1
|
Yao Y, Jin C, Liao Y, Huang X, Wei Z, Zhang Y, Li D, Su H, Han W, Qin D. Schizophrenia-Like Behaviors Arising from Dysregulated Proline Metabolism Are Associated with Altered Neuronal Morphology and Function in Mice with Hippocampal PRODH Deficiency. Aging Dis 2024; 15:1952-1968. [PMID: 37815900 PMCID: PMC11272211 DOI: 10.14336/ad.2023.0902] [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: 06/01/2023] [Accepted: 09/02/2023] [Indexed: 10/12/2023] Open
Abstract
Despite decades of research being conducted to understand what physiological deficits in the brain are an underlying basis of psychiatric diseases like schizophrenia, it has remained difficult to establish a direct causal relationship between neuronal dysfunction and specific behavioral phenotypes. Moreover, it remains unclear how metabolic processes, including amino acid metabolism, affect neuronal function and consequently modulate animal behaviors. PRODH, which catalyzes the first step of proline degradation, has been reported as a susceptibility gene for schizophrenia. It has consistently been shown that PRODH knockout mice exhibit schizophrenia-like behaviors. However, whether the loss of PRODH directly impacts neuronal function or whether such neuronal deficits are linked to schizophrenia-like behaviors has not yet been examined. Herein, we first ascertained that dysregulated proline metabolism in humans is associated with schizophrenia. We then found that PRODH was highly expressed in the oreins layer of the mouse dorsal hippocampus. By using AAV-mediated shRNA, we depleted PRODH expression in the mouse dorsal hippocampus and subsequently observed hyperactivity and impairments in the social behaviors, learning, and memory of these mice. Furthermore, the loss of PRODH led to altered neuronal morphology and function both in vivo and in vitro. Our study demonstrates that schizophrenia-like behaviors may arise from dysregulated proline metabolism due to the loss of PRODH and are associated with altered neuronal morphology and function in mice.
Collapse
Affiliation(s)
- Yuxiao Yao
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China.
| | - Chenchen Jin
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Yilie Liao
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Xiang Huang
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Ziying Wei
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China.
| | - Yahong Zhang
- Guangzhou Laboratory, Guangzhou, Guangdong, China.
| | - Dongwei Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China.
| | - Huanxing Su
- Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| | - Weiping Han
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China.
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences; Hong Kong SAR, China.
| |
Collapse
|
2
|
Rose K, Jepson T, Shukla S, Maya-Romero A, Kampmann M, Xu K, Hurley JH. Tau fibrils induce nanoscale membrane damage and nucleate cytosolic tau at lysosomes. Proc Natl Acad Sci U S A 2024; 121:e2315690121. [PMID: 38781206 PMCID: PMC11145263 DOI: 10.1073/pnas.2315690121] [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: 09/09/2023] [Accepted: 04/08/2024] [Indexed: 05/25/2024] Open
Abstract
The prion-like spread of protein aggregates is a leading hypothesis for the propagation of neurofibrillary lesions in the brain, including the spread of tau inclusions associated with Alzheimer's disease. The mechanisms of cellular uptake of tau seeds and subsequent nucleated polymerization of cytosolic tau are major questions in the field, and the potential for coupling between the entry and nucleation mechanisms has been little explored. We found that in primary astrocytes and neurons, endocytosis of tau seeds leads to their accumulation in lysosomes. This in turn leads to lysosomal swelling, deacidification, and recruitment of ESCRT proteins, but not Galectin-3, to the lysosomal membrane. These observations are consistent with nanoscale damage of the lysosomal membrane. Live cell imaging and STORM superresolution microscopy further show that the nucleation of cytosolic tau occurs primarily at the lysosome membrane under these conditions. These data suggest that tau seeds escape from lysosomes via nanoscale damage rather than wholesale rupture and that nucleation of cytosolic tau commences as soon as tau fibril ends emerge from the lysosomal membrane.
Collapse
Affiliation(s)
- Kevin Rose
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
| | - Tyler Jepson
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
- Graduate Group in Biophysics, University of California, Berkeley, CA94720
| | - Sankalp Shukla
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
| | - Alex Maya-Romero
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA94158
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA94158
| | - Ke Xu
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
- Graduate Group in Biophysics, University of California, Berkeley, CA94720
- Department of Chemistry, University of California, Berkeley, CA94720
| | - James H. Hurley
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
- Graduate Group in Biophysics, University of California, Berkeley, CA94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA94720
| |
Collapse
|
3
|
Forrest SL, Lee S, Nassir N, Martinez-Valbuena I, Sackmann V, Li J, Ahmed A, Tartaglia MC, Ittner LM, Lang AE, Uddin M, Kovacs GG. Cell-specific MAPT gene expression is preserved in neuronal and glial tau cytopathologies in progressive supranuclear palsy. Acta Neuropathol 2023; 146:395-414. [PMID: 37354322 PMCID: PMC10412651 DOI: 10.1007/s00401-023-02604-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
Microtubule-associated protein tau (MAPT) aggregates in neurons, astrocytes and oligodendrocytes in a number of neurodegenerative diseases, including progressive supranuclear palsy (PSP). Tau is a target of therapy and the strategy includes either the elimination of pathological tau aggregates or reducing MAPT expression, and thus the amount of tau protein made to prevent its aggregation. Disease-associated tau affects brain regions in a sequential manner that includes cell-to-cell spreading. Involvement of glial cells that show tau aggregates is interpreted as glial cells taking up misfolded tau assuming that glial cells do not express enough MAPT. Although studies have evaluated MAPT expression in human brain tissue homogenates, it is not clear whether MAPT expression is compromised in cells accumulating pathological tau. To address these perplexing aspects of disease pathogenesis, this study used RNAscope combined with immunofluorescence (AT8), and single-nuclear(sn) RNAseq to systematically map and quantify MAPT expression dynamics across different cell types and brain regions in controls (n = 3) and evaluated whether tau cytopathology affects MAPT expression in PSP (n = 3). MAPT transcripts were detected in neurons, astrocytes and oligodendrocytes, and varied between brain regions and within each cell type, and were preserved in all cell types with tau aggregates in PSP. These results propose a complex scenario in all cell types, where, in addition to the ingested misfolded tau, the preserved cellular MAPT expression provides a pool for local protein production that can (1) be phosphorylated and aggregated, or (2) feed the seeding of ingested misfolded tau by providing physiological tau, both accentuating the pathological process. Since tau cytopathology does not compromise MAPT gene expression in PSP, a complete loss of tau protein expression as an early pathogenic component is less likely. These observations provide rationale for a dual approach to therapy by decreasing cellular MAPT expression and targeting removal of misfolded tau.
Collapse
Affiliation(s)
- Shelley L Forrest
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Nasna Nassir
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Ivan Martinez-Valbuena
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Valerie Sackmann
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Jun Li
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
| | - Awab Ahmed
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada
- University Health Network Memory Clinic, Krembil Brain Institute, Toronto, ON, Canada
| | - Lars M Ittner
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
| | - Mohammed Uddin
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
- Cellular Intelligence (Ci) Lab, GenomeArc Inc., Toronto, ON, Canada
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease (CRND), University of Toronto, Krembil Discovery Tower, 60 Leonard Ave, Toronto, ON, M5T 0S8, Canada.
- Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada.
- Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology and Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Rose K, Jepson T, Shukla S, Maya-Romero A, Kampmann M, Xu K, Hurley JH. Tau fibrils induce nanoscale membrane damage and nucleate cytosolic tau at lysosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555157. [PMID: 37693477 PMCID: PMC10491128 DOI: 10.1101/2023.08.28.555157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The prion-like spread of protein aggregates is a leading hypothesis for the propagation of neurofibrillary lesions in the brain, including the spread of tau inclusions associated with Alzheimer's disease. The mechanisms of cellular uptake of tau seeds and subsequent nucleated polymerization of cytosolic tau are major questions in the field, and the potential for coupling between the entry and nucleation mechanisms has been little explored. We found that in primary astrocytes, endocytosis of tau seeds leads to their accumulation in lysosomes. This in turn leads to lysosomal swelling, deacidification and recruitment of ESCRT proteins, but not Galectin-3, to the lysosomal membrane. These observations are consistent with nanoscale damage of the lysosomal membrane. Using live cell and STORM, imaging, nucleation of cytosolic tau occurs primarily at the lysosome membrane under these conditions. These data suggest that tau seeds escape from lysosomes via nanoscale damage rather than wholesale rupture, and that nucleation of cytosolic tau commences as soon as tau fibril ends emerge from the lysosomal membrane.
Collapse
Affiliation(s)
- Kevin Rose
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
| | - Tyler Jepson
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, University of California, Berkeley, CA 94720
| | - Sankalp Shukla
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
| | - Alex Maya-Romero
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California 94158
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158
| | - Ke Xu
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, University of California, Berkeley, CA 94720
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - James H. Hurley
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, University of California, Berkeley, CA 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720
| |
Collapse
|
5
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [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: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
6
|
Torii T, Miyamoto Y, Nakata R, Higashi Y, Shinmyo Y, Kawasaki H, Miyasaka T, Misonou H. Identification of Tau protein as a novel marker for maturation and pathological changes of oligodendrocytes. Glia 2023; 71:1002-1017. [PMID: 36565228 DOI: 10.1002/glia.24322] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/25/2022]
Abstract
Microtubule-associated protein Tau is primarily expressed in axons of neurons, but also in Olig2-positive oligodendrocytes in adult rodent and monkey brains. In this study, we sought to determine at what cell stage Tau becomes expressed in the oligodendrocyte lineage. We performed immunostaining of adult mouse brain sections using well-known markers of oligodendrocyte lineage and found that Tau is expressed in mature oligodendrocytes, but not in oligodendrocyte progenitors and immature pre-oligodendrocytes. We also investigated Tau expression in developing mouse brain. Surprisingly, Tau expression occurred after the peak of myelination and even exceeded GSTπ expression, which has been considered as a marker of myelinating oligodendrocytes. These results suggest Tau as a novel marker of oligodendrocyte maturation. We then investigated whether Tau is important for oligodendrocyte development and/or myelination and how Tau changes in demyelination. First, we found no changes in myelination and oligodendrocyte markers in Tau knockout mice, suggesting that Tau is dispensable. Next, we analyzed the proteolipid protein 1 transgenic model of Pelizaeus-Merzbacher disease, which is a rare leukodystrophy. In hemizygous transgenic mice, the number of Tau-positive cells were significantly increased as compared with wild type mice. These cells were also positive for Olig2, CC1, and GSTπ, but not PDGFRα and GPR17. In stark contrast, the expression level of Tau, as well as GSTπ, was dramatically decreased in the cuprizone-induced model of multiple sclerosis. Taken together, we propose Tau as a new marker of oligodendrocyte lineage and for investigating demyelination lesions.
Collapse
Affiliation(s)
- Tomohiro Torii
- Laboratory of Ion Channel Pathophysiology, Graduate School of Brain Science, Doshisha University, Kyotanabe-shi, Kyoto, Japan.,Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| | - Yuki Miyamoto
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagayaku, Tokyo, Japan
| | - Rinaho Nakata
- Laboratory of Ion Channel Pathophysiology, Graduate School of Brain Science, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| | - Yuto Higashi
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa-shi, Ishikawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa-shi, Ishikawa, Japan
| | - Tomohiro Miyasaka
- Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe-shi, Kyoto, Japan.,Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| | - Hiroaki Misonou
- Laboratory of Ion Channel Pathophysiology, Graduate School of Brain Science, Doshisha University, Kyotanabe-shi, Kyoto, Japan.,Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| |
Collapse
|
7
|
Perbet R, Zufferey V, Leroux E, Parietti E, Espourteille J, Culebras L, Perriot S, Du Pasquier R, Bégard S, Deramecourt V, Déglon N, Toni N, Buée L, Colin M, Richetin K. Tau Transfer via Extracellular Vesicles Disturbs the Astrocytic Mitochondrial System. Cells 2023; 12:cells12070985. [PMID: 37048058 PMCID: PMC10093208 DOI: 10.3390/cells12070985] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/08/2023] [Accepted: 03/18/2023] [Indexed: 04/14/2023] Open
Abstract
Tauopathies are neurodegenerative disorders involving the accumulation of tau isoforms in cell subpopulations such as astrocytes. The origins of the 3R and 4R isoforms of tau that accumulate in astrocytes remain unclear. Extracellular vesicles (EVs) were isolated from primary neurons overexpressing 1N3R or 1N4R tau or from human brain extracts (progressive supranuclear palsy or Pick disease patients or controls) and characterized (electron microscopy, nanoparticle tracking analysis (NTA), proteomics). After the isolated EVs were added to primary astrocytes or human iPSC-derived astrocytes, tau transfer and mitochondrial system function were evaluated (ELISA, immunofluorescence, MitoTracker staining). We demonstrated that neurons in which 3R or 4R tau accumulated had the capacity to transfer tau to astrocytes and that EVs were essential for the propagation of both isoforms of tau. Treatment with tau-containing EVs disrupted the astrocytic mitochondrial system, altering mitochondrial morphology, dynamics, and redox state. Although similar levels of 3R and 4R tau were transferred, 3R tau-containing EVs were significantly more damaging to astrocytes than 4R tau-containing EVs. Moreover, EVs isolated from the brain fluid of patients with different tauopathies affected mitochondrial function in astrocytes derived from human iPSCs. Our data indicate that tau pathology spreads to surrounding astrocytes via EVs-mediated transfer and modifies their function.
Collapse
Affiliation(s)
- Romain Perbet
- Univ. Lille, Inserm, CHU Lille, LilNCog-Lille Neuroscience & Cognition, 59000 Lille, France
| | - Valentin Zufferey
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Elodie Leroux
- Univ. Lille, Inserm, CHU Lille, LilNCog-Lille Neuroscience & Cognition, 59000 Lille, France
| | - Enea Parietti
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Jeanne Espourteille
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Lucas Culebras
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Sylvain Perriot
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, 1011 Lausanne, Switzerland
| | - Renaud Du Pasquier
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, 1011 Lausanne, Switzerland
| | - Séverine Bégard
- Univ. Lille, Inserm, CHU Lille, LilNCog-Lille Neuroscience & Cognition, 59000 Lille, France
| | - Vincent Deramecourt
- Univ. Lille, Inserm, CHU Lille, LilNCog-Lille Neuroscience & Cognition, 59000 Lille, France
| | - Nicole Déglon
- Lausanne University Hospital (CHUV) and University of Lausanne, Neuroscience Research Center (CRN), Laboratory of Neurotherapies and Neuromodulation, 1011 Lausanne, Switzerland
| | - Nicolas Toni
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, LilNCog-Lille Neuroscience & Cognition, 59000 Lille, France
| | - Morvane Colin
- Univ. Lille, Inserm, CHU Lille, LilNCog-Lille Neuroscience & Cognition, 59000 Lille, France
| | - Kevin Richetin
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
8
|
Bustos LM, Sattler R. The Fault in Our Astrocytes - cause or casualties of proteinopathies of ALS/FTD and other neurodegenerative diseases? FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1075805. [PMID: 39165755 PMCID: PMC11334001 DOI: 10.3389/fmmed.2023.1075805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/08/2023] [Indexed: 08/22/2024]
Abstract
Many neurodegenerative diseases fall under the class of diseases known as proteinopathies, whereby the structure and localization of specific proteins become abnormal. These aberrant proteins often aggregate within cells which disrupts vital homeostatic and physiological cellular functions, ultimately contributing to cell death. Although neurodegenerative disease research is typically neurocentric, there is evidence supporting the role of non-neuronal cells in the pathogenesis of these diseases. Specifically, the role of astrocytes in neurodegenerative diseases has been an ever-growing area of research. Astrocytes are one of the most abundant cell types in the central nervous system (CNS) and provide an array of essential homeostatic functions that are disrupted in neurodegenerative diseases. Astrocytes can exhibit a reactive phenotype that is characterized by molecular changes, as well as changes in morphology and function. In neurodegenerative diseases, there is potential for reactive astrocytes to assume a loss-of-function phenotype in homeostatic operations such as synapse maintenance, neuronal metabolic support, and facilitating cell-cell communication between glia and neurons. They are also able to concurrently exhibit gain-of-function phenotypes that can be destructive to neural networks and the astrocytes themselves. Additionally, astrocytes have been shown to internalize disease related proteins and reflect similar or exacerbated pathology that has been observed in neurons. Here, we review several major neurodegenerative disease-specific proteinopathies and what is known about their presence in astrocytes and the potential consequences regarding cell and non-cell autonomous neurodegeneration.
Collapse
Affiliation(s)
- Lynette M. Bustos
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Barrow Neurological Institute, Phoenix, AZ, United States
| | - Rita Sattler
- Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
9
|
Mahakud AK, Shaikh J, Rifa Iqbal VV, Gupta A, Tiwari A, Saleem M. Amyloids on Membrane Interfaces: Implications for Neurodegeneration. J Membr Biol 2022; 255:705-722. [PMID: 35670831 DOI: 10.1007/s00232-022-00245-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/12/2022] [Indexed: 12/24/2022]
Abstract
Membrane interfaces are vital for various cellular processes, and their involvement in neurodegenerative disorders such as Alzheimer's and Parkinson's disease has taken precedence in recent years. The amyloidogenic proteins associated with neurodegenerative diseases interact with the neuronal membrane through various means, which has implications for both the onset and progression of the disease. The parameters that regulate the interaction between the membrane and the amyloids remain poorly understood. The review focuses on the various aspects of membrane interactions of amyloids, particularly amyloid-β (Aβ) peptides and Tau involved in Alzheimer's and α-synuclein involved in Parkinson's disease. The genetic, cell biological, biochemical, and biophysical studies that form the basis for our current understanding of the membrane interactions of Aβ peptides, Tau, and α-synuclein are discussed.
Collapse
Affiliation(s)
- Amaresh Kumar Mahakud
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - Jafarulla Shaikh
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - V V Rifa Iqbal
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - Abhinav Gupta
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India.,Homi Bhabha National Institute, Mumbai, India
| | - Anuj Tiwari
- Department of Life Sciences, National Institute of Technology, Rourkela, India
| | - Mohammed Saleem
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India. .,Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
10
|
Hromadkova L, Siddiqi MK, Liu H, Safar JG. Populations of Tau Conformers Drive Prion-like Strain Effects in Alzheimer's Disease and Related Dementias. Cells 2022; 11:2997. [PMID: 36230957 PMCID: PMC9562632 DOI: 10.3390/cells11192997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Recent findings of diverse populations of prion-like conformers of misfolded tau protein expand the prion concept to Alzheimer's disease (AD) and monogenic frontotemporal lobar degeneration (FTLD)-MAPT P301L, and suggest that distinct strains of misfolded proteins drive the phenotypes and progression rates in many neurodegenerative diseases. Notable progress in the previous decades has generated many lines of proof arguing that yeast, fungal, and mammalian prions determine heritable as well as infectious traits. The extraordinary phenotypic diversity of human prion diseases arises from structurally distinct prion strains that target, at different progression speeds, variable brain structures and cells. Although human prion research presents beneficial lessons and methods to study the mechanism of strain diversity of protein-only pathogens, the fundamental molecular mechanism by which tau conformers are formed and replicate in diverse tauopathies is still poorly understood. In this review, we summarize up to date advances in identification of diverse tau conformers through biophysical and cellular experimental paradigms, and the impact of heterogeneity of pathological tau strains on personalized structure- and strain-specific therapeutic approaches in major tauopathies.
Collapse
Affiliation(s)
- Lenka Hromadkova
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - He Liu
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jiri G. Safar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
11
|
Mai M, Guo X, Huang Y, Zhang W, Xu Y, Zhang Y, Bai X, Wu J, Zu H. DHCR24 Knockdown Induces Tau Hyperphosphorylation at Thr181, Ser199, Ser262, and Ser396 Sites via Activation of the Lipid Raft-Dependent Ras/MEK/ERK Signaling Pathway in C8D1A Astrocytes. Mol Neurobiol 2022; 59:5856-5873. [PMID: 35804281 PMCID: PMC9395500 DOI: 10.1007/s12035-022-02945-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 06/28/2022] [Indexed: 02/01/2023]
Abstract
The synthetase 3β-hydroxysterol-Δ24 reductase (DHCR24) is a key regulator involved in cholesterol synthesis and homeostasis. A growing body of evidence indicates that DHCR24 is downregulated in the brain of various models of Alzheimer's disease (AD), such as astrocytes isolated from AD mice. For the past decades, astrocytic tau pathology has been found in AD patients, while the origin of phosphorylated tau in astrocytes remains unknown. A previous study suggests that downregulation of DHCR24 is associated with neuronal tau hyperphosphorylation. Herein, the present study is to explore whether DHCR24 deficiency can also affect tau phosphorylation in astrocytes. Here, we showed that DHCR24 knockdown could induce tau hyperphosphorylation at Thr181, Ser199, Thr231, Ser262, and Ser396 sites in C8D1A astrocytes. Meanwhile, we found that DHCR24-silencing cells had reduced the level of free cholesterol in the plasma membrane and intracellular organelles, as well as cholesterol esters. Furthermore, reduced cellular cholesterol level caused a decreased level of the caveolae-associated protein, cavin1, which disrupted lipid rafts/caveolae and activated rafts/caveolae-dependent Ras/MEK/ERK signaling pathway. In contrast, overexpression of DHCR24 prevented the overactivation of Ras/MEK/ERK signaling by increasing cellular cholesterol content, therefore decreasing tau hyperphosphorylation in C8D1A astrocytes. Herein, we firstly found that DHCR24 knockdown can lead to tau hyperphosphorylation in the astrocyte itself by activating lipid raft-dependent Ras/MEK/ERK signaling, which might contribute to the pathogenesis of AD and other degenerative tauopathies.
Collapse
Affiliation(s)
- Meiting Mai
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Xiaorou Guo
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Yue Huang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Wenbin Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Yixuan Xu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Ying Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Xiaojing Bai
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Junfeng Wu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-hang Road, Jinshan district, Shanghai, 201508 China
| |
Collapse
|
12
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [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] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
13
|
Han ZZ, Kang SG, Arce L, Westaway D. Prion-like strain effects in tauopathies. Cell Tissue Res 2022; 392:179-199. [PMID: 35460367 PMCID: PMC9034081 DOI: 10.1007/s00441-022-03620-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
Tau is a microtubule-associated protein that plays crucial roles in physiology and pathophysiology. In the realm of dementia, tau protein misfolding is associated with a wide spectrum of clinicopathologically diverse neurodegenerative diseases, collectively known as tauopathies. As proposed by the tau strain hypothesis, the intrinsic heterogeneity of tauopathies may be explained by the existence of structurally distinct tau conformers, “strains”. Tau strains can differ in their associated clinical features, neuropathological profiles, and biochemical signatures. Although prior research into infectious prion proteins offers valuable lessons for studying how a protein-only pathogen can encompass strain diversity, the underlying mechanism by which tau subtypes are generated remains poorly understood. Here we summarize recent advances in understanding different tau conformers through in vivo and in vitro experimental paradigms, and the implications of heterogeneity of pathological tau species for drug development.
Collapse
Affiliation(s)
- Zhuang Zhuang Han
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Luis Arce
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2M8, Canada. .,Department of Medicine, University of Alberta, Edmonton, AB, Canada. .,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
14
|
Shahpasand‐Kroner H, Portillo J, Lantz C, Seidler PM, Sarafian N, Loo JA, Bitan G. Three-repeat and four-repeat tau isoforms form different oligomers. Protein Sci 2022; 31:613-627. [PMID: 34902187 PMCID: PMC8862439 DOI: 10.1002/pro.4257] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 11/11/2022]
Abstract
Different tauopathies are characterized by the isoform-specific composition of the aggregates found in the brain and by structurally distinct tau strains. Although tau oligomers have been implicated as important neurotoxic species, little is known about how the primary structures of the six human tau isoforms affect tau oligomerization because the oligomers are metastable and difficult to analyze. To address this knowledge gap, here, we analyzed the initial oligomers formed by the six tau isoforms in the absence of posttranslational modifications or other manipulations using dot blots probed by an oligomer-specific antibody, native-PAGE/western blots, photo-induced cross-linking of unmodified proteins, mass-spectrometry, and ion-mobility spectroscopy. We found that under these conditions, three-repeat (3R) isoforms are more prone than four-repeat (4R) isoforms to form oligomers. We also tested whether known inhibitors of tau aggregation affect its oligomerization using three small molecules representing different classes of tau aggregation inhibitors, Methylene Blue (MB), the molecular tweezer CLR01, and the all-D peptide TLKIVW, for their ability to inhibit or modulate the oligomerization of the six tau isoforms. Unlike their reported inhibitory effect on tau fibrillation, the inhibitors had little or no effect on the initial oligomerization. Our study provides novel insight into the primary-quaternary structure relationship of human tau and suggests that 3R-tau oligomers may be an important target for future development of compounds targeting pathological tau assemblies.
Collapse
Affiliation(s)
- Hedieh Shahpasand‐Kroner
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Jennifer Portillo
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Carter Lantz
- Department of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Paul M. Seidler
- Department of Pharmacology and Pharmaceutical SciencesUniversity of Southern California School of PharmacyLos AngelesCaliforniaUSA
| | - Natalie Sarafian
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Joseph A. Loo
- Department of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCaliforniaUSA,Molecular Biology InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA,Department of Biological ChemistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Gal Bitan
- Department of NeurologyDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA,Molecular Biology InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA,Brain Research InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
15
|
Sinsky J, Pichlerova K, Hanes J. Tau Protein Interaction Partners and Their Roles in Alzheimer's Disease and Other Tauopathies. Int J Mol Sci 2021; 22:9207. [PMID: 34502116 PMCID: PMC8431036 DOI: 10.3390/ijms22179207] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Tau protein plays a critical role in the assembly, stabilization, and modulation of microtubules, which are important for the normal function of neurons and the brain. In diseased conditions, several pathological modifications of tau protein manifest. These changes lead to tau protein aggregation and the formation of paired helical filaments (PHF) and neurofibrillary tangles (NFT), which are common hallmarks of Alzheimer's disease and other tauopathies. The accumulation of PHFs and NFTs results in impairment of physiological functions, apoptosis, and neuronal loss, which is reflected as cognitive impairment, and in the late stages of the disease, leads to death. The causes of this pathological transformation of tau protein haven't been fully understood yet. In both physiological and pathological conditions, tau interacts with several proteins which maintain their proper function or can participate in their pathological modifications. Interaction partners of tau protein and associated molecular pathways can either initiate and drive the tau pathology or can act neuroprotective, by reducing pathological tau proteins or inflammation. In this review, we focus on the tau as a multifunctional protein and its known interacting partners active in regulations of different processes and the roles of these proteins in Alzheimer's disease and tauopathies.
Collapse
Affiliation(s)
| | | | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia; (J.S.); (K.P.)
| |
Collapse
|
16
|
Root J, Merino P, Nuckols A, Johnson M, Kukar T. Lysosome dysfunction as a cause of neurodegenerative diseases: Lessons from frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2021; 154:105360. [PMID: 33812000 PMCID: PMC8113138 DOI: 10.1016/j.nbd.2021.105360] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are fatal neurodegenerative disorders that are thought to exist on a clinical and pathological spectrum. FTD and ALS are linked by shared genetic causes (e.g. C9orf72 hexanucleotide repeat expansions) and neuropathology, such as inclusions of ubiquitinated, misfolded proteins (e.g. TAR DNA-binding protein 43; TDP-43) in the CNS. Furthermore, some genes that cause FTD or ALS when mutated encode proteins that localize to the lysosome or modulate endosome-lysosome function, including lysosomal fusion, cargo trafficking, lysosomal acidification, autophagy, or TFEB activity. In this review, we summarize evidence that lysosomal dysfunction, caused by genetic mutations (e.g. C9orf72, GRN, MAPT, TMEM106B) or toxic-gain of function (e.g. aggregation of TDP-43 or tau), is an important pathogenic disease mechanism in FTD and ALS. Further studies into the normal function of many of these proteins are required and will help uncover the mechanisms that cause lysosomal dysfunction in FTD and ALS. Mutations or polymorphisms in genes that encode proteins important for endosome-lysosome function also occur in other age-dependent neurodegenerative diseases, including Alzheimer's (e.g. APOE, PSEN1, APP) and Parkinson's (e.g. GBA, LRRK2, ATP13A2) disease. A more complete understanding of the common and unique features of lysosome dysfunction across the spectrum of neurodegeneration will help guide the development of therapies for these devastating diseases.
Collapse
Affiliation(s)
- Jessica Root
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Paola Merino
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Austin Nuckols
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Michelle Johnson
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia
| | - Thomas Kukar
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta 30322, Georgia; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta 30322, Georgia; Department of Neurology, Emory University, School of Medicine, Atlanta 30322, Georgia.
| |
Collapse
|
17
|
Fleeman RM, Proctor EA. Astrocytic Propagation of Tau in the Context of Alzheimer's Disease. Front Cell Neurosci 2021; 15:645233. [PMID: 33815065 PMCID: PMC8010320 DOI: 10.3389/fncel.2021.645233] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/19/2021] [Indexed: 01/14/2023] Open
Abstract
More than 6 million Americans are currently living with Alzheimer's disease (AD), and the incidence is growing rapidly with our aging population. Numerous therapeutics have failed to make it to the clinic, potentially due to a focus on presumptive pathogenic proteins instead of cell-type-specific signaling mechanisms. The tau propagation hypothesis that inter-neuronal tau transfer drives AD pathology has recently garnered attention, as accumulation of pathological tau in the brain has high clinical significance in correlating with progression of cognitive AD symptoms. However, studies on tau pathology in AD are classically neuron-centric and have greatly overlooked cell-type specific effects of tau internalization, degradation, and propagation. While the contribution of microglia to tau processing and propagation is beginning to be recognized and understood, astrocytes, glial cells in the brain important for maintaining neuronal metabolic, synaptic, trophic, and immune function which can produce, internalize, degrade, and propagate tau are understudied in their ability to affect AD progression through tau pathology. Here, we showcase evidence for whether tau uptake by astrocytes may be beneficial or detrimental to neuronal health and how astrocytes and their immunometabolic functions may be key targets for future successful AD therapies.
Collapse
Affiliation(s)
- Rebecca M Fleeman
- Department of Neurosurgery, Department of Pharmacology, College of Medicine, Pennsylvania State University (PSU), Hershey, PA, United States.,Center for Neural Engineering, Pennsylvania State University (PSU), University Park, PA, United States
| | - Elizabeth A Proctor
- Department of Neurosurgery, Department of Pharmacology, College of Medicine, Pennsylvania State University (PSU), Hershey, PA, United States.,Department of Biomedical Engineering, Department of Engineering Science and Mechanics, Center for Neural Engineering, Pennsylvania State University (PSU), University Park, PA, United States
| |
Collapse
|
18
|
Ayubcha C, Moghbel M, Borja AJ, Newberg A, Werner TJ, Alavi A, Revheim ME. Tau Imaging in Head Injury. PET Clin 2021; 16:249-260. [PMID: 33648666 DOI: 10.1016/j.cpet.2020.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Tau proteins play a significant role in a variety of degenerative neurologic conditions. Postmortem neuropathology studies of victims of repeat and severe head trauma have defined a unique spatial expression of neurologic tauopathies in these individuals, known as chronic traumatic encephalopathy. Established and newly developed radiotracers are now being applied to head injury populations with the intent of diagnosis and disease monitoring. This review assesses the role of tau in head injury, the state of tau radiotracer development, and the potential clinical value of tau-PET as derived from head injury studies.
Collapse
Affiliation(s)
- Cyrus Ayubcha
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Mateen Moghbel
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Austin J Borja
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Andrew Newberg
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA; Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Mona-Elisabeth Revheim
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Sognsvannsveien 20, Oslo 0372, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Problemveien 7, Oslo 0315, Norway.
| |
Collapse
|
19
|
Wegmann S, DeVos SL, Zeitler B, Marlen K, Bennett RE, Perez-Rando M, MacKenzie D, Yu Q, Commins C, Bannon RN, Corjuc BT, Chase A, Diez L, Nguyen HOB, Hinkley S, Zhang L, Goodwin A, Ledeboer A, Lam S, Ankoudinova I, Tran H, Scarlott N, Amora R, Surosky R, Miller JC, Robbins AB, Rebar EJ, Urnov FD, Holmes MC, Pooler AM, Riley B, Zhang HS, Hyman BT. Persistent repression of tau in the brain using engineered zinc finger protein transcription factors. SCIENCE ADVANCES 2021; 7:7/12/eabe1611. [PMID: 33741591 PMCID: PMC7978433 DOI: 10.1126/sciadv.abe1611] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/05/2021] [Indexed: 05/12/2023]
Abstract
Neuronal tau reduction confers resilience against β-amyloid and tau-related neurotoxicity in vitro and in vivo. Here, we introduce a novel translational approach to lower expression of the tau gene MAPT at the transcriptional level using gene-silencing zinc finger protein transcription factors (ZFP-TFs). Following a single administration of adeno-associated virus (AAV), either locally into the hippocampus or intravenously to enable whole-brain transduction, we selectively reduced tau messenger RNA and protein by 50 to 80% out to 11 months, the longest time point studied. Sustained tau lowering was achieved without detectable off-target effects, overt histopathological changes, or molecular alterations. Tau reduction with AAV ZFP-TFs was able to rescue neuronal damage around amyloid plaques in a mouse model of Alzheimer's disease (APP/PS1 line). The highly specific, durable, and controlled knockdown of endogenous tau makes AAV-delivered ZFP-TFs a promising approach for the treatment of tau-related human brain diseases.
Collapse
Affiliation(s)
- Susanne Wegmann
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA.
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Sarah L DeVos
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | | | | | - Rachel E Bennett
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Marta Perez-Rando
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Danny MacKenzie
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Qi Yu
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | - Caitlin Commins
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Riley N Bannon
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Bianca T Corjuc
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Alison Chase
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Lisa Diez
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | | | | | - Lei Zhang
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | | | - Stephen Lam
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | - Hung Tran
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | | | | | | | - Ashley B Robbins
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA
| | | | | | | | - Amy M Pooler
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | - Brigit Riley
- Sangamo Therapeutics Inc., Richmond, CA 94804, USA
| | | | - Bradley T Hyman
- Massachusetts General Hospital, Massachusetts Institute of Neurodegenerative Disease, Charlestown, MA 02129, USA.
| |
Collapse
|
20
|
Maxwell SP, Cash MK, Rockwood K, Fisk JD, Darvesh S. Clinical and neuropathological variability in the rare IVS10 + 14 tau mutation. Neurobiol Aging 2021; 101:298.e1-298.e10. [PMID: 33612310 DOI: 10.1016/j.neurobiolaging.2021.01.004] [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: 09/28/2020] [Revised: 11/24/2020] [Accepted: 01/09/2021] [Indexed: 10/22/2022]
Abstract
Mutations in the microtubule-associated protein tau gene are known to cause progressive neurodegenerative disorders with variable clinical and neuropathological phenotypes, including the intronic 10 + 14 (IVS10 + 14) splice site mutation. Three families have been reported with the IVS10 + 14 microtubule-associated protein tau mutation. Here, we describe the clinical and neuropathological data from an additional family. Neuropathological data were available for 2 of the 3 cases, III-4, and III-5. While III-5 had widespread tau deposition and atrophy, III-4 exhibited more mild neuropathological changes except for the substantia nigra. The previously reported families that express the IVS10 + 14 mutation exhibited significant interfamilial heterogeneity, with symptoms including amyotrophy, dementia, disinhibition, parkinsonism, and breathing problems. In addition to expressing many of these symptoms, members of this fourth family experienced profound sensory abnormalities and sleep disturbance. Although there were probable clinicopathological correlates for the symptoms expressed by the earlier families and III-5 from our cohort, pathology in III-4 did not appear sufficient to explain symptom severity. This indicates the need to explore alternate mechanisms of tau-induced brain dysfunction.
Collapse
Affiliation(s)
- Selena P Maxwell
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Meghan K Cash
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kenneth Rockwood
- Department of Medicine (Division of Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medicine (Division of Neurology), Dalhousie University, Halifax, Nova Scotia, Canada
| | - John D Fisk
- Department of Medicine (Division of Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sultan Darvesh
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medicine (Division of Geriatric Medicine), Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medicine (Division of Neurology), Dalhousie University, Halifax, Nova Scotia, Canada; Department of Chemistry and Physics, Mount Saint Vincent University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
21
|
Kang SG, Eskandari-Sedighi G, Hromadkova L, Safar JG, Westaway D. Cellular Biology of Tau Diversity and Pathogenic Conformers. Front Neurol 2020; 11:590199. [PMID: 33304310 PMCID: PMC7693435 DOI: 10.3389/fneur.2020.590199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Tau accumulation is a prominent feature in a variety of neurodegenerative disorders and remarkable effort has been expended working out the biochemistry and cell biology of this cytoplasmic protein. Tau's wayward properties may derive from germline mutations in the case of frontotemporal lobar degeneration (FTLD-MAPT) but may also be prompted by less understood cues—perhaps environmental or from molecular damage as a consequence of chronological aging—in the case of idiopathic tauopathies. Tau properties are undoubtedly affected by its covalent structure and in this respect tau protein is not only subject to changes in length produced by alternative splicing and endoproteolysis, but different types of posttranslational modifications that affect different amino acid residues. Another layer of complexity concerns alternate conformations—“conformers”—of the same covalent structures; in vivo conformers can encompass soluble oligomeric species, ramified fibrillar structures evident by light and electron microscopy and other forms of the protein that have undergone liquid-liquid phase separation to make demixed liquid droplets. Biological concepts based upon conformers have been charted previously for templated replication mechanisms for prion proteins built of the PrP polypeptide; these are now providing useful explanations to feature tau pathobiology, including how this protein accumulates within cells and how it can exhibit predictable patterns of spread across different neuroanatomical regions of an affected brain. In sum, the documented, intrinsic heterogeneity of tau forms and conformers now begins to speak to a fundamental basis for diversity in clinical presentation of tauopathy sub-types. In terms of interventions, emphasis upon subclinical events may be worthwhile, noting that irrevocable cell loss and ramified protein assemblies feature at end-stage tauopathy, whereas earlier events may offer better opportunities for diverting pathogenic processes. Nonetheless, the complexity of tau sub-types, which may be present even within intermediate disease stages, likely mitigates against one-size-fits-all therapeutic strategies and may require a suite of interventions. We consider the extent to which animal models of tauopathy can be reasonably enrolled in the campaign to produce such interventions and to slow the otherwise inexorable march of disease progression.
Collapse
Affiliation(s)
- Sang-Gyun Kang
- Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | | | - Lenka Hromadkova
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Jiri G Safar
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - David Westaway
- Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
22
|
Richetin K, Steullet P, Pachoud M, Perbet R, Parietti E, Maheswaran M, Eddarkaoui S, Bégard S, Pythoud C, Rey M, Caillierez R, Q Do K, Halliez S, Bezzi P, Buée L, Leuba G, Colin M, Toni N, Déglon N. Tau accumulation in astrocytes of the dentate gyrus induces neuronal dysfunction and memory deficits in Alzheimer's disease. Nat Neurosci 2020; 23:1567-1579. [PMID: 33169029 DOI: 10.1038/s41593-020-00728-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/24/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of the tau protein in neurons, neurodegeneration and memory loss. However, the role of non-neuronal cells in this chain of events remains unclear. In the present study, we found accumulation of tau in hilar astrocytes of the dentate gyrus of individuals with AD. In mice, the overexpression of 3R tau specifically in hilar astrocytes of the dentate gyrus altered mitochondrial dynamics and function. In turn, these changes led to a reduction of adult neurogenesis, parvalbumin-expressing neurons, inhibitory synapses and hilar gamma oscillations, which were accompanied by impaired spatial memory performances. Together, these results indicate that the loss of tau homeostasis in hilar astrocytes of the dentate gyrus is sufficient to induce AD-like symptoms, through the impairment of the neuronal network. These results are important for our understanding of disease mechanisms and underline the crucial role of astrocytes in hippocampal function.
Collapse
Affiliation(s)
- Kevin Richetin
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland. .,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland. .,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Pascal Steullet
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Mathieu Pachoud
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Romain Perbet
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Enea Parietti
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Mathischan Maheswaran
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Séverine Bégard
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Catherine Pythoud
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Maria Rey
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Raphaëlle Caillierez
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Kim Q Do
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Sophie Halliez
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Geneviève Leuba
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Morvane Colin
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Nicolas Toni
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Nicole Déglon
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
23
|
Merezhko M, Uronen RL, Huttunen HJ. The Cell Biology of Tau Secretion. Front Mol Neurosci 2020; 13:569818. [PMID: 33071756 PMCID: PMC7539664 DOI: 10.3389/fnmol.2020.569818] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022] Open
Abstract
The progressive accumulation and spread of misfolded tau protein in the nervous system is the hallmark of tauopathies, progressive neurodegenerative diseases with only symptomatic treatments available. A growing body of evidence suggests that spreading of tau pathology can occur via cell-to-cell transfer involving secretion and internalization of pathological forms of tau protein followed by templated misfolding of normal tau in recipient cells. Several studies have addressed the cell biological mechanisms of tau secretion. It now appears that instead of a single mechanism, cells can secrete tau via three coexisting pathways: (1) translocation through the plasma membrane; (2) membranous organelles-based secretion; and (3) ectosomal shedding. The relative importance of these pathways in the secretion of normal and pathological tau is still elusive, though. Moreover, glial cells contribute to tau propagation, and the involvement of different cell types, as well as different secretion pathways, complicates the understanding of prion-like propagation of tauopathy. One of the important regulators of tau secretion in neuronal activity, but its mechanistic connection to tau secretion remains unclear and may involve all three secretion pathways of tau. This review article summarizes recent advancements in the field of tau secretion with an emphasis on cell biological aspects of the secretion process and discusses the role of neuronal activity and glial cells in the spread of pathological forms of tau.
Collapse
Affiliation(s)
- Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Villa González M, Vallés-Saiz L, Hernández IH, Avila J, Hernández F, Pérez-Alvarez MJ. Focal cerebral ischemia induces changes in oligodendrocytic tau isoforms in the damaged area. Glia 2020; 68:2471-2485. [PMID: 32515854 DOI: 10.1002/glia.23865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/31/2022]
Abstract
Ischemic stroke is a major cause of death and the first leading cause of long-term disability worldwide. The only therapeutic strategy available to date is reperfusion and not all the patients are suitable for this treatment. Blood flow blockage or reduction leads to considerable brain damage, affecting both gray and white matter. The detrimental effects of ischemia have been studied extensively in the former but not in the latter. Previous reports indicate that preservation of white matter integrity reduces deleterious effect of ischemia on the brain. Oligodendrocytes are sensitive to ischemic damage, however, some reports demonstrate that oligodendrogenesis occurs after ischemia. These glial cells have a complex cytoskeletal network, including tau, that plays a key role to proper myelination. 4R-Tau/3R-Tau, which differ in the presence/absence of Exon 10, are found in oligodendrocytes; but the precise role of each isoform is not understood. Using permanent middle cerebral artery occlusion model and immunofluorescence, we demonstrate that cerebral ischemia induces an increase in 3R-Tau versus 4R-Tau in oligodendrocytes in the damaged area. In addition, cellular distribution of Tau undergoes a change after ischemia, with some oligodendrocytic processes showing positive staining for 3R-Tau. This occurs simultaneously with the amelioration of neurological damage in ischemic rats. We propose that ischemia triggers an endogenous mechanism involving 3R-Tau, that induces colonization of the ischemic damaged area by oligodendrocytes in an attempt to myelinate-injured axons. Understanding the molecular mechanism of this phenomenon could pave the way for the design of therapeutic strategies that exploit glial cells for the treatment of ischemia.
Collapse
Affiliation(s)
- Mario Villa González
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain
| | - Laura Vallés-Saiz
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain
| | - Ivó H Hernández
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús Avila
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Félix Hernández
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María José Pérez-Alvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
25
|
Ashrafizadeh M, Zarrabi A, Najafi M, Samarghandian S, Mohammadinejad R, Ahn KS. Resveratrol targeting tau proteins, amyloid-beta aggregations, and their adverse effects: An updated review. Phytother Res 2020; 34:2867-2888. [PMID: 32491273 DOI: 10.1002/ptr.6732] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/18/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
Resveratrol (Res) is a non-flavonoid compound with pharmacological actions such as antioxidant, antiinflammatory, hepatoprotective, antidiabetes, and antitumor. This plant-derived chemical has a long history usage in treatment of diseases. The excellent therapeutic impacts of Res and its capability in penetration into blood-brain barrier have made it an appropriate candidate in the treatment of neurological disorders (NDs). Tau protein aggregations and amyloid-beta (Aβ) deposits are responsible for the induction of NDs. A variety of studies have elucidated the role of these aggregations in NDs and the underlying molecular pathways in their development. In the present review, based on the recently published articles, we describe that how Res administration could inhibit amyloidogenic pathway and stimulate processes such as autophagy to degrade Aβ aggregations. Besides, we demonstrate that Res supplementation is beneficial in dephosphorylation of tau proteins and suppressing their aggregations. Then, we discuss molecular pathways and relate them to the treatment of NDs.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Turkey
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Brunello CA, Merezhko M, Uronen RL, Huttunen HJ. Mechanisms of secretion and spreading of pathological tau protein. Cell Mol Life Sci 2020; 77:1721-1744. [PMID: 31667556 PMCID: PMC7190606 DOI: 10.1007/s00018-019-03349-1] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
Accumulation of misfolded and aggregated forms of tau protein in the brain is a neuropathological hallmark of tauopathies, such as Alzheimer's disease and frontotemporal lobar degeneration. Tau aggregates have the ability to transfer from one cell to another and to induce templated misfolding and aggregation of healthy tau molecules in previously healthy cells, thereby propagating tau pathology across different brain areas in a prion-like manner. The molecular mechanisms involved in cell-to-cell transfer of tau aggregates are diverse, not mutually exclusive and only partially understood. Intracellular accumulation of misfolded tau induces several mechanisms that aim to reduce the cellular burden of aggregated proteins and also promote secretion of tau aggregates. However, tau may also be released from cells physiologically unrelated to protein aggregation. Tau secretion involves multiple vesicular and non-vesicle-mediated pathways, including secretion directly through the plasma membrane. Consequently, extracellular tau can be found in various forms, both as a free protein and in vesicles, such as exosomes and ectosomes. Once in the extracellular space, tau aggregates can be internalized by neighboring cells, both neurons and glial cells, via endocytic, pinocytic and phagocytic mechanisms. Importantly, accumulating evidence suggests that prion-like propagation of misfolding protein pathology could provide a general mechanism for disease progression in tauopathies and other related neurodegenerative diseases. Here, we review the recent literature on cellular mechanisms involved in cell-to-cell transfer of tau, with a particular focus in tau secretion.
Collapse
Affiliation(s)
- Cecilia A Brunello
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Riikka-Liisa Uronen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland.
| |
Collapse
|
27
|
Sung K, Jimenez-Sanchez M. Autophagy in Astrocytes and its Implications in Neurodegeneration. J Mol Biol 2020; 432:2605-2621. [PMID: 31931011 DOI: 10.1016/j.jmb.2019.12.041] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022]
Abstract
Autophagy is a major degradation pathway where double-membrane vesicles called autophagosomes deliver cytoplasmic content to the lysosome. Increasing evidence suggests that autophagy dysfunction contributes to the pathogenesis of neurodegenerative diseases. In addition, misfolded proteins that accumulate in these diseases and constitute a common pathological hallmark are substrates for autophagic degradation. Astrocytes, a major type of glial cells, are emerging as a critical component in most neurodegenerative diseases. This review will summarize the recent efforts to investigate the role that autophagy plays in astrocytes in the context of neurodegenerative diseases. While the field has mostly focused on the implications of autophagy in neurons, autophagy may also be involved in the clearance of disease-related proteins in astrocytes as well as in maintaining astrocyte function, which could impact the cell autonomous and non-cell autonomous contribution of astrocytes to neurodegeneration.
Collapse
Affiliation(s)
- Katherine Sung
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RX, UK
| | - Maria Jimenez-Sanchez
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RX, UK.
| |
Collapse
|
28
|
Transmission of tauopathy strains is independent of their isoform composition. Nat Commun 2020; 11:7. [PMID: 31911587 PMCID: PMC6946697 DOI: 10.1038/s41467-019-13787-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/15/2019] [Indexed: 01/12/2023] Open
Abstract
The deposition of pathological tau is a common feature in several neurodegenerative tauopathies. Although equal ratios of tau isoforms with 3 (3R) and 4 (4R) microtubule-binding repeats are expressed in the adult human brain, the pathological tau from different tauopathies have distinct isoform compositions and cell type specificities. The underlying mechanisms of tauopathies are unknown, partially due to the lack of proper models. Here, we generate a new transgenic mouse line expressing equal ratios of 3R and 4R human tau isoforms (6hTau mice). Intracerebral injections of distinct human tauopathy brain-derived tau strains into 6hTau mice recapitulate the deposition of pathological tau with distinct tau isoform compositions and cell type specificities as in human tauopathies. Moreover, through in vivo propagation of these tau strains among different mouse lines, we demonstrate that the transmission of distinct tau strains is independent of strain isoform compositions, but instead intrinsic to unique pathological conformations. Although normal human brains express 6 tau isoforms in equal ratio with 3 or 4 microtubule-binding repeat domains (3R and 4R), tau inclusions from different human tauopathy brains, now considered as different strains, have distinct isoform compositions and strain properties and the relationship between these two parts is unclear. Here the authors generate a new transgenic mouse line expressing 6 human tau isoforms with equal 3R and 4R ratios, recapitulate distinct human tau strains in mouse brains with similar isoform compositions and cell type specificities, and further show the strain transmission pattern is independent of its isoform composition.
Collapse
|
29
|
Xu H, Dzhashiashvili Y, Shah A, Kunjamma RB, Weng YL, Elbaz B, Fei Q, Jones JS, Li YI, Zhuang X, Ming GL, He C, Popko B. m 6A mRNA Methylation Is Essential for Oligodendrocyte Maturation and CNS Myelination. Neuron 2019; 105:293-309.e5. [PMID: 31901304 DOI: 10.1016/j.neuron.2019.12.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 10/15/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022]
Abstract
The molecular mechanisms that govern the maturation of oligodendrocyte lineage cells remain unclear. Emerging studies have shown that N6-methyladenosine (m6A), the most common internal RNA modification of mammalian mRNA, plays a critical role in various developmental processes. Here, we demonstrate that oligodendrocyte lineage progression is accompanied by dynamic changes in m6A modification on numerous transcripts. In vivo conditional inactivation of an essential m6A writer component, METTL14, results in decreased oligodendrocyte numbers and CNS hypomyelination, although oligodendrocyte precursor cell (OPC) numbers are normal. In vitro Mettl14 ablation disrupts postmitotic oligodendrocyte maturation and has distinct effects on OPC and oligodendrocyte transcriptomes. Moreover, the loss of Mettl14 in oligodendrocyte lineage cells causes aberrant splicing of myriad RNA transcripts, including those that encode the essential paranodal component neurofascin 155 (NF155). Together, our findings indicate that dynamic RNA methylation plays an important regulatory role in oligodendrocyte development and CNS myelination.
Collapse
Affiliation(s)
- Huan Xu
- Center for Peripheral Neuropathy and Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Yulia Dzhashiashvili
- Center for Peripheral Neuropathy and Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Ankeeta Shah
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL 60637, USA; Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Rejani B Kunjamma
- Center for Peripheral Neuropathy and Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Yi-Lan Weng
- Department of Neuroscience and Mahoney Institute for Neurosciences, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benayahu Elbaz
- Center for Peripheral Neuropathy and Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Qili Fei
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Joshua S Jones
- Center for Peripheral Neuropathy and Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Yang I Li
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Xiaoxi Zhuang
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chuan He
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Brian Popko
- Center for Peripheral Neuropathy and Department of Neurology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
30
|
Spencer B, Brüschweiler S, Sealey-Cardona M, Rockenstein E, Adame A, Florio J, Mante M, Trinh I, Rissman RA, Konrat R, Masliah E. Selective targeting of 3 repeat Tau with brain penetrating single chain antibodies for the treatment of neurodegenerative disorders. Acta Neuropathol 2018; 136:69-87. [PMID: 29934874 PMCID: PMC6112111 DOI: 10.1007/s00401-018-1869-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly affecting more than 5 million people in the U.S. AD is characterized by the accumulation of β-amyloid (Aβ) and Tau in the brain, and is manifested by severe impairments in memory and cognition. Therefore, removing tau pathology has become one of the main therapeutic goals for the treatment of AD. Tau (tubulin-associated unit) is a major neuronal cytoskeletal protein found in the CNS encoded by the gene MAPT. Alternative splicing generates two major isoforms of tau containing either 3 or 4 repeat (R) segments. These 3R or 4RTau species are differentially expressed in neurodegenerative diseases. Previous studies have been focused on reducing Tau accumulation with antibodies against total Tau, 4RTau or phosphorylated isoforms. Here, we developed a brain penetrating, single chain antibody that specifically recognizes a pathogenic 3RTau. This single chain antibody was modified by the addition of a fragment of the apoB protein to facilitate trafficking into the brain, once in the CNS these antibody fragments reduced the accumulation of 3RTau and related deficits in a transgenic mouse model of tauopathy. NMR studies showed that the single chain antibody recognized an epitope at aa 40-62 of 3RTau. This single chain antibody reduced 3RTau transmission and facilitated the clearance of Tau via the endosomal-lysosomal pathway. Together, these results suggest that targeting 3RTau with highly specific, brain penetrating, single chain antibodies might be of potential value for the treatment of tauopathies such as Pick's Disease.
Collapse
Affiliation(s)
- Brian Spencer
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Sven Brüschweiler
- Department of Computational and Structural Biology, University of Vienna, Vienna, Austria
| | - Marco Sealey-Cardona
- Department of Computational and Structural Biology, University of Vienna, Vienna, Austria
| | - Edward Rockenstein
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Jazmin Florio
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Michael Mante
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Ivy Trinh
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, USA
| | - Robert Konrat
- Department of Computational and Structural Biology, University of Vienna, Vienna, Austria
| | - Eliezer Masliah
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA.
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA.
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, 7201 Wisconsin Ave, Bethesda, MD, 20892, USA.
| |
Collapse
|
31
|
Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol 2017; 133:665-704. [PMID: 28386764 PMCID: PMC5390006 DOI: 10.1007/s00401-017-1707-9] [Citation(s) in RCA: 587] [Impact Index Per Article: 83.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/26/2017] [Accepted: 03/26/2017] [Indexed: 01/18/2023]
Abstract
Tau is well established as a microtubule-associated protein in neurons. However, under pathological conditions, aberrant assembly of tau into insoluble aggregates is accompanied by synaptic dysfunction and neural cell death in a range of neurodegenerative disorders, collectively referred to as tauopathies. Recent advances in our understanding of the multiple functions and different locations of tau inside and outside neurons have revealed novel insights into its importance in a diverse range of molecular pathways including cell signalling, synaptic plasticity, and regulation of genomic stability. The present review describes the physiological and pathophysiological properties of tau and how these relate to its distribution and functions in neurons. We highlight the post-translational modifications of tau, which are pivotal in defining and modulating tau localisation and its roles in health and disease. We include discussion of other pathologically relevant changes in tau, including mutation and aggregation, and how these aspects impinge on the propensity of tau to propagate, and potentially drive neuronal loss, in diseased brain. Finally, we describe the cascade of pathological events that may be driven by tau dysfunction, including impaired axonal transport, alterations in synapse and mitochondrial function, activation of the unfolded protein response and defective protein degradation. It is important to fully understand the range of neuronal functions attributed to tau, since this will provide vital information on its involvement in the development and pathogenesis of disease. Such knowledge will enable determination of which critical molecular pathways should be targeted by potential therapeutic agents developed for the treatment of tauopathies.
Collapse
Affiliation(s)
- Tong Guo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK
| | - Diane P Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK.
| |
Collapse
|
32
|
Arendt T, Stieler JT, Holzer M. Tau and tauopathies. Brain Res Bull 2016; 126:238-292. [DOI: 10.1016/j.brainresbull.2016.08.018] [Citation(s) in RCA: 333] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022]
|
33
|
Lopes S, Lopes A, Pinto V, Guimarães MR, Sardinha VM, Duarte‐Silva S, Pinheiro S, Pizarro J, Oliveira JF, Sousa N, Leite‐Almeida H, Sotiropoulos I. Absence of Tau triggers age-dependent sciatic nerve morphofunctional deficits and motor impairment. Aging Cell 2016; 15:208-16. [PMID: 26748966 PMCID: PMC4783352 DOI: 10.1111/acel.12391] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2015] [Indexed: 11/29/2022] Open
Abstract
Dementia is the cardinal feature of Alzheimer's disease (AD), yet the clinical symptoms of this disorder also include a marked loss of motor function. Tau abnormal hyperphosphorylation and malfunction are well‐established key events in AD neuropathology but the impact of the loss of normal Tau function in neuronal degeneration and subsequent behavioral deficits is still debated. While Tau reduction has been increasingly suggested as therapeutic strategy against neurodegeneration, particularly in AD, there is controversial evidence about whether loss of Tau progressively impacts on motor function arguing about damage of CNS motor components. Using a variety of motor‐related tests, we herein provide evidence of an age‐dependent motor impairment in Tau−/− animals that is accompanied by ultrastructural and functional impairments of the efferent fibers that convey motor‐related information. Specifically, we show that the sciatic nerve of old (17–22‐months) Tau−/− mice displays increased degenerating myelinated fibers and diminished conduction properties, as compared to age‐matched wild‐type (Tau+/+) littermates and younger (4–6 months) Tau−/− and Tau+/+ mice. In addition, the sciatic nerves of Tau−/− mice exhibit a progressive hypomyelination (assessed by g‐ratio) specifically affecting large‐diameter, motor‐related axons in old animals. These findings suggest that loss of Tau protein may progressively impact on peripheral motor system.
Collapse
Affiliation(s)
- Sofia Lopes
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - André Lopes
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Vítor Pinto
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Marco R. Guimarães
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Vanessa Morais Sardinha
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Sara Duarte‐Silva
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Sara Pinheiro
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - João Pizarro
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - João Filipe Oliveira
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Hugo Leite‐Almeida
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS) School of Health Sciences University of Minho Campus Gualtar 4710‐057 Braga Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| |
Collapse
|
34
|
Kahlson MA, Colodner KJ. Glial Tau Pathology in Tauopathies: Functional Consequences. J Exp Neurosci 2016; 9:43-50. [PMID: 26884683 PMCID: PMC4750898 DOI: 10.4137/jen.s25515] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 12/22/2022] Open
Abstract
Tauopathies are a class of neurodegenerative diseases characterized by the presence of hyperphosphorylated and aggregated tau pathology in neuronal and glial cells. Though the ratio of neuronal and glial tau aggregates varies across diseases, glial tau aggregates can populate the same degenerating brain regions as neuronal tau aggregates. While much is known about the deleterious consequences of tau pathology in neurons, the relative contribution of glial tau pathology to these diseases is less clear. Recent studies using a number of model systems implicate glial tau pathology in contributing to tauopathy pathogenesis. This review aims to highlight the functional consequences of tau overexpression in glial cells and explore the potential contribution of glial tau pathology in the pathogenesis of neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Martha A Kahlson
- Department of Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, USA
| | - Kenneth J Colodner
- Department of Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, USA
| |
Collapse
|
35
|
ADNP/ADNP2 expression in oligodendrocytes: implication for myelin-related neurodevelopment. J Mol Neurosci 2015; 57:304-13. [DOI: 10.1007/s12031-015-0640-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
36
|
Seiberlich V, Bauer NG, Schwarz L, Ffrench-Constant C, Goldbaum O, Richter-Landsberg C. Downregulation of the microtubule associated protein Tau impairs process outgrowth and myelin basic protein mRNA transport in oligodendrocytes. Glia 2015; 63:1621-35. [DOI: 10.1002/glia.22832] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 03/17/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Veronika Seiberlich
- Department for Neuroscience; Molecular Neurobiology; University of Oldenburg; Oldenburg Germany
| | - Nina G. Bauer
- Department for Neuroscience; Molecular Neurobiology; University of Oldenburg; Oldenburg Germany
- MRC Centre for Regenerative Medicine; The University of Edinburgh, Edinburgh BioQuarter; Edinburgh United Kingdom
| | - Lisa Schwarz
- Department for Neuroscience; Molecular Neurobiology; University of Oldenburg; Oldenburg Germany
| | - Charles Ffrench-Constant
- MRC Centre for Regenerative Medicine; The University of Edinburgh, Edinburgh BioQuarter; Edinburgh United Kingdom
| | - Olaf Goldbaum
- Department for Neuroscience; Molecular Neurobiology; University of Oldenburg; Oldenburg Germany
| | | |
Collapse
|
37
|
Abstract
Common cellular and molecular mechanisms including protein aggregation and inclusion body formation are involved in many neurodegenerative diseases. α-Synuclein is a major component of Lewy bodies in Parkinson's disease (PD) as well as in glial cytoplasmic inclusions in multiple system atrophy (MSA). Tau is a principal component of neurofibrillary and glial tangles in tauopathies. Recently, TDP-43 was identified as a component of ubiquitinated inclusions in amyotrophic lateral sclerosis and frontotemporal lobar degeneration. PD is traditionally considered a movement disorder with hallmark lesions in the brainstem pigmented nuclei. However, pathological changes occur in widespread regions of the central and peripheral nervous systems in this disease. Furthermore, primary glial involvement ("gliodegeneration") can be observed in PD and MSA as well as in tauopathy. The present article reviews abnormal protein accumulation and inclusion body formation inside and outside the central nervous system.
Collapse
Affiliation(s)
- Koichi Wakabayashi
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Japan
| |
Collapse
|
38
|
Noack M, Leyk J, Richter-Landsberg C. HDAC6 inhibition results in tau acetylation and modulates tau phosphorylation and degradation in oligodendrocytes. Glia 2014; 62:535-47. [PMID: 24464872 DOI: 10.1002/glia.22624] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/11/2013] [Accepted: 12/16/2013] [Indexed: 01/06/2023]
Abstract
Histone deacetylase 6 (HDAC6) is a unique member of the HDAC family. It is localized within the cytoplasm and has unique substrate specificities for nonhistone proteins, such as α-tubulin. Furthermore, it plays a major role in protein aggregate formation and recently was demonstrated to interact with the microtubule associated protein tau and tau was identified as a possible substrate for HDAC6 in neurons. This study was undertaken to investigate whether HDAC6 is present in oligodendrocytes and whether it is involved in tubulin and tau acetylation in these cells. We show for the first time that HDAC6 is expressed in cultured rat brain oligodendrocytes. Its inhibition by the specific HDAC6 inhibitor tubastatin A (TST) leads to morphological alterations, microtubule bundling, and tubulin acetylation, and changes in tau-isoform expression and phosphorylation. Furthermore, the microtubule binding activity of tau was reduced. Using the oligodendroglial cell lines OLN-t40 and OLN-t44, which were genetically engineered to express either the longest human tau isoform with four microtubule binding repeats (4R-tau), or the shortest tau isoform with three repeats (3R-tau), respectively, we demonstrate that tau is acetylated by HDAC6 within the 4R-binding domain. Tau acetylation reduced its turnover rate and acetylated tau was degraded slower in these cells. TST and shRNA-mediated knockdown of HDAC6 in oligodendroglia cells caused an increase in pathological hyperphosphorylated tau detectable with the 12E8 antibody. Hence HDAC6 and dysregulation of the deacetylation and acetylation process in oligodendrocytes may contribute to diseases with oligodendroglial pathology.
Collapse
Affiliation(s)
- Monika Noack
- Department of Biology, Molecular Neurobiology, University of Oldenburg, D-26111, Oldenburg, Germany
| | | | | |
Collapse
|
39
|
Fressinaud C, Eyer J. Neurofilament-tubulin binding site peptide NFL-TBS.40-63 increases the differentiation of oligodendrocytes in vitro and partially prevents them from lysophosphatidyl choline toxiciy. J Neurosci Res 2013; 92:243-53. [PMID: 24327347 DOI: 10.1002/jnr.23308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 08/30/2013] [Accepted: 09/06/2013] [Indexed: 01/05/2023]
Abstract
During multiple sclerosis (MS), the main axon cystoskeleton proteins, neurofilaments (NF), are altered, and their release into the cerebrospinal fluid correlates with disease severity. The role of NF in the extraaxonal location is unknown. Therefore, we tested whether synthetic peptides corresponding to the tubulin-binding site (TBS) sequence identified on light NF chain (NFL-TBS.40-63) and keratin (KER-TBS.1-24), which could be released during MS, modulate remyelination in vitro. Biotinylated NFL-TBS.40-63, NFL-Scramble2, and KER-TBS.1-54 (1-100 μM, 24 hr) were added to rat oligodendrocyte (OL) and astrocyte (AS) cultures, grown in chemically defined medium. Proliferation and differentiation were characterized by using specific antibodies (A2B5, CNP, MBP, GFAP) and compared with untreated cultures. Lysophosphatidyl choline (LPC; 2 × 10(-5) M) was used to induce OL death and to test the effects of TBS peptides under these conditions. NFL-TBS.40-63 significantly increased OL differentiation and maturation, with more CNP(+) and MBP(+) cells characterized by numerous ramified processes, along with myelin balls. When OL were challenged with LPC, concomitant treatment with NFL-TBS.40-63 rescued more than 50% of OL compared with cultures treated with LPC only. Proliferation of OL progenitors was not affected, nor were AS proliferation and differentiation. NFL-TBS.40-63 peptide induces specific effects in vitro, increasing OL differentiation and maturation without altering AS fate. In addition, it partially protects OL from demyelinating injury. Thus release of NFL-TBS.40-63 caused by axonal damage in vivo could improve repair through increased OL differentiation, which is a prerequisite for remyelination.
Collapse
Affiliation(s)
- Catherine Fressinaud
- LUNAM, Neurology Department, University Hospital, Angers, France; LUNAM, Neurobiology and Transgenesis Laboratory, UPRES EA 3143, University Hospital, Angers, France
| | | |
Collapse
|
40
|
Reduced beta-catenin expression in the hippocampal CA1 region following transient cerebral ischemia in the gerbil. Neurochem Res 2013; 38:1045-54. [PMID: 23504294 DOI: 10.1007/s11064-013-1015-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 02/08/2013] [Accepted: 03/05/2013] [Indexed: 12/16/2022]
Abstract
Beta-catenin, a transcription factor, plays a critical role in cell survival and degradation after stroke. In this study, we examined changes of expression in beta-catenin in the hippocampal CA1 region of the gerbil following 5 min of transient cerebral ischemia. We observed neuronal damage using cresyl violet staining, neuronal nuclei immunohistochemistry and Fluro-Jade B immunofluorescence. Four days after ischemia-reperfusion (I-R), most of pyramidal cells in the CA1 region were damaged. In addition, early damage in dendrites was detected 1 day after I-R by immunohistochemical staining for microtubule-associated protein 2 (MAP-2), and MAP-2 immunoreactivity was hardly detected in the CA1 region 4 days after I-R. We found that beta-catenin (a synapse-enriched cell adhesion molecule) was well expressed in dendrites before I-R. Its immunoreactivity was well colocalized with MAP-2. Chronological change of beta-catenin immunoreactivity was novelty in the present study. Twelve hours after I-R, its immunoreactivity was decreased in the stratum radiatum of the CA1 region, however, its immunoreactivity was increased 1 and 2 days after I-R, and decreased sharply 4 days after I-R. However, we did not find any change in beta-catenin immunoreactivity in the CA2 and CA3 region. In brief, we suggest that early change of beta-catenin expression in the stratum pyramidale of ischemic hippocampal CA1 region is associated with early dendrite damage following transient cerebral ischemia.
Collapse
|
41
|
Harauz G, Boggs JM. Myelin management by the 18.5-kDa and 21.5-kDa classic myelin basic protein isoforms. J Neurochem 2013; 125:334-61. [PMID: 23398367 DOI: 10.1111/jnc.12195] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 12/15/2022]
Abstract
The classic myelin basic protein (MBP) splice isoforms range in nominal molecular mass from 14 to 21.5 kDa, and arise from the gene in the oligodendrocyte lineage (Golli) in maturing oligodendrocytes. The 18.5-kDa isoform that predominates in adult myelin adheres the cytosolic surfaces of oligodendrocyte membranes together, and forms a two-dimensional molecular sieve restricting protein diffusion into compact myelin. However, this protein has additional roles including cytoskeletal assembly and membrane extension, binding to SH3-domains, participation in Fyn-mediated signaling pathways, sequestration of phosphoinositides, and maintenance of calcium homeostasis. Of the diverse post-translational modifications of this isoform, phosphorylation is the most dynamic, and modulates 18.5-kDa MBP's protein-membrane and protein-protein interactions, indicative of a rich repertoire of functions. In developing and mature myelin, phosphorylation can result in microdomain or even nuclear targeting of the protein, supporting the conclusion that 18.5-kDa MBP has significant roles beyond membrane adhesion. The full-length, early-developmental 21.5-kDa splice isoform is predominantly karyophilic due to a non-traditional P-Y nuclear localization signal, with effects such as promotion of oligodendrocyte proliferation. We discuss in vitro and recent in vivo evidence for multifunctionality of these classic basic proteins of myelin, and argue for a systematic evaluation of the temporal and spatial distributions of these protein isoforms, and their modified variants, during oligodendrocyte differentiation.
Collapse
Affiliation(s)
- George Harauz
- Department of Molecular and Cellular Biology, Biophysics Interdepartmental Group and Collaborative Program in Neuroscience, University of Guelph, Guelph, Ontario, Canada.
| | | |
Collapse
|
42
|
Miller DW, Cookson MR, Dickson DW. Glial cell inclusions and the pathogenesis of neurodegenerative diseases. ACTA ACUST UNITED AC 2012; 1:13-21. [PMID: 16614753 PMCID: PMC1435946 DOI: 10.1017/s1740925x04000043] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this review, we discuss examples that show how glial-cell pathology is increasingly recognized in several neurodegenerative diseases. We also discuss the more provocative idea that some of the disorders that are currently considered to be neurodegenerative diseases might, in fact, be due to primary abnormalities in glia. Although the mechanism of glial pathology (i.e. modulating glutamate excitotoxicity) might be better established for amyotrophic lateral sclerosis (ALS), a role for neuronal-glial interactions in the pathogenesis of most neurodegenerative diseases is plausible. This burgeoning area of neuroscience will receive much attention in the future and it is expected that further understanding of basic neuronal-glial interactions will have a significant impact on the understanding of the fundamental nature of human neurodegenerative disorders.
Collapse
Affiliation(s)
- David W. Miller
- Laboratory of Neurogenetics National Institute on Aging 9000 Rockville Pike Bethesda MD 20892-1589
| | - Mark R. Cookson
- Laboratory of Neurogenetics National Institute on Aging 9000 Rockville Pike Bethesda MD 20892-1589
- Correspondence should be addressed to: Mark R. Cookson, Ph.D., Laboratory of Neurogenetics, National Institute on Aging, NIH, Building 10, Room 6C103, MSC1589, 9000 Rockville Pike, Bethesda MD 20892, USA, phone: +1 301 451, 3870 fax: +1 301 480 0315
| | - Dennis W. Dickson
- Department of Neuroscience Mayo Clinic Jacksonville 4500 San Pablo Road Jacksonville FL 32224
| |
Collapse
|
43
|
Zhong Q, Congdon EE, Nagaraja HN, Kuret J. Tau isoform composition influences rate and extent of filament formation. J Biol Chem 2012; 287:20711-9. [PMID: 22539343 DOI: 10.1074/jbc.m112.364067] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The risk of developing tauopathic neurodegenerative disease depends in part on the levels and composition of six naturally occurring Tau isoforms in human brain. These proteins, which form filamentous aggregates in disease, vary only by the presence or absence of three inserts encoded by alternatively spliced exons 2, 3, and 10 of the Tau gene (MAPT). To determine the contribution of alternatively spliced segments to Tau aggregation propensity, the aggregation kinetics of six unmodified, recombinant human Tau isoforms were examined in vitro using electron microscopy assay methods. Aggregation propensity was then compared at the level of elementary rate constants for nucleation and extension phases. We found that all three alternatively spliced segments modulated Tau aggregation but through differing kinetic mechanisms that could synergize or compete depending on sequence context. Overall, segments encoded by exons 2 and 10 promoted aggregation, whereas the segment encoded by exon 3 depressed it with its efficacy dependent on the presence or absence of a fourth microtubule binding repeat. In general, aggregation propensity correlated with genetic risk reported for multiple tauopathies, implicating aggregation as one candidate mechanism rationalizing the correlation between Tau expression patterns and disease.
Collapse
Affiliation(s)
- Qi Zhong
- Department of Molecular & Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|
44
|
Regulation of gene expression during early neuronal differentiation: evidence for patterns conserved across neuron populations and vertebrate classes. Cell Tissue Res 2012; 348:1-27. [PMID: 22437873 DOI: 10.1007/s00441-012-1367-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 02/08/2012] [Indexed: 12/19/2022]
Abstract
Analysis of transcription factor function during neurogenesis has provided a huge amount of data on the generation and specification of diverse neuron populations in the central and peripheral nervous systems of vertebrates. However, an understanding of the induction of key neuron functions including electrical information processing and synaptic transmission lags seriously behind. Whereas pan-neuronal markers such as neurofilaments, neuron-specific tubulin and RNA-binding proteins have often been included in developmental analysis, the molecular players underlying electrical activity and transmitter release have been neglected in studies addressing gene expression during neuronal induction. Here, I summarize the evidence for a distinct accumulation pattern of mRNAs for synaptic proteins, a pattern that is delayed compared with pan-neuronal gene expression during neurogenesis. The conservation of this pattern across diverse avian and mammalian neuron populations suggests a common mechanism for the regulation of various sets of neuronal genes during initial neuronal differentiation. The co-regulation of genes coding for synaptic proteins from embryonic to postnatal development indicates that the expression of the players required for synaptic transmission shares common regulatory features. For the ion channels involved in neuronal electrical activity, such as voltage-gated sodium channels, the situation is less clear because of the lack of comparative studies early during neurogenesis. Transcription factors have been characterized that regulate the expression of synaptic proteins in vitro and in vivo. They currently do not explain the co-regulation of these genes across different neuron populations. The neuron-restrictive silencing factor NRSF/REST targets a large gene set, but not all of the genes coding for pan-neuronal, synaptic and ion channel proteins. The discrepancy between NRSF/REST loss-of-function and silencer-to-activator-switch studies leaves the full functional implications of this factor open. Together with microRNAs, splicing regulators, chromatin remodellers and an increasing list of transcriptional regulators, the factor is embedded in feedback circuits with the potential to orchestrate neuronal differentiation. The precise regulation of the coordinated expression of proteins underlying key neuronal functions by these circuits during neuronal induction is a major emerging topic.
Collapse
|
45
|
Fressinaud C, Berges R, Eyer J. Axon cytoskeleton proteins specifically modulate oligodendrocyte growth and differentiation in vitro. Neurochem Int 2012; 60:78-90. [DOI: 10.1016/j.neuint.2011.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 09/26/2011] [Accepted: 10/26/2011] [Indexed: 12/19/2022]
|
46
|
Abstract
Tauopathies are a group of disorders that have in common abnormal accumulation of tau protein in the brain. Although the different tauopathies have long been considered to be separate diseases, it is now clear that progressive supranuclear palsy, corticobasal degeneration and some forms of tau-positive frontotemporal lobar degeneration share clinical, pathological and genetic features. The important overlap between these disorders suggest they may represent different phenotypes of a single disease process, the clinical result depending on the topography of pathological lesions as well as other unknown factors.
Collapse
|
47
|
Aureli M, Loberto N, Lanteri P, Chigorno V, Prinetti A, Sonnino S. Cell surface sphingolipid glycohydrolases in neuronal differentiation and aging in culture. J Neurochem 2011; 116:891-9. [DOI: 10.1111/j.1471-4159.2010.07019.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
Fukui M, Ozawa S, Nakamichi N, Nakazato R, Takarada T, Yoneda Y. Gradual downregulation of protein expression of the partner GABA(B)R2 subunit during postnatal brain development in mice defective of GABA(B)R1 subunit. J Pharmacol Sci 2010; 115:45-55. [PMID: 21160134 DOI: 10.1254/jphs.10254fp] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
We have previously shown the functional expression of GABA(B) receptors (GABA(B)R) composed of GABA(B)R1 and GABA(B)R2 subunits with ability to promote proliferation and neuronal differentiation in cultured neural progenitor cells (NPC) isolated from embryonic mouse brains. In this study, we evaluated postnatal changes in the expression profiles of different markers for progenitor, neuronal, astroglial, and microglial cells in brains of GABA(B)R1-null mice. Consistent with undifferentiated murine NPC cultured with epidermal growth factor, a significant and selective decrease was seen in mRNA expression of the proneural gene Mash1 in brains of GABA(B)R1-null mice at 1 day after birth. The expression of several NPC marker proteins was similarly decreased in brains of both wild-type and GABA(B)R1-null mice from 1 to 7 days after birth, while slight changes were induced in both mRNA and proteins for neuronal, astroglial, and microglial markers between wild-type and GABA(B)R1-null mouse brains within this developmental stage. In particular discrete brain structures of adult GABA(B)R1-null mice at 56 days after birth, a significant decrease was seen in neuronal marker protein levels along with a significant increase in both astroglial and microglial marker protein expression. Although no significant difference was found in mRNA expression of the partner GABA(B)R2 subunit between wild-type and GABA(B)R1-null mouse brains, GABA(B)R2 subunit protein levels were gradually declined during postnatal development within 56 days after birth in GABA(B)R1-null mouse brains. These results suggest that GABA(B)R2 protein levels are closely correlated with the partner subunit GABA(B)R1 protein levels in mouse brains during postnatal development in vivo.
Collapse
Affiliation(s)
- Masaki Fukui
- Kanazawa University Graduate School of Natural Science and Technology, Kanazawa, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Bauer NG, Richter-Landsberg C, Ffrench-Constant C. Role of the oligodendroglial cytoskeleton in differentiation and myelination. Glia 2010; 57:1691-705. [PMID: 19455583 DOI: 10.1002/glia.20885] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oligodendrocytes, the myelin-forming cells of the central nervous system, are in culture characterized by an elaborate process network, terminating in flat membranous sheets that are rich in myelin-specific proteins and lipids, and spirally wrap axons forming a compact insulating layer in vivo. By analogy with other cell types, maintenance and stability of these processes, as well as the formation of the myelin sheath, likely rely on a pronounced cytoskeleton consisting of microtubules and microfilaments. While the specialized process of wrapping and compaction forming the myelin sheath is not well understood, considerably more is known about how cytoskeletal organization is mediated by extracellular and intracellular signals and other interaction partners during oligodendrocyte differentiation and myelination. Here, we review the current state of knowledge on the role of the oligodendrocyte cytoskeleton in differentiation with an emphasis on signal transduction mechanisms and will attempt to draw out implications for its significance in myelination.
Collapse
Affiliation(s)
- Nina G Bauer
- MRC Centre for Regenerative Medicine, Centre for Multiple Sclerosis Research, The University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom.
| | | | | |
Collapse
|
50
|
Rao RC, Boyd J, Padmanabhan R, Chenoweth JG, McKay RD. Efficient serum-free derivation of oligodendrocyte precursors from neural stem cell-enriched cultures. Stem Cells 2009; 27:116-25. [PMID: 18403757 DOI: 10.1634/stemcells.2007-0205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Oligodendrocytes derived in the laboratory from stem cells have been proposed as a treatment for acute and chronic injury to the central nervous system. Platelet-derived growth factor (PDGF) receptor alpha (PDGFRalpha) signaling is known to regulate oligodendrocyte precursor cell numbers both during development and adulthood. Here, we analyze the effects of PDGFRalpha signaling on central nervous system (CNS) stem cell-enriched cultures. We find that AC133 selection for CNS progenitors acutely isolated from the fetal cortex enriches for PDGF-AA-responsive cells. PDGF-AA treatment of fibroblast growth factor 2-expanded CNS stem cell-enriched cultures increases nestin(+) cell number, viability, proliferation, and glycolytic rate. We show that a brief exposure to PDGF-AA rapidly and efficiently permits the derivation of O4(+) oligodendrocyte-lineage cells from CNS stem cell-enriched cultures. The derivation of oligodendrocyte-lineage cells demonstrated here may support the effective use of stem cells in understanding fate choice mechanisms and the development of new therapies targeting this cell type.
Collapse
Affiliation(s)
- Rajesh C Rao
- Laboratory of Molecular Biology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|