remodeling was absent in TgA53T/mTau-/- mice, indicating that postsynaptic deficits, aberrant network hyperactivity, and memory deficits are mechanistically linked. Our results directly implicate tau as a mediator of specific human mutant A53T αS-mediated abnormalities related to deficits in hippocampal neurotransmission and suggest a mechanism for memory impairment that occurs as a consequence of synaptic dysfunction rather than synaptic or neuronal loss. We hypothesize that these initial synaptic deficits contribute to network hyperexcitability which, in turn, exacerbate cognitive dysfunction. Our results indicate that these synaptic changes present potential therapeutic targets for amelioration of memory deficits in α-synucleinopathies.
Collapse
Affiliation(s)
- Balvindar Singh
- Medical Scientist Training Program, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Graduate Program in Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Ana Covelo
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Héctor Martell-Martínez
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Carmen Nanclares
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Mathew A Sherman
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Emmanuel Okematti
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Joyce Meints
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Peter J Teravskis
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Christopher Gallardo
- Graduate Program in Pharmacology, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Alena V Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA
| | - Michael A Benneyworth
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Mouse Behavior Core, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Sylvain E Lesné
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- N. Budd Grossman Center for Memory Research and Care, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
- Institute for Translational Neuroscience, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
- Geriatric Research Education and Clinical Center, Minneapolis Veterans Affairs Health Care System, University of Minnesota Medical School, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
44
|
Mroczko B, Groblewska M, Litman-Zawadzka A. The Role of Protein Misfolding and Tau Oligomers (TauOs) in Alzheimer's Disease (AD). Int J Mol Sci 2019; 20:E4661. [PMID: 31547024 PMCID: PMC6802364 DOI: 10.3390/ijms20194661] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 11/25/2022] Open
Abstract
Although the causative role of the accumulation of amyloid β 1-42 (Aβ42) deposits in the pathogenesis of Alzheimer's disease (AD) has been under debate for many years, it is supposed that the toxicity soluble oligomers of Tau protein (TauOs) might be also the pathogenic factor acting on the initial stages of this disease. Therefore, we performed a thorough search for literature pertaining to our investigation via the MEDLINE/PubMed database. It was shown that soluble TauOs, especially granular forms, may be the most toxic form of this protein. Hyperphosphorylated TauOs can reduce the number of synapses by missorting into axonal compartments of neurons other than axon. Furthermore, soluble TauOs may be also responsible for seeding Tau pathology within AD brains, with probable link to AβOs toxicity. Additionally, the concentrations of TauOs in the cerebrospinal fluid (CSF) and plasma of AD patients were higher than in non-demented controls, and revealed a negative correlation with mini-mental state examination (MMSE) scores. It was postulated that adding the measurements of TauOs to the panel of CSF biomarkers could improve the diagnosis of AD.
Collapse
Affiliation(s)
- Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland.
- Department of Biochemical Diagnostics, University Hospital of Białystok, 15-269 Białystok, Poland.
| | - Magdalena Groblewska
- Department of Biochemical Diagnostics, University Hospital of Białystok, 15-269 Białystok, Poland.
| | - Ala Litman-Zawadzka
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland.
