1
|
Bryan Iii MR, Tian X, Tseng JH, Evangelista BA, Ragusa JV, Bryan AF, Trotman W, Irwin D, Cohen TJ. Development and characterization of novel anti-acetylated tau monoclonal antibodies to probe pathogenic tau species in Alzheimer's disease. Acta Neuropathol Commun 2024; 12:163. [PMID: 39396065 PMCID: PMC11470691 DOI: 10.1186/s40478-024-01865-1] [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: 09/04/2024] [Accepted: 09/22/2024] [Indexed: 10/14/2024] Open
Abstract
Tauopathies, including Alzheimer's disease (AD), are a class of neurodegenerative diseases characterized by the presence of insoluble tau inclusions. Tau phosphorylation has traditionally been viewed as the dominant post-translational modification (PTM) controlling tau function and pathogenesis in tauopathies. However, we and others have identified tau acetylation as a primary PTM regulating both normal tau function as well as abnormal pathogenic features including aggregation. Prior work showed robust tau acetylation in aggregation hotspots located within the 2nd and 3rd repeat regions of tau (residues K280 and K311) in tauopathy brains, including AD, compared to non-tauopathy controls. By screening thousands of hybridoma clones, we generated site-specific and modification-specific monoclonal antibodies targeting acetylated tau at residues K280 or K311. To validate these antibodies in a bona fide neuronal system, we targeted the acetyltransferase CBP to the cytoplasm of neurons to promote tau acetylation. Several antibody clones specifically detected CBP-acetylated tau and co-localized with ac-tau in neurons. Additionally, our lead optimal anti-acetylated-tau monoclonal antibodies detected robust tau pathology in tangles and neuritic plaques of human AD brains. Given the now emerging interest in acetylated tau as critical regulator of tau functions, these sensitive and highly specific tools will allow us to further unravel the tau PTM code and, importantly, could be deployed as diagnostic or disease-modifying agents.
Collapse
Affiliation(s)
- Miles R Bryan Iii
- Department of Neurology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xu Tian
- Department of Neurology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jui-Heng Tseng
- Department of Neurology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Baggio A Evangelista
- Department of Neurology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joey V Ragusa
- Department of Cell Biology and Physiology, University of North Carolia at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Audra F Bryan
- Department of Neurology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Winifred Trotman
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David Irwin
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Todd J Cohen
- Department of Neurology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Cell Biology and Physiology, University of North Carolia at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
2
|
Gaikwad S, Puangmalai N, Sonawane M, Montalbano M, Price R, Iyer MS, Ray A, Moreno S, Kayed R. Nasal tau immunotherapy clears intracellular tau pathology and improves cognitive functions in aged tauopathy mice. Sci Transl Med 2024; 16:eadj5958. [PMID: 38959324 DOI: 10.1126/scitranslmed.adj5958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Pathological tau aggregates cause cognitive decline in neurodegenerative tauopathies, including Alzheimer's disease (AD). These aggregates are prevalent within intracellular compartments. Current tau immunotherapies have shown limited efficacy in clearing intracellular tau aggregates and improving cognition in clinical trials. In this study, we developed toxic tau conformation-specific monoclonal antibody-2 (TTCM2), which selectively recognized pathological tau aggregates in brain tissues from patients with AD, dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). TTCM2 potently inhibited tau-seeding activity, an essential mechanism underlying tauopathy progression. To effectively target intracellular tau aggregates and ensure rapid delivery to the brain, TTCM2 was loaded in micelles (TTCM2-ms) and administered through the intranasal route. We found that intranasally administered TTCM2-ms efficiently entered the brain in hTau-tauopathy mice, targeting pathological tau in intracellular compartments. Moreover, a single intranasal dose of TTCM2-ms effectively cleared pathological tau, elevated synaptic proteins, and improved cognitive functions in aged tauopathy mice. Mechanistic studies revealed that TTCM2-ms cleared intracellular, synaptic, and seed-competent tau aggregates through tripartite motif-containing 21 (TRIM21), an intracellular antibody receptor and E3 ubiquitin ligase known to facilitate proteasomal degradation of cytosolic antibody-bound proteins. TRIM21 was found to be essential for TTCM2-ms-mediated clearance of tau pathology. Our study collectively provides evidence of the effectiveness of nasal tau immunotherapy in targeting and clearing intracellular tau pathology through TRIM21 and enhancing cognition in aged tauopathy mice. This study could be valuable in designing effective tau immunotherapies for AD and other tauopathies.
