1
|
Sahoo TA, Chand J, Kandy AT, Antony S, Subramanian G. Unravelling the Proteinopathic Engagement of α-Synuclein, Tau, and Amyloid Beta in Parkinson's Disease: Mitochondrial Collapse as a Pivotal Driver of Neurodegeneration. Neurochem Res 2025; 50:145. [PMID: 40240583 DOI: 10.1007/s11064-025-04399-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
Parkinson's disease is a complex neurological ailment manifested by dopaminergic neurodegeneration in the substantia nigra of the brain. This study investigates the molecular tripartite interaction between Lewy bodies, amyloid beta, and tau protein in the pathogenesis of Parkinson's disease. Lewy bodies which have been found as the important pathological hallmark in the degenerative neurons of Parkinson's patients, are mainly composed of α-synuclein. The accumulation of α-synuclein has been directly and indirectly linked to the severity and degree of progression of the disease. In addition, approximately 50% of Parkinson's disease cases are also described by hyperphosphorylation of tau protein indicating its significant involvement in the disease. The study further explains how α-synuclein, tau and amyloid beta can spread via cross-seeding mechanisms and accelerate each other's aggregation leading to neuronal death. Both GSK-3β and CDK5 are involved in phosphorylation which among other effects contributes to the misfolding of both α-synuclein and tau proteins that lead to neurodegeneration in Alzheimer's disease. Several mediators, that contribute to mitochondrial damage through elevated oxidative stress pathology are clearly described. Because of the increase in the incidence of Parkinson's disease, as predicted to be 17 million when the study was being conducted, studying these pathological mechanisms is very important in trying to establish treatments. This work contributes a path to finding a multi-target treatment regimen to alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Tarini Ashish Sahoo
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Jagdish Chand
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Amarjith Thiyyar Kandy
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Shanish Antony
- Department of Pharmaceutical Sciences, Government Medical College, Kottayam, Kerala, 686008, India
| | - Gomathy Subramanian
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India.
| |
Collapse
|
2
|
Kirimtay K, Huang W, Sun X, Qiang L, Wang DV, Sprouse CT, Craig EM, Baas PW. Tau and MAP6 establish labile and stable domains on microtubules. iScience 2025; 28:111785. [PMID: 40040809 PMCID: PMC11879653 DOI: 10.1016/j.isci.2025.111785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/01/2024] [Accepted: 01/08/2025] [Indexed: 03/06/2025] Open
Abstract
We previously documented that individual microtubules in the axons of cultured juvenile rodent neurons consist of a labile domain and a stable domain and that experimental depletion of tau results in selective shortening and partial stabilization of the labile domain. After first confirming these findings in adult axons, we sought to understand the mechanism that accounts for the formation and maintenance of these microtubule domains. We found that fluorescent tau and MAP6 ectopically expressed in RFL-6 fibroblasts predominantly segregate on different microtubules or different domains on the same microtubule, with the tau-rich ones becoming more labile than in control cells and the MAP6-rich ones being more stable than in control cells. These and other experimental findings, which we studied further using computational modeling with tunable parameters, indicate that these two MAPs do not merely bind to pre-existing stable and labile domains but actually create stable and labile domains on microtubules.