| |
Collapse
|
45
|
Dumitrescu L, Barnes LL, Thambisetty M, Beecham G, Kunkle B, Bush WS, Gifford KA, Chibnik LB, Mukherjee S, De Jager PL, Kukull W, Crane PK, Resnick SM, Keene CD, Montine TJ, Schellenberg GD, Deming Y, Chao MJ, Huentelman M, Martin ER, Hamilton-Nelson K, Shaw LM, Trojanowski JQ, Peskind ER, Cruchaga C, Pericak-Vance MA, Goate AM, Cox NJ, Haines JL, Zetterberg H, Blennow K, Larson EB, Johnson SC, Albert M, Bennett DA, Schneider JA, Jefferson AL, Hohman TJ. Sex differences in the genetic predictors of Alzheimer's pathology. Brain 2019; 142:2581-2589. [PMID: 31497858 PMCID: PMC6736148 DOI: 10.1093/brain/awz206] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/03/2019] [Accepted: 05/15/2019] [Indexed: 01/01/2023] Open
Abstract
Autopsy measures of Alzheimer's disease neuropathology have been leveraged as endophenotypes in previous genome-wide association studies (GWAS). However, despite evidence of sex differences in Alzheimer's disease risk, sex-stratified models have not been incorporated into previous GWAS analyses. We looked for sex-specific genetic associations with Alzheimer's disease endophenotypes from six brain bank data repositories. The pooled dataset included 2701 males and 3275 females, the majority of whom were diagnosed with Alzheimer's disease at autopsy (70%). Sex-stratified GWAS were performed within each dataset and then meta-analysed. Loci that reached genome-wide significance (P < 5 × 10-8) in stratified models were further assessed for sex interactions. Additional analyses were performed in independent datasets leveraging cognitive, neuroimaging and CSF endophenotypes, along with age-at-onset data. Outside of the APOE region, one locus on chromosome 7 (rs34331204) showed a sex-specific association with neurofibrillary tangles among males (P = 2.5 × 10-8) but not females (P = 0.85, sex-interaction P = 2.9 × 10-4). In follow-up analyses, rs34331204 was also associated with hippocampal volume, executive function, and age-at-onset only among males. These results implicate a novel locus that confers male-specific protection from tau pathology and highlight the value of assessing genetic associations in a sex-specific manner.
Collapse
Affiliation(s)
- Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa L Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Gary Beecham
- John T MacDonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
- John P. Hussman Institute for Human Genomics, University of Miami School of Medicine, Miami, FL, USA
| | - Brian Kunkle
- John P. Hussman Institute for Human Genomics, University of Miami School of Medicine, Miami, FL, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Katherine A Gifford
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lori B Chibnik
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
- Cell Circuits Program, Broad Institute, Cambridge MA, USA
| | - Walter Kukull
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael J Chao
- Ronald M Loeb Center for Alzheimer’s Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matt Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Eden R Martin
- John T MacDonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
- John P. Hussman Institute for Human Genomics, University of Miami School of Medicine, Miami, FL, USA
| | - Kara Hamilton-Nelson
- John P. Hussman Institute for Human Genomics, University of Miami School of Medicine, Miami, FL, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine R Peskind
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami School of Medicine, Miami, FL, USA
| | - Alison M Goate
- Ronald M Loeb Center for Alzheimer’s Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Sterling C Johnson
- Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Marilyn Albert
- Department of Neurology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Angela L Jefferson
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
46
|
Stamelou M, Giagkou N, Höglinger GU. One decade ago, one decade ahead in progressive supranuclear palsy. Mov Disord 2019; 34:1284-1293. [DOI: 10.1002/mds.27788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Affiliation(s)
- Maria Stamelou
- Parkinson's disease and Movement Disorders DepartmentHYGEIA Hospital Athens Greece
- Neurology ClinicPhilipps University Marburg Germany
- First Department of Neurology, Aiginiteion HospitalUniversity of Athens Athens Greece
| | - Nikolaos Giagkou
- Parkinson's disease and Movement Disorders DepartmentHYGEIA Hospital Athens Greece
| | - Günter U Höglinger
- Department of NeurologyTechnische Universität München Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Germany
| |
Collapse
|
47
|
Strang KH, Golde TE, Giasson BI. MAPT mutations, tauopathy, and mechanisms of neurodegeneration. J Transl Med 2019; 99:912-928. [PMID: 30742061 PMCID: PMC7289372 DOI: 10.1038/s41374-019-0197-x] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 11/09/2022] Open
Abstract
In multiple neurodegenerative diseases, including Alzheimer's disease (AD), a prominent pathological feature is the aberrant aggregation and inclusion formation of the microtubule-associated protein tau. Because of the pathological association, these disorders are often referred to as tauopathies. Mutations in the MAPT gene that encodes tau can cause frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), providing the clearest evidence that tauopathy plays a causal role in neurodegeneration. However, large gaps in our knowledge remain regarding how various FTDP-17-linked tau mutations promote tau aggregation and neurodegeneration, and, more generally, how the tauopathy is linked to neurodegeneration. Herein, we review what is known about how FTDP-17-linked pathogenic MAPT mutations cause disease, with a major focus on the prion-like properties of wild-type and mutant tau proteins. The hypothesized mechanisms by which mutations in the MAPT gene promote tauopathy are quite varied and may not provide definitive insights into how tauopathy arises in the absence of mutation. Further, differences in the ability of tau and mutant tau proteins to support prion-like propagation in various model systems raise questions about the generalizability of this mechanism in various tauopathies. Notably, understanding the mechanisms of tauopathy induction and spread and tau-induced neurodegeneration has important implications for tau-targeting therapeutics.