Collapse
Affiliation(s)
- Sagar Gaikwad
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Minal Sonawane
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rachel Price
- Department of Science, University "Roma Tre," Viale G. Marconi 446 00146 Rome, Italy
| | | | | | - Sandra Moreno
- Department of Science, University "Roma Tre," Viale G. Marconi 446 00146 Rome, Italy
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
3
|
Parra Bravo C, Krukowski K, Barker S, Wang C, Li Y, Fan L, Vázquez-Rosa E, Shin MK, Wong MY, McCullough LD, Kitagawa RS, Choi HA, Cacace A, Sinha SC, Pieper AA, Rosi S, Chen X, Gan L. Anti-acetylated-tau immunotherapy is neuroprotective in tauopathy and brain injury. Mol Neurodegener 2024; 19:51. [PMID: 38915105 PMCID: PMC11197196 DOI: 10.1186/s13024-024-00733-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/15/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Tau is aberrantly acetylated in various neurodegenerative conditions, including Alzheimer's disease, frontotemporal lobar degeneration (FTLD), and traumatic brain injury (TBI). Previously, we reported that reducing acetylated tau by pharmacologically inhibiting p300-mediated tau acetylation at lysine 174 reduces tau pathology and improves cognitive function in animal models. METHODS We investigated the therapeutic efficacy of two different antibodies that specifically target acetylated lysine 174 on tau (ac-tauK174). We treated PS19 mice, which harbor the P301S tauopathy mutation that causes FTLD, with anti-ac-tauK174 and measured effects on tau pathology, neurodegeneration, and neurobehavioral outcomes. Furthermore, PS19 mice received treatment post-TBI to evaluate the ability of the immunotherapy to prevent TBI-induced exacerbation of tauopathy phenotypes. Ac-tauK174 measurements in human plasma following TBI were also collected to establish a link between trauma and acetylated tau levels, and single nuclei RNA-sequencing of post-TBI brain tissues from treated mice provided insights into the molecular mechanisms underlying the observed treatment effects. RESULTS Anti-ac-tauK174 treatment mitigates neurobehavioral impairment and reduces tau pathology in PS19 mice. Ac-tauK174 increases significantly in human plasma 24 h after TBI, and anti-ac-tauK174 treatment of PS19 mice blocked TBI-induced neurodegeneration and preserved memory functions. Anti-ac-tauK174 treatment rescues alterations of microglial and oligodendrocyte transcriptomic states following TBI in PS19 mice. CONCLUSIONS The ability of anti-ac-tauK174 treatment to rescue neurobehavioral impairment, reduce tau pathology, and rescue glial responses demonstrates that targeting tau acetylation at K174 is a promising neuroprotective therapeutic approach to human tauopathies resulting from TBI or genetic disease.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Karen Krukowski
- Department of Physical Therapy & Rehabilitation Science, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah Barker
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Yaqiao Li
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Li Fan
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Edwin Vázquez-Rosa
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
| | - Min-Kyoo Shin
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Man Ying Wong
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ryan S Kitagawa
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Alex Choi
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Subhash C Sinha
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Susanna Rosi
- Department of Physical Therapy & Rehabilitation Science, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Xu Chen
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
- Department of Neurosciences, School of Medicine, University of California, San Diego, USA.
| | - Li Gan
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Tan W, Thiruppathi J, Hong SH, Puth S, Pheng S, Mun BR, Choi WS, Lee KH, Park HS, Nguyen DT, Lee MC, Jeong K, Zheng JH, Kim Y, Lee SE, Rhee JH. Development of an anti-tauopathy mucosal vaccine specifically targeting pathologic conformers. NPJ Vaccines 2024; 9:108. [PMID: 38879560 PMCID: PMC11180213 DOI: 10.1038/s41541-024-00904-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/28/2024] [Indexed: 06/19/2024] Open
Abstract
Alzheimer's disease (AD) and related tauopathies are associated with pathological tau protein aggregation, which plays an important role in neurofibrillary degeneration and dementia. Targeted immunotherapy to eliminate pathological tau aggregates is known to improve cognitive deficits in AD animal models. The tau repeat domain (TauRD) plays a pivotal role in tau-microtubule interactions and is critically involved in the aggregation of hyperphosphorylated tau proteins. Because TauRD forms the structural core of tau aggregates, the development of immunotherapies that selectively target TauRD-induced pathological aggregates holds great promise for the modulation of tauopathies. In this study, we generated recombinant TauRD polypeptide that form neurofibrillary tangle-like structures and evaluated TauRD-specific immune responses following intranasal immunization in combination with the mucosal adjuvant FlaB. In BALB/C mice, repeated immunizations at one-week intervals induced robust TauRD-specific antibody responses in a TLR5-dependent manner. Notably, the resulting antiserum recognized only the aggregated form of TauRD, while ignoring monomeric TauRD. The antiserum effectively inhibited TauRD filament formation and promoted the phagocytic degradation of TauRD aggregate fragments by microglia. The antiserum also specifically recognized pathological tau conformers in the human AD brain. Based on these results, we engineered a built-in flagellin-adjuvanted TauRD (FlaB-TauRD) vaccine and tested its efficacy in a P301S transgenic mouse model. Mucosal immunization with FlaB-TauRD improved quality of life, as indicated by the amelioration of memory deficits, and alleviated tauopathy progression. Notably, the survival of the vaccinated mice was dramatically extended. In conclusion, we developed a mucosal vaccine that exclusively targets pathological tau conformers and prevents disease progression.
Collapse
Affiliation(s)
- Wenzhi Tan
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Jayalakshmi Thiruppathi
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Seol Hee Hong
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sao Puth
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Sophea Pheng
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Bo-Ram Mun
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kyung-Hwa Lee
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Pathology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Hyun-Sun Park
- Department of Pharmacology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Duc Tien Nguyen
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Min-Cheol Lee
- Department of Pathology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Seegene Inc, Seoul, 05548, Republic of Korea
| | - Kwangjoon Jeong
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Jin Hai Zheng
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Young Kim
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
| |
Collapse
|
5
|
Basheer N, Buee L, Brion JP, Smolek T, Muhammadi MK, Hritz J, Hromadka T, Dewachter I, Wegmann S, Landrieu I, Novak P, Mudher A, Zilka N. Shaping the future of preclinical development of successful disease-modifying drugs against Alzheimer's disease: a systematic review of tau propagation models. Acta Neuropathol Commun 2024; 12:52. [PMID: 38576010 PMCID: PMC10993623 DOI: 10.1186/s40478-024-01748-5] [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: 01/11/2024] [Accepted: 02/21/2024] [Indexed: 04/06/2024] Open
Abstract
The transcellular propagation of the aberrantly modified protein tau along the functional brain network is a key hallmark of Alzheimer's disease and related tauopathies. Inoculation-based tau propagation models can recapitulate the stereotypical spread of tau and reproduce various types of tau inclusions linked to specific tauopathy, albeit with varying degrees of fidelity. With this systematic review, we underscore the significance of judicious selection and meticulous functional, biochemical, and biophysical characterization of various tau inocula. Furthermore, we highlight the necessity of choosing suitable animal models and inoculation sites, along with the critical need for validation of fibrillary pathology using confirmatory staining, to accurately recapitulate disease-specific inclusions. As a practical guide, we put forth a framework for establishing a benchmark of inoculation-based tau propagation models that holds promise for use in preclinical testing of disease-modifying drugs.
Collapse
Affiliation(s)
- Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Luc Buee
- Inserm, CHU Lille, CNRS, LilNCog - Lille Neuroscience & Cognition, University of Lille, 59000, Lille, France.
| | - Jean-Pierre Brion
- Faculty of Medicine, Laboratory of Histology, Alzheimer and Other Tauopathies Research Group (CP 620), ULB Neuroscience Institute (UNI), Université Libre de Bruxelles, 808, Route de Lennik, 1070, Brussels, Belgium
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Muhammad Khalid Muhammadi
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Jozef Hritz
- CEITEC Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Tomas Hromadka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Ilse Dewachter
- Biomedical Research Institute, BIOMED, Hasselt University, 3500, Hasselt, Belgium
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases, Charitéplatz 1, 10117, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Isabelle Landrieu
- CNRS EMR9002 - BSI - Integrative Structural Biology, 59000, Lille, France
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, 59000, Lille, France
| | - Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia
| | - Amritpal Mudher
- School of Biological Sciences, Faculty of Environment and Life Sciences, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10, Bratislava, Slovakia.
- AXON Neuroscience R&D Services SE, Dubravska Cesta 9, 845 10, Bratislava, Slovakia.
| |
Collapse
|
6
|
Alhadidy MM, Kanaan NM. Biochemical approaches to assess the impact of post-translational modifications on pathogenic tau conformations using recombinant protein. Biochem Soc Trans 2024; 52:301-318. [PMID: 38348781 PMCID: PMC10903483 DOI: 10.1042/bst20230596] [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: 11/10/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/29/2024]
Abstract
Tau protein is associated with many neurodegenerative disorders known as tauopathies. Aggregates of tau are thought of as a main contributor to neurodegeneration in these diseases. Increasingly, evidence points to earlier, soluble conformations of abnormally modified monomers and multimeric tau as toxic forms of tau. The biological processes driving tau from physiological species to pathogenic conformations remain poorly understood, but certain avenues are currently under investigation including the functional consequences of various pathological tau changes (e.g. mutations, post-translational modifications (PTMs), and protein-protein interactions). PTMs can regulate several aspects of tau biology such as proteasomal and autophagic clearance, solubility, and aggregation. Moreover, PTMs can contribute to the transition of tau from normal to pathogenic conformations. However, our understating of how PTMs specifically regulate the transition of tau into pathogenic conformations is partly impeded by the relative lack of structured frameworks to assess and quantify these conformations. In this review, we describe a set of approaches that includes several in vitro assays to determine the contribution of PTMs to tau's transition into known pathogenic conformations. The approaches begin with different methods to create recombinant tau proteins carrying specific PTMs followed by validation of the PTMs status. Then, we describe a set of biochemical and biophysical assays that assess the contribution of a given PTM to different tau conformations, including aggregation, oligomerization, exposure of the phosphatase-activating domain, and seeding. Together, these approaches can facilitate the advancement of our understanding of the relationships between PTMs and tau conformations.
Collapse
Affiliation(s)
- Mohammed M. Alhadidy
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, U.S.A
- Neuroscience Program, Michigan State University, East Lansing, MI, U.S.A
| | - Nicholas M. Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, U.S.A
- Neuroscience Program, Michigan State University, East Lansing, MI, U.S.A
| |
Collapse
|
7
|
Gaikwad S, Senapati S, Haque MA, Kayed R. Senescence, brain inflammation, and oligomeric tau drive cognitive decline in Alzheimer's disease: Evidence from clinical and preclinical studies. Alzheimers Dement 2024; 20:709-727. [PMID: 37814508 PMCID: PMC10841264 DOI: 10.1002/alz.13490] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/11/2023]
Abstract
Aging, tau pathology, and chronic inflammation in the brain play crucial roles in synaptic loss, neurodegeneration, and cognitive decline in tauopathies, including Alzheimer's disease. Senescent cells accumulate in the aging brain, accelerate the aging process, and promote tauopathy progression through their abnormal inflammatory secretome known as the senescence-associated secretory phenotype (SASP). Tau oligomers (TauO)-the most neurotoxic tau species-are known to induce senescence and the SASP, which subsequently promote neuropathology, inflammation, oxidative stress, synaptic dysfunction, neuronal death, and cognitive dysfunction. TauO, brain inflammation, and senescence are associated with heterogeneity in tauopathy progression and cognitive decline. However, the underlying mechanisms driving the disease heterogeneity remain largely unknown, impeding the development of therapies for tauopathies. Based on clinical and preclinical evidence, this review highlights the critical role of TauO and senescence in neurodegeneration. We discuss key knowledge gaps and potential strategies for targeting senescence and TauO to treat tauopathies. HIGHLIGHTS: Senescence, oligomeric Tau (TauO), and brain inflammation accelerate the aging process and promote the progression of tauopathies, including Alzheimer's disease. We discuss their role in contributing to heterogeneity in tauopathy and cognitive decline. We highlight strategies to target senescence and TauO to treat tauopathies while addressing key knowledge gaps.
Collapse
Affiliation(s)
- Sagar Gaikwad
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Sudipta Senapati
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Md. Anzarul Haque
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- The Mitchell Center for Neurodegenerative Diseasesand Department of NeurologyUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
8
|
Paterno G, Torrellas J, Bell BM, Gorion KMM, Quintin SS, Hery GP, Prokop S, Giasson BI. Novel Conformation-Dependent Tau Antibodies Are Modulated by Adjacent Phosphorylation Sites. Int J Mol Sci 2023; 24:13676. [PMID: 37761979 PMCID: PMC10530490 DOI: 10.3390/ijms241813676] [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: 07/18/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Tau proteins within the adult central nervous system (CNS) are found to be abnormally aggregated into heterogeneous filaments in neurodegenerative diseases, termed tauopathies. These tau inclusions are pathological hallmarks of Alzheimer's disease (AD), Pick's disease (PiD), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP). The neuropathological hallmarks of these diseases burden several cell types within the CNS, and have also been shown to be abundantly phosphorylated. The mechanism(s) by which tau aggregates in the CNS is not fully known, but it is hypothesized that hyperphosphorylated tau may precede and further promote filament formation, leading to the production of these pathological inclusions. In the studies herein, we generated and thoroughly characterized two novel conformation-dependent tau monoclonal antibodies that bind to residues Pro218-Glu222, but are sensitive to denaturing conditions and highly modulated by adjacent downstream phosphorylation sites. These epitopes are present in the neuropathological hallmarks of several tauopathies, including AD, PiD, CBD, and PSP. These novel antibodies will further enable investigation of tau-dependent pathological inclusion formation and enhance our understanding of the phosphorylation signatures within tauopathies with the possibility of new biomarker developments.
Collapse
Affiliation(s)
- Giavanna Paterno
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Jose Torrellas
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Brach M. Bell
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Kimberly-Marie M. Gorion
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Stephan S. Quintin
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
| | - Gabriela P. Hery
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
- Department of Pathology, 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; (G.P.); (J.T.); (B.M.B.); (K.-M.M.G.); (S.S.Q.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.H.); (S.P.)
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
9
|
Zhang ZY, Harischandra DS, Wang R, Ghaisas S, Zhao JY, McMonagle TP, Zhu G, Lacuarta KD, Song J, Trojanowski JQ, Xu H, Lee VMY, Yang X. TRIM11 protects against tauopathies and is down-regulated in Alzheimer's disease. Science 2023; 381:eadd6696. [PMID: 37499037 DOI: 10.1126/science.add6696] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 06/01/2023] [Indexed: 07/29/2023]
Abstract
Aggregation of tau into filamentous inclusions underlies Alzheimer's disease (AD) and numerous other neurodegenerative tauopathies. The pathogenesis of tauopathies remains unclear, which impedes the development of disease-modifying treatments. Here, by systematically analyzing human tripartite motif (TRIM) proteins, we identified a few TRIMs that could potently inhibit tau aggregation. Among them, TRIM11 was markedly down-regulated in AD brains. TRIM11 promoted the proteasomal degradation of mutant tau as well as superfluous normal tau. It also enhanced tau solubility by acting as both a molecular chaperone to prevent tau misfolding and a disaggregase to dissolve preformed tau fibrils. TRIM11 maintained the connectivity and viability of neurons. Intracranial delivery of TRIM11 through adeno-associated viruses ameliorated pathology, neuroinflammation, and cognitive impairments in multiple animal models of tauopathies. These results suggest that TRIM11 down-regulation contributes to the pathogenesis of tauopathies and that restoring TRIM11 expression may represent an effective therapeutic strategy.
Collapse
Affiliation(s)
- Zi-Yang Zhang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dilshan S Harischandra
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ruifang Wang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shivani Ghaisas
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janet Y Zhao
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas P McMonagle
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guixin Zhu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenzo D Lacuarta
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jianing Song
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hong Xu
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaolu Yang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Karimi N, Bayram Çatak F, Arslan E, Saghazadeh A, Rezaei N. Tau immunotherapy in Alzheimer’s disease and progressive supranuclear palsy. Int Immunopharmacol 2022; 113:109445. [DOI: 10.1016/j.intimp.2022.109445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
|
11
|
Bittar A, Al-Lahham R, Bhatt N, Moore K, Montalbano M, Jerez C, Fung L, McAllen S, Ellsworth A, Kayed R. Passive Immunotherapy Targeting Tau Oligomeric Strains Reverses Tauopathy Phenotypes in Aged Human-Tau Mice in a Mouse Model-Specific Manner. J Alzheimers Dis 2022; 90:1103-1122. [DOI: 10.3233/jad-220518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Tau oligomers are one of the most toxic species, displaying prion-like strains which have different conformations resulting in different tauopathies. Passive immunotherapy targeting different tau species is a promising therapeutic approach. Age is one of the greatest risk factors; however, most immunotherapy studies are done in young to middle-aged mice tauopathy models, which is not representative of the many clinical trials done with older humans with established tauopathies. Objective: We utilized two different clones of tau oligomer monoclonal antibodies (TOMAs) in aged Htau and JNPL3 mouse models to investigate the potential of passive immunotherapy. Methods: Aged mice received a single intravenous injection of 120 μg/animal of either TOMA1, TOMA3 clones or a non-specific IgG. Their cognitive functions were assessed one-week post-injection using Y-maze and novel object recognition tests. Brain tissues were analyzed using biochemical and immunological assays. Results: TOMA 1 and 3 rescues cognitive phenotypes in aged animals in a mouse model-specific manner, indicative by a reduction in tau oligomers levels. The TOMAs were shown to have strong reactivity with different tau oligomeric species in the different mouse models in vitro and ex vivo. Conclusion: This is the first study testing tau passive immunotherapy in aged animals and supports our previous reports on of the role of oligomeric tau in disease progression further validating the potential of TOMAs to rescue the late-stage disease pathology and phenotype. Moreover, this study suggests that multiple tau oligomeric strains exist in aged animals; therefore, it is of great importance to further characterize these strains.
Collapse
Affiliation(s)
- Alice Bittar
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rabab Al-Lahham
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kenya Moore
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Leiana Fung
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Anna Ellsworth
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
12
|
Vagenknecht P, Luzgin A, Ono M, Ji B, Higuchi M, Noain D, Maschio CA, Sobek J, Chen Z, Konietzko U, Gerez JA, Riek R, Razansky D, Klohs J, Nitsch RM, Dean-Ben XL, Ni R. Non-invasive imaging of tau-targeted probe uptake by whole brain multi-spectral optoacoustic tomography. Eur J Nucl Med Mol Imaging 2022; 49:2137-2152. [PMID: 35128565 PMCID: PMC9165274 DOI: 10.1007/s00259-022-05708-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Abnormal tau accumulation within the brain plays an important role in tauopathies such as Alzheimer's disease and frontotemporal dementia. High-resolution imaging of tau deposits at the whole-brain scale in animal disease models is highly desired. METHODS We approached this challenge by non-invasively imaging the brains of P301L mice of 4-repeat tau with concurrent volumetric multi-spectral optoacoustic tomography (vMSOT) at ~ 115 μm spatial resolution using the tau-targeted pyridinyl-butadienyl-benzothiazole derivative PBB5 (i.v.). In vitro probe characterization, concurrent vMSOT and epi-fluorescence imaging of in vivo PBB5 targeting (i.v.) was performed in P301L and wild-type mice, followed by ex vivo validation using AT-8 antibody for phosphorylated tau. RESULTS PBB5 showed specific binding to recombinant K18 tau fibrils by fluorescence assay, to post-mortem Alzheimer's disease brain tissue homogenate by competitive binding against [11C]PBB3 and to tau deposits (AT-8 positive) in post-mortem corticobasal degeneration and progressive supranuclear palsy brains. Dose-dependent optoacoustic and fluorescence signal intensities were observed in the mouse brains following i.v. administration of different concentrations of PBB5. In vivo vMSOT brain imaging of P301L mice showed higher retention of PBB5 in the tau-laden cortex and hippocampus compared to wild-type mice, as confirmed by ex vivo vMSOT, epi-fluorescence, multiphoton microscopy, and immunofluorescence staining. CONCLUSIONS We demonstrated non-invasive whole-brain imaging of tau in P301L mice with vMSOT system using PBB5 at a previously unachieved ~ 115 μm spatial resolution. This platform provides a new tool to study tau spreading and clearance in a tauopathy mouse model, foreseeable in monitoring tau targeting putative therapeutics.
Collapse
Affiliation(s)
- Patrick Vagenknecht
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Artur Luzgin
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Maiko Ono
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Bin Ji
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Makoto Higuchi
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Daniela Noain
- Neurology Department, University Hospital Zurich, Zurich, Switzerland
| | - Cinzia A Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
| | - Jens Sobek
- Functional Genomics Center, University of Zurich, Zurich, Switzerland
| | - Zhenyue Chen
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Uwe Konietzko
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Juan A Gerez
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Roland Riek
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Jan Klohs
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland
| | - Xose Luis Dean-Ben
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland.
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.
- Zentrum für Neurowissenschaften Zürich (ZNZ), Zurich, Switzerland.
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, ETH Zurich & University of Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Congdon EE, Jiang Y, Sigurdsson EM. Targeting tau only extracellularly is likely to be less efficacious than targeting it both intra- and extracellularly. Semin Cell Dev Biol 2022; 126:125-137. [PMID: 34896021 PMCID: PMC9680670 DOI: 10.1016/j.semcdb.2021.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
Aggregation of the tau protein is thought to be responsible for the neurodegeneration and subsequent functional impairments in diseases that are collectively named tauopathies. Alzheimer's disease is the most common tauopathy, but the group consists of over 20 different diseases, many of which have tau pathology as their primary feature. The development of tau therapies has mainly focused on preventing the formation of and/or clearing these aggregates. Of these, immunotherapies that aim to either elicit endogenous tau antibodies or deliver exogenous ones are the most common approach in clinical trials. While their mechanism of action can involve several pathways, both extra- and intracellular, pharmaceutical companies have primarily focused on antibody-mediated clearance of extracellular tau. As we have pointed out over the years, this is rather surprising because it is well known that most of pathological tau protein is found intracellularly. It has been repeatedly shown by several groups over the past decades that antibodies can enter neurons and that their cellular uptake can be enhanced by various means, particularly by altering their charge. Here, we will briefly describe the potential extra- and intracellular mechanisms involved in antibody-mediated clearance of tau pathology, discuss these in the context of recent failures of some of the tau antibody trials, and finally provide a brief overview of how the intracellular efficacy of tau antibodies can potentially be further improved by certain modifications that aim to enhance tau clearance via specific intracellular degradation pathways.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States.
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
14
|
Dregni AJ, Duan P, Xu H, Changolkar L, El Mammeri N, Lee VMY, Hong M. Fluent molecular mixing of Tau isoforms in Alzheimer's disease neurofibrillary tangles. Nat Commun 2022; 13:2967. [PMID: 35624093 PMCID: PMC9142584 DOI: 10.1038/s41467-022-30585-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/04/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is defined by intracellular neurofibrillary tangles formed by the microtubule-associated protein tau and extracellular plaques formed by the β-amyloid peptide. AD tau tangles contain a mixture of tau isoforms with either four (4R) or three (3R) microtubule-binding repeats. Here we use solid-state NMR to determine how 4R and 3R tau isoforms mix at the molecular level in AD tau aggregates. By seeding differentially isotopically labeled 4R and 3R tau monomers with AD brain-derived tau, we measured intermolecular contacts of the two isoforms. The NMR data indicate that 4R and 3R tau are well mixed in the AD-tau seeded fibrils, with a 60:40 incorporation ratio of 4R to 3R tau and a small homotypic preference. The AD-tau templated 4R tau, 3R tau, and mixed 4R and 3R tau fibrils exhibit no structural differences in the rigid β-sheet core or the mobile domains. Therefore, 4R and 3R tau are fluently recruited into the pathological fold of AD tau aggregates, which may explain the predominance of AD among neurodegenerative disorders.
Collapse
Affiliation(s)
- Aurelio J Dregni
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA
| | - Pu Duan
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA
| | - Hong Xu
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Lakshmi Changolkar
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Nadia El Mammeri
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
15
|
Tashima T. Delivery of Intravenously Administered Antibodies Targeting Alzheimer's Disease-Relevant Tau Species into the Brain Based on Receptor-Mediated Transcytosis. Pharmaceutics 2022; 14:411. [PMID: 35214143 PMCID: PMC8876001 DOI: 10.3390/pharmaceutics14020411] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 01/23/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that causes memory loss, cognitive decline, and eventually dementia. The etiology of AD and its pathological mechanisms remain unclear due to its complex pathobiology. At the same time, the number of patients with AD is increasing worldwide. However, no therapeutic agents for AD are currently available for definitive care. Several phase 3 clinical trials using agents targeting amyloid β (Aβ) and its related molecules have failed, with the exception of aducanumab, an anti-Aβ monoclonal antibody (mAb), clinically approved by the US Food and Drug Administration in 2021, which could be modified for AD drug development due to controversial approval. Neurofibrillary tangles (NFTs) composed of tau rather than senile plaques composed of Aβ are correlated with AD pathogenesis. Moreover, Aβ and tau pathologies initially proceed independently. At a certain point in the progression of AD symptoms, the Aβ pathology is involved in the alteration and spreading of the tau pathology. Therefore, tau-targeting therapies have attracted the attention of pharmaceutical scientists, as well as Aβ-targeting therapies. In this review, I introduce the implementations and potential of AD immunotherapy using intravenously administered anti-tau and anti-receptor bispecific mAbs. These cross the blood-brain barrier (BBB) based on receptor-mediated transcytosis and are subsequently cleared by microglia based on Fc-mediated endocytosis after binding to tau and lysosomal degradation.
Collapse
Affiliation(s)
- Toshihiko Tashima
- Tashima Laboratories of Arts and Sciences, 1239-5 Toriyama-cho, Kohoku-ku, Yokohama 222-0035, Japan
| |
Collapse
|
16
|
Benn JA, Mukadam AS, McEwan WA. Targeted protein degradation using intracellular antibodies and its application to neurodegenerative disease. Semin Cell Dev Biol 2021; 126:138-149. [PMID: 34654628 DOI: 10.1016/j.semcdb.2021.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 01/10/2023]
Abstract
Antibodies mediate the majority of their effects in the extracellular domain, or in intracellular compartments isolated from the cytosol. Under a growing list of circumstances, however, antibodies are found to gain access to the cytoplasm. Cytosolic immune complexes are bound by the atypical antibody receptor TRIM21, which mediates the rapid degradation of the immune complexes at the proteasome. These discoveries have informed the development of TRIM-Away, a technique to selectively deplete proteins using delivery of antibodies into cells. A range of related approaches that elicit selective protein degradation using intracellular constructs linking antibody fragments to degradative effector functions have also been developed. These methods hold promise for inducing the degradation of proteins as both research tools and as a novel therapeutic approach. Protein aggregates are a pathophysiological feature of neurodegenerative diseases and are considered to have a causal role in pathology. Immunotherapy is emerging as a promising route towards their selective targeting, and a role of antibodies in the cytosol has been demonstrated in cell-based assays. This review will explore the mechanisms by which therapeutic antibodies engage and eliminate intracellularly aggregated proteins. We will discuss how future developments in intracellular antibody technology may enhance the therapeutic potential of such antibody-derived therapies.
Collapse
Affiliation(s)
- Jonathan A Benn
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, UK
| | - Aamir S Mukadam
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, UK
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, UK.
| |
Collapse
|
17
|
Shimogawa M, Petersson EJ. New strategies for fluorescently labeling proteins in the study of amyloids. Curr Opin Chem Biol 2021; 64:57-66. [PMID: 34091264 PMCID: PMC8585672 DOI: 10.1016/j.cbpa.2021.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 01/25/2023]
Abstract
Amyloid proteins are widely studied, both for their unusual biophysical properties and their association with disorders such as Alzheimer's and Parkinson's disease. Fluorescence-based methods using site-specifically labeled proteins can provide information on the details of their structural dynamics and their roles in specific biological processes. Here, we describe the application of different labeling methods and novel fluorescent probe strategies to the study of amyloid proteins, both for in vitro biophysical experiments and for in vivo imaging. These labeling tools can be elegantly used to answer important questions on the function and pathology of amyloid proteins.
Collapse
Affiliation(s)
- Marie Shimogawa
- Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, PA, 19104, USA
| | - E James Petersson
- Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
18
|
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
|
19
|
Darling AL, Shorter J. Combating deleterious phase transitions in neurodegenerative disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2021; 1868:118984. [PMID: 33549703 PMCID: PMC7965345 DOI: 10.1016/j.bbamcr.2021.118984] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022]
Abstract
Protein aggregation is a hallmark of neurodegenerative diseases. However, the mechanism that induces pathogenic aggregation is not well understood. Recently, it has emerged that several of the pathological proteins found in an aggregated or mislocalized state in neurodegenerative diseases are also able to undergo liquid-liquid phase separation (LLPS) under physiological conditions. Although these phase transitions are likely important for various physiological functions, neurodegenerative disease-related mutations and conditions can alter the LLPS behavior of these proteins, which can elicit toxicity. Therefore, therapeutics that antagonize aberrant LLPS may be able to mitigate toxicity and aggregation that is ubiquitous in neurodegenerative disease. Here, we discuss the mechanisms by which aberrant protein phase transitions may contribute to neurodegenerative disease. We also outline potential therapeutic strategies to counter deleterious phases. State without borders: Membrane-less organelles and liquid-liquid phase transitions edited by Vladimir N Uversky.
Collapse
Affiliation(s)
- April L Darling
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Bachstetter AD, Garrett FG, Jicha GA, Nelson PT. Space-occupying brain lesions, trauma-related tau astrogliopathy, and ARTAG: a report of two cases and a literature review. Acta Neuropathol Commun 2021; 9:49. [PMID: 33757579 PMCID: PMC7986305 DOI: 10.1186/s40478-021-01152-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes with intracellular accumulations of misfolded phosphorylated tau protein have been observed in advanced-stage chronic traumatic encephalopathy (CTE) and in other neurodegenerative conditions. There is a growing awareness that astrocytic tau inclusions are also relatively common in the brains of persons over 70 years of age-affecting approximately one-third of autopsied individuals. The pathologic hallmarks of aging-related tau astrogliopathy (ARTAG) include phosphorylated tau protein within thorn-shaped astrocytes (TSA) in subpial, subependymal, perivascular, and white matter regions, whereas granular-fuzzy astrocytes are often seen in gray matter. CTE and ARTAG share molecular and histopathologic characteristics, suggesting that trauma-related mechanism(s) may predispose to the development of tau astrogliopathy. There are presently few experimental systems to study the pathobiology of astrocytic-tau aggregation, but human studies have made recent progress. For example, leucotomy (also referred to as lobotomy) is associated with a localized ARTAG-like neuropathology decades after the surgical brain injury, suggesting that chronic brain injury of any type may predispose to later life ARTAG. To examine this idea in a different context, we report clinical and pathologic features of two middle-aged men who came to autopsy with large (> 6 cm in greatest dimension) arachnoid cysts that had physically displaced and injured the subjects' left temporal lobes through chronic mechanical stress. Despite the similarity of the size and location of the arachnoid cysts, these individuals had dissimilar neurologic outcomes and neuropathologic findings. We review the evidence for ARTAG in response to brain injury, and discuss how the location and molecular properties of astroglial tau inclusions might alter the physiology of resident astrocytes. These cases and literature review point toward possible mechanism(s) of tau aggregation in astrocytes in response to chronic brain trauma.
Collapse
Affiliation(s)
- Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA.
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
| | - Filip G Garrett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|