Collapse
Affiliation(s)
- Koray Kirimtay
- Department Neurobiology and Anatomy, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Wenqiang Huang
- Department Neurobiology and Anatomy, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Xiaohuan Sun
- Department Neurobiology and Anatomy, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Liang Qiang
- Department Neurobiology and Anatomy, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Dong V. Wang
- Department Neurobiology and Anatomy, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Calvin T. Sprouse
- Department Physics, Central Washington University, Ellensburg, WA 98926, USA
| | - Erin M. Craig
- Department Physics, Central Washington University, Ellensburg, WA 98926, USA
| | - Peter W. Baas
- Department Neurobiology and Anatomy, Drexel University, 2900 Queen Lane, Philadelphia, PA 19129, USA
| |
Collapse
|
3
|
Di Lorenzo D. Tau Protein and Tauopathies: Exploring Tau Protein-Protein and Microtubule Interactions, Cross-Interactions and Therapeutic Strategies. ChemMedChem 2024; 19:e202400180. [PMID: 39031682 DOI: 10.1002/cmdc.202400180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/22/2024]
Abstract
Tau, a microtubule-associated protein (MAP), is essential to maintaining neuronal stability and function in the healthy brain. However, aberrant modifications and pathological aggregations of Tau are implicated in various neurodegenerative disorders, collectively known as tauopathies. The most common Tauopathy is Alzheimer's Disease (AD) counting nowadays more than 60 million patients worldwide. This comprehensive review delves into the multifaceted realm of Tau protein, puzzling out its intricate involvement in both physiological and pathological roles. Emphasis is put on Tau Protein-Protein Interactions (PPIs), depicting its interaction with tubulin, microtubules and its cross-interaction with other proteins such as Aβ1-42, α-synuclein, and the chaperone machinery. In the realm of therapeutic strategies, an overview of diverse possibilities is presented with their relative clinical progresses. The focus is mostly addressed to Tau protein aggregation inhibitors including recent small molecules, short peptides and peptidomimetics with specific focus on compounds that showed a double anti aggregative activity on both Tau protein and Aβ amyloid peptide. This review amalgamates current knowledge on Tau protein and evolving therapeutic strategies, providing a comprehensive resource for researchers seeking to deepen their understanding of the Tau protein and for scientists involved in the development of new peptide-based anti-aggregative Tau compounds.
Collapse
Affiliation(s)
- Davide Di Lorenzo
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, D-33615, Bielefeld, Germany
| |
Collapse
|
4
|
Hedna R, Kovacic H, Pagano A, Peyrot V, Robin M, Devred F, Breuzard G. Tau Protein as Therapeutic Target for Cancer? Focus on Glioblastoma. Cancers (Basel) 2022; 14:5386. [PMID: 36358803 PMCID: PMC9653627 DOI: 10.3390/cancers14215386] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 08/27/2023] Open
Abstract
Despite being extensively studied for several decades, the microtubule-associated protein Tau has not finished revealing its secrets. For long, Tau has been known for its ability to promote microtubule assembly. A less known feature of Tau is its capability to bind to cancer-related protein kinases, suggesting a possible role of Tau in modulating microtubule-independent cellular pathways that are associated with oncogenesis. With the intention of finding new therapeutic targets for cancer, it appears essential to examine the interaction of Tau with these kinases and their consequences. This review aims at collecting the literature data supporting the relationship between Tau and cancer with a particular focus on glioblastoma tumors in which the pathological significance of Tau remains largely unexplored. We will first treat this subject from a mechanistic point of view showing the pivotal role of Tau in oncogenic processes. Then, we will discuss the involvement of Tau in dysregulating critical pathways in glioblastoma. Finally, we will outline promising strategies to target Tau protein for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Rayane Hedna
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Hervé Kovacic
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Alessandra Pagano
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Vincent Peyrot
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Maxime Robin
- Faculté de Pharmacie, Institut Méditerranéen de Biodiversité et Ecologie marine et continentale (IMBE), UMR 7263, CNRS, IRD 237, Aix-Marseille Université, 13005 Marseille, France
| | - François Devred
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Gilles Breuzard
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| |
Collapse
|
5
|
Haynes BM, Cunningham K, Shekhar MPV. RAD6 inhibition enhances paclitaxel sensitivity of triple negative breast cancer cells by aggravating mitotic spindle damage. BMC Cancer 2022; 22:1073. [PMID: 36258187 PMCID: PMC9578210 DOI: 10.1186/s12885-022-10119-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022] Open
Abstract
Background Paclitaxel (PTX), a first-line therapy for triple negative breast cancers (TNBC) induces anti-tumor activity by microtubule stabilization and inhibition of cell division. Its dose-limiting toxicity and short half-life, however, pose clinical challenges underscoring the need for strategies that increase its efficiency. RAD6, a E2 ubiquitin conjugating enzyme, is associated with centrosomes at all phases of cell cycle. Constitutive overexpression of the RAD6B homolog in normal breast cells induces centrosome amplification and multipolar spindle formation, indicating its importance in centrosome regulation. Methods TNBC centrosome numbers were scored by pericentrin immunostaining. PTX sensitivities and interactions with SMI#9, a RAD6-selective small molecule inhibitor, on TNBC cell survival were analyzed by MTT and colony forming assays and an isogenic MDA-MB-468 TNBC model of PTX resistance. The molecular mechanisms underlying PTX and SMI#9 induced cytotoxicity were determined by flow cytometry, immunoblot analysis of cyclin B1 and microtubule associated protein TAU, and dual immunofluorescence staining of TAU and α-tubulin. Results Our data show aberrant centrosome numbers and that PTX sensitivities are not correlated with TNBC BRCA1 status. Combining PTX with SMI#9 synergistically enhances PTX sensitivities of BRCA1 wild-type and mutant TNBC cells. Whereas SMI#9/PTX combination treatment increased cyclin B1 levels in MDA-MB-468 cells, it induced cyclin B1 loss in HCC1937 cells with accumulation of reproductively dead giant cells, a characteristic of mitotic catastrophe. Cell cycle analysis revealed drug-induced accumulation of tetraploid cells in S and G2/M phases, and robust increases in cells with 4 N DNA content in HCC1937 cells. TAU overexpression is associated with reduced PTX efficacy. Among the six TAU isoforms, both SMI#9 and PTX downregulated 1N3R TAU in MDA-MB-468 and HCC1937 cells, suggesting a common mechanism of 1N3R regulation. Dual TAU and α-tubulin immunostaining showed that SMI#9 induces monopolar mitotic spindles. Using the isogenic model of PTX resistance, we show that SMI#9 treatment restores PTX sensitivity. Conclusions These data support a common mechanism of microtubule regulation by SMI#9 and PTX and suggest that combining PTX with RAD6 inhibitor may be beneficial for increasing TNBC sensitivities to PTX and alleviating toxicity. This study demonstrates a new role for RAD6 in regulating microtubule dynamics. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10119-z.
Collapse
Affiliation(s)
- Brittany M Haynes
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, USA.,Department of Oncology, Wayne State University School of Medicine, 421 E. Canfield Avenue, Detroit, MI, 48201, USA.,Present address: Office of Policy Communications, and Education, National Center for Advancing Translational Sciences, Besthesda, USA
| | - Kristen Cunningham
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, USA.,Department of Oncology, Wayne State University School of Medicine, 421 E. Canfield Avenue, Detroit, MI, 48201, USA
| | - Malathy P V Shekhar
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, USA. .,Department of Oncology, Wayne State University School of Medicine, 421 E. Canfield Avenue, Detroit, MI, 48201, USA. .,Department of Pathology, Wayne State University School of Medicine, 421 E. Canfield Avenue, Detroit, MI, 48201, USA.
| |
Collapse
|
6
|
Wattanathamsan O, Pongrakhananon V. Emerging role of microtubule-associated proteins on cancer metastasis. Front Pharmacol 2022; 13:935493. [PMID: 36188577 PMCID: PMC9515585 DOI: 10.3389/fphar.2022.935493] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/29/2022] [Indexed: 12/29/2022] Open
Abstract
The major cause of death in cancer patients is strongly associated with metastasis. While much remains to be understood, microtubule-associated proteins (MAPs) have shed light on metastatic progression’s molecular mechanisms. In this review article, we focus on the role of MAPs in cancer aggressiveness, particularly cancer metastasis activity. Increasing evidence has shown that a growing number of MAP member proteins might be fundamental regulators involved in altering microtubule dynamics, contributing to cancer migration, invasion, and epithelial-to-mesenchymal transition. MAP types have been established according to their microtubule-binding site and function in microtubule-dependent activities. We highlight that altered MAP expression was commonly found in many cancer types and related to cancer progression based on available evidence. Furthermore, we discuss and integrate the relevance of MAPs and related molecular signaling pathways in cancer metastasis. Our review provides a comprehensive understanding of MAP function on microtubules. It elucidates how MAPs regulate cancer progression, preferentially in metastasis, providing substantial scientific information on MAPs as potential therapeutic targets and prognostic markers for cancer management.
Collapse
Affiliation(s)
- Onsurang Wattanathamsan
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Varisa Pongrakhananon
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- *Correspondence: Varisa Pongrakhananon,
| |
Collapse
|
7
|
Pagano A, Breuzard G, Parat F, Tchoghandjian A, Figarella-Branger D, De Bessa TC, Garrouste F, Douence A, Barbier P, Kovacic H. Tau Regulates Glioblastoma Progression, 3D Cell Organization, Growth and Migration via the PI3K-AKT Axis. Cancers (Basel) 2021; 13:cancers13225818. [PMID: 34830972 PMCID: PMC8616151 DOI: 10.3390/cancers13225818] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The Microtubule-associated protein Tau is expressed in different cancers; however, its role and prognostic value are still debated. In the present work, we evaluated the role of Tau in glioblastoma by down-regulating its expression in glioblastoma cells. We showed that Tau: (1) is required for tumor progression in nude mice; (2) is necessary for glioblastoma 3D cell organization, growth, and migration; and (3) regulates the PI3K/AKT signaling pathway. Abstract The Microtubule-Associated Protein Tau is expressed in several cancers, including low-grade gliomas and glioblastomas. We have previously shown that Tau is crucial for the 2D motility of several glioblastoma cell lines, including U87-MG cells. Using an RNA interference (shRNA), we tested if Tau contributed to glioblastoma in vivo tumorigenicity and analyzed its function in a 3D model of multicellular spheroids (MCS). Tau depletion significantly increased median mouse survival in an orthotopic glioblastoma xenograft model. This was accompanied by the inhibition of MCS growth and cell evasion, as well as decreased MCS compactness, implying N-cadherin mislocalization. Intracellular Signaling Array analysis revealed a defective activation of the PI3K/AKT pathway in Tau-depleted cells. Such a defect in PI3K/AKT signaling was responsible for reduced MCS growth and cell evasion, as demonstrated by the inhibition of the pathway in control MCS using LY294002 or Perifosine, which did not significantly affect Tau-depleted MCS. Finally, analysis of the glioblastoma TCGA dataset showed a positive correlation between the amount of phosphorylated Akt-Ser473 and the expression of MAPT RNA encoding Tau, underlining the relevance of our findings in glioblastoma disease. We suggest a role for Tau in glioblastoma by controlling 3D cell organization and functions via the PI3K/AKT signaling axis.
Collapse
Affiliation(s)
- Alessandra Pagano
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
- Correspondence:
| | - Gilles Breuzard
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Fabrice Parat
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Aurélie Tchoghandjian
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 8, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (A.T.); (D.F.-B.)
| | - Dominique Figarella-Branger
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 8, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (A.T.); (D.F.-B.)
- Service d’Anatomie Pathologique et de Neuropathologie, CHU Timone, APHM, 13005 Marseille, France
| | - Tiphany Coralie De Bessa
- LIM 64: Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-090, SP, Brazil;
| | - Françoise Garrouste
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Alexis Douence
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Pascale Barbier
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Hervé Kovacic
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| |
Collapse
|
8
|
Lo CH. Recent advances in cellular biosensor technology to investigate tau oligomerization. Bioeng Transl Med 2021; 6:e10231. [PMID: 34589603 PMCID: PMC8459642 DOI: 10.1002/btm2.10231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Tau is a microtubule binding protein which plays an important role in physiological functions but it is also involved in the pathogenesis of Alzheimer's disease and related tauopathies. While insoluble and β-sheet containing tau neurofibrillary tangles have been the histopathological hallmark of these diseases, recent studies suggest that soluble tau oligomers, which are formed prior to fibrils, are the primary toxic species. Substantial efforts have been made to generate tau oligomers using purified recombinant protein strategies to study oligomer conformations as well as their toxicity. However, no specific toxic tau species has been identified to date, potentially due to the lack of cellular environment. Hence, there is a need for cell-based models for direct monitoring of tau oligomerization and aggregation. This review will summarize the recent advances in the cellular biosensor technology, with a focus on fluorescence resonance energy transfer, bimolecular fluorescence complementation, and split luciferase complementation approaches, to monitor formation of tau oligomers and aggregates in living cells. We will discuss the applications of the cellular biosensors in examining the heterogeneous tau conformational ensembles and factors affecting tau self-assembly, as well as detecting cell-to-cell propagation of tau pathology. We will also compare the advantages and limitations of each type of tau biosensors, and highlight their translational applications in biomarker development and therapeutic discovery.
Collapse
Affiliation(s)
- Chih Hung Lo
- Department of Neurology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
9
|
Barbolina MV. Dichotomous role of microtubule associated protein tau as a biomarker of response to and a target for increasing efficacy of taxane treatment in cancers of epithelial origin. Pharmacol Res 2021; 168:105585. [PMID: 33798735 PMCID: PMC8165012 DOI: 10.1016/j.phrs.2021.105585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/19/2022]
Abstract
Cancer is the second leading cause of death worldwide, and the World Health Organization estimates that one in six deaths globally is due to cancer. Chemotherapy is one of the major modalities used to treat advanced cancers and their metastasis. However, the existence of acquired and intrinsic resistance to anti-cancer drugs often diminishes their therapeutic effect. In order to pre-select patients who could benefit the most from these treatments, the efforts of many research groups have been focused on identification of biomarkers of therapy response. Taxanes paclitaxel (Taxol) and docetaxel (Taxotere) have been introduced as chemotherapy for treatment of cancers of ovary in 1992 and breast in 1996, respectively. Since then, clinical use of taxanes has expanded to include lung, prostate, gastric, head and neck, esophageal, pancreatic, and cervical cancers, as well as Kaposi sarcoma. Several independent molecular mechanisms have been shown to support taxane chemoresistance. One such mechanism is dependent on microtubule associated protein tau. Tau binds to the same site on the inner side of the microtubules that is also occupied by paclitaxel or docetaxel, and several studies have demonstrated that low/no tau expression significantly correlated with better response to the taxane treatment, suggesting that levels of tau expression could have a predictive value in pre-selecting patient cohorts that are likely to benefit from the treatment. However, several other studies have found no correlation between tau expression and taxane response, introducing a controversy and precluding its wide use as a predictive biomarker. Based on the knowledge of tau biology accumulated thus far, in this review we attempt to critically analyze the studies that evaluated tau as a biomarker of taxane response. Further, we identify yet unknown aspects of tau biology understanding of which is necessary for improvement of development of tau as a biomarker of response and a target for increasing response to taxane treatment.
Collapse
Affiliation(s)
- Maria V Barbolina
- University of Illinois at Chicago, College of Pharmacy, Department of Pharmaceutical Sciences, 833 South Wood Street, Chicago, IL 60612, USA.
| |
Collapse
|
10
|
Dioli C, Patrício P, Pinto LG, Marie C, Morais M, Vyas S, Bessa JM, Pinto L, Sotiropoulos I. Adult neurogenic process in the subventricular zone-olfactory bulb system is regulated by Tau protein under prolonged stress. Cell Prolif 2021; 54:e13027. [PMID: 33988263 PMCID: PMC8249793 DOI: 10.1111/cpr.13027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/08/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives The area of the subventricular zone (SVZ) in the adult brain exhibits the highest number of proliferative cells, which, together with the olfactory bulb (OB), maintains constant brain plasticity through the generation, migration and integration of newly born neurons. Despite Tau and its malfunction is increasingly related to deficits of adult hippocampal neurogenesis and brain plasticity under pathological conditions [e.g. in Alzheimer's disease (AD)], it remains unknown whether Tau plays a role in the neurogenic process of the SVZ and OB system under conditions of chronic stress, a well‐known sculptor of brain and risk factor for AD. Materials and methods Different types of newly born cells in SVZ and OB were analysed in animals that lack Tau gene (Tau‐KO) and their wild‐type littermates (WT) under control or chronic stress conditions. Results We demonstrate that chronic stress reduced the number of proliferating cells and neuroblasts in the SVZ leading to decreased number of newborn neurons in the OB of adult WT, but not Tau‐KO, mice. Interestingly, while stress‐evoked changes were not detected in OB granular cell layer, Tau‐KO exhibited increased number of mature neurons in this layer indicating altered neuronal migration due to Tau loss. Conclusions Our findings suggest the critical involvement of Tau in the neurogenesis suppression of SVZ and OB neurogenic niche under stressful conditions highlighting the role of Tau protein as an essential regulator of stress‐driven plasticity deficits.
Collapse
Affiliation(s)
- Chrysoula Dioli
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.,Institute of Biology Paris Seine, Team Gene Regulation and Adaptive Behaviors, Department of Neurosciences Paris Seine, Sorbonne Université, CNRS UMR 8246, INSERM U1130, Paris, France
| | - Patrícia Patrício
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lucilia-Goreti Pinto
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Clemence Marie
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mónica Morais
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sheela Vyas
- Institute of Biology Paris Seine, Team Gene Regulation and Adaptive Behaviors, Department of Neurosciences Paris Seine, Sorbonne Université, CNRS UMR 8246, INSERM U1130, Paris, France
| | - João M Bessa
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Luisa Pinto
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ioannis Sotiropoulos
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
11
|
Li J, Snyder EY, Tang FHF, Pasqualini R, Arap W, Sidman RL. Nna1 gene deficiency triggers Purkinje neuron death by tubulin hyperglutamylation and ER dysfunction. JCI Insight 2020; 5:136078. [PMID: 33004692 PMCID: PMC7566705 DOI: 10.1172/jci.insight.136078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Posttranslational glutamylation/deglutamylation balance in tubulins influences dendritic maturation and neuronal survival of cerebellar Purkinje neurons (PNs). PNs and some additional neuronal types degenerate in several spontaneous, independently occurring Purkinje cell degeneration (pcd) mice featuring mutant neuronal nuclear protein induced by axotomy (Nna1), a deglutamylase gene. This defective deglutamylase allows glutamylases to form hyperglutamylated tubulins. In pcd, all PNs die during postnatal “adolescence.” Neurons in some additional brain regions also die, mostly later than PNs. We show in laser capture microdissected single PNs, in cerebellar granule cell neuronal clusters, and in dissected hippocampus and substantia nigra that deglutamase mRNA and protein were virtually absent before pcd PNs degenerated, whereas glutaminase mRNA and protein remained normal. Hyperglutamylated microtubules and dimeric tubulins accumulated in pcd PNs and were involved in pcd PN death by glutamylase/deglutamylase imbalance. Importantly, treatment with a microtubule depolymerizer corrected the glutamylation/deglutamylation ratio, increasing PN survival. Further, before onset of neuronal death, pcd PNs displayed prominent basal polylisosomal masses rich in ER. We propose a “seesaw” metamorphic model summarizing mutant Nna1-induced tubulin hyperglutamylation, the pcd’s PN phenotype, and report that the neuronal disorder involved ER stress, unfolded protein response, and protein synthesis inhibition preceding PN death by apoptosis/necroptosis. Purkinje cell degeneration is due to ER stress, unfolded protein response, and protein synthesis inhibition preceding Purkinje neuron death by apoptosis/necroptosis.
Collapse
Affiliation(s)
- Jianxue Li
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Evan Y Snyder
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Fenny HF Tang
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey and Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Richard L Sidman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Kim YC, Jeong BH. In Silico Evaluation of Acetylation Mimics in the 27 Lysine Residues of Human Tau Protein. Curr Alzheimer Res 2020; 16:379-387. [PMID: 30907318 DOI: 10.2174/1567205016666190321161032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/11/2019] [Accepted: 03/17/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND Various neurodegenerative diseases, including Alzheimer's disease (AD), are related to abnormal hyperphosphorylated microtubule-associated protein tau accumulation in brain lesions. Recent studies have focused on toxicity caused by another post-translational modification (PTM), acetylation of the lysine (K) residues of tau protein. Because there are numerous acetylation sites, several studies have introduced mimics of tau acetylation using amino acid substitutions from lysine to glutamine (Q). However, human tau protein contains over 20 acetylation sites; thus, investigation of the effects of an acetylated tau is difficult. OBJECTIVE Here, the authors in silico evaluated acetylation effects using SIFT, PolyPhen-2 and PROVEAN which can estimate the effects of amino acid substitutions based on the sequence homology or protein structure in tau isoforms. In addition, they also investigated 27 acetylation effects on the amyloid formation of tau proteins using Waltz. RESULTS 15 acetylation mimics were estimated to be the most detrimental, which indicates that there may be novel pathogenic acetylation sites in the human tau protein. Interestingly, the deleterious effect of acetylation mimics was different according to the type of isoforms. Furthermore, all acetylation mimics were predicted to be a region of amyloid formation at the codons 274-279 of human tau protein. Notably, acetylation mimic of codon 311 (K311Q) induced the formation of an additional amyloid region located on codons 306-311 of the human tau protein. CONCLUSION To the best of our knowledge, this is the first simultaneous in-silico evaluation of the acetylation state of 27 human tau protein residues.
Collapse
Affiliation(s)
- Yong-Chan Kim
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk 570-390, Korea.,Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk 561-756, Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk 570-390, Korea.,Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk 561-756, Korea
| |
Collapse
|
13
|
Rajaei S, Karima S, Sepasi Tehrani H, Shateri S, Mahmoodi Baram S, Mahdavi M, Mokhtari F, Alimohammadi A, Tafakhori A, Amiri A, Aghamollaii V, Fatemi H, Rajabibazl M, Kobarfard F, Gorji A. Conformational change and GTPase activity of human tubulin: A comparative study on Alzheimer's disease and healthy brain. J Neurochem 2020; 155:207-224. [PMID: 32196663 DOI: 10.1111/jnc.15009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 11/26/2022]
Abstract
In Alzheimer's disease (AD), the most common form of dementia, microtubules (MTs) play a pivotal role through their highly dynamic structure and instability. They mediate axonal transport that is crucial to synaptic viability. MT assembly, dynamic instability and stabilization are modulated by tau proteins, whose detachment initiates MT disintegration. Albeit extensive research, the role of GTPase activity in molecular mechanism of stability remains controversial. We hypothesized that GTPase activity is altered in AD leading to microtubule dynamic dysfunction and ultimately to neuronal death. In this paper, fresh tubulin was purified by chromatography from normal young adult, normal aged, and Alzheimer's brain tissues. Polymerization pattern, assembly kinetics and dynamics, critical concentration, GTPase activity, interaction with tau, intermolecular geometry, and conformational changes were explored via Förster Resonance Energy Transfer (FRET) and various spectroscopy methods. Results showed slower MT assembly process in samples from the brains of people with AD compared with normal young and aged brains. This observation was characterized by prolonged lag phase and increased critical and inactive concentration of tubulin. In addition, the GTPase activity in samples from AD brains was significantly higher than in both normal young and normal aged samples, concurrent with profound conformational changes and contracted intermolecular MT-tau distances as revealed by FRET. These alterations were partially restored in the presence of a microtubule stabilizer, paclitaxel. We proposed that alterations of both tubulin function and GTPase activity may be involved in the molecular neuropathogenesis of AD, thus providing new avenues for therapeutic approaches.
Collapse
Affiliation(