Collapse
Affiliation(s)
- Kevin H Strang
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Todd E Golde
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
48
|
Heckman MG, Kasanuki K, Brennan RR, Labbé C, Vargas ER, Soto AI, Murray ME, Koga S, Dickson DW, Ross OA. Association of MAPT H1 subhaplotypes with neuropathology of lewy body disease. Mov Disord 2019; 34:1325-1332. [PMID: 31234228 DOI: 10.1002/mds.27773] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/30/2019] [Accepted: 05/20/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Genetic variation at the microtubule-associated protein tau locus is associated with clinical parkinsonism. However, it is unclear as to whether microtubule-associated protein tau H1 subhaplotypes are associated with the burden of neuropathological features of Lewy body disease. OBJECTIVES To evaluate associations of microtubule-associated protein tau haplotypes with severity of Lewy body pathology and markers of SN neuronal loss in Lewy body disease cases. METHODS Five hundred eighty-five autopsy-confirmed Lewy body disease cases were included. Six microtubule-associated protein tau variants (rs1467967, rs242557, rs3785883, rs2471738, rs8070723, and rs7521) were genotyped to define common microtubule-associated protein tau haplotypes. Lewy body counts were measured in five cortical regions. Ventrolateral and medial SN neuronal loss were assessed semiquantitatively. Nigrostriatal dopaminergic degeneration was quantified by image analysis of tyrosine hydroxylase immunoreactivity in the dorsolateral and ventromedial putamen. RESULTS The common microtubule-associated protein tau H2 haplotype did not show a strong effect on pathological burden in Lewy body disease. The rare H1j haplotype (1.3%) was significantly associated with a lower dorsolateral putaminal tyrosine hydroxylase immunoreactivity (and therefore greater dopaminergic degeneration) compared to other microtubule-associated protein tau haplotypes (P = 0.0016). Microtubule-associated protein tau H1j was also nominally (P ≤ 0.05) associated with a lower ventromedial putaminal tyrosine hydroxylase immunoreactivity (P = 0.010), but this did not survive multiple testing correction. Other nominally significant associations between microtubule-associated protein tau H1 subhaplotypes and neuropathological outcomes were observed. CONCLUSIONS A rare microtubule-associated protein tau H1 subhaplotype (H1j) may be associated with more severe putaminal dopaminergic degeneration in Lewy body disease cases. Microtubule-associated protein tau H1j has been associated previously with an increased risk of PD, and therefore our exploratory findings provide insight into the mechanism by which H1j modulates PD risk. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Michael G Heckman
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida, USA
| | - Koji Kasanuki
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Catherine Labbé
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Emily R Vargas
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida, USA
| | - Alexandra I Soto
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA.,Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
49
|
Li YI, Wong G, Humphrey J, Raj T. Prioritizing Parkinson's disease genes using population-scale transcriptomic data. Nat Commun 2019; 10:994. [PMID: 30824768 PMCID: PMC6397174 DOI: 10.1038/s41467-019-08912-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 02/05/2019] [Indexed: 12/23/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified over 41 susceptibility loci associated with Parkinson's Disease (PD) but identifying putative causal genes and the underlying mechanisms remains challenging. Here, we leverage large-scale transcriptomic datasets to prioritize genes that are likely to affect PD by using a transcriptome-wide association study (TWAS) approach. Using this approach, we identify 66 gene associations whose predicted expression or splicing levels in dorsolateral prefrontal cortex (DLFPC) and peripheral monocytes are significantly associated with PD risk. We uncover many novel genes associated with PD but also novel mechanisms for known associations such as MAPT, for which we find that variation in exon 3 splicing explains the common genetic association. Genes identified in our analyses belong to the same or related pathways including lysosomal and innate immune function. Overall, our study provides a strong foundation for further mechanistic studies that will elucidate the molecular drivers of PD.
Collapse
Affiliation(s)
- Yang I Li
- Section of Genetic Medicine, Department of Medicine, and Department of Human Genetics, University of Chicago, Chicago, 60637, IL, USA
| | - Garrett Wong
- Departments of Neuroscience, and Genetics and Genomic Sciences, Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - Jack Humphrey
- UCL Genetics Institute, Gower Street, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1E 6BT, UK
| | - Towfique Raj
- Departments of Neuroscience, and Genetics and Genomic Sciences, Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.
| |
Collapse
|
50
|
Jadhav S, Avila J, Schöll M, Kovacs GG, Kövari E, Skrabana R, Evans LD, Kontsekova E, Malawska B, de Silva R, Buee L, Zilka N. A walk through tau therapeutic strategies. Acta Neuropathol Commun 2019; 7:22. [PMID: 30767766 PMCID: PMC6376692 DOI: 10.1186/s40478-019-0664-z] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/21/2019] [Indexed: 12/18/2022] Open
Abstract
Tau neuronal and glial pathologies drive the clinical presentation of Alzheimer's disease and related human tauopathies. There is a growing body of evidence indicating that pathological tau species can travel from cell to cell and spread the pathology through the brain. Throughout the last decade, physiological and pathological tau have become attractive targets for AD therapies. Several therapeutic approaches have been proposed, including the inhibition of protein kinases or protein-3-O-(N-acetyl-beta-D-glucosaminyl)-L-serine/threonine Nacetylglucosaminyl hydrolase, the inhibition of tau aggregation, active and passive immunotherapies, and tau silencing by antisense oligonucleotides. New tau therapeutics, across the board, have demonstrated the ability to prevent or reduce tau lesions and improve either cognitive or motor impairment in a variety of animal models developing neurofibrillary pathology. The most advanced strategy for the treatment of human tauopathies remains immunotherapy, which has already reached the clinical stage of drug development. Tau vaccines or humanised antibodies target a variety of tau species either in the intracellular or extracellular spaces. Some of them recognise the amino-terminus or carboxy-terminus, while others display binding abilities to the proline-rich area or microtubule binding domains. The main therapeutic foci in existing clinical trials are on Alzheimer's disease, progressive supranuclear palsy and non-fluent primary progressive aphasia. Tau therapy offers a new hope for the treatment of many fatal brain disorders. First efficacy data from clinical trials will be available by the end of this decade.
Collapse
Affiliation(s)
- Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska 9, 845 10, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Jesus Avila
- Centro de Biologia Molecular "Severo Ochoa", Consejo Superior de Investigaciones, Cientificas, Universidad Autonoma de Madrid, C/ Nicolas Cabrera, 1. Campus de Cantoblanco, 28049, Madrid, Spain
- Networking Research Center on Neurodegenerative, Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of, Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Dementia Research Centre, University College London, London, UK
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | - Enikö Kövari
- Department of Mental Health and Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Rostislav Skrabana
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Lewis D Evans
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Eva Kontsekova
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Cracow, Poland
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Luc Buee
- Universite of Lille, Inserm, CHU-Lille, UMRS1172, Alzheimer & Tauopathies, Place de Verdun, 59045, Lille cedex, France.
| | - Norbert Zilka
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia.
| |
Collapse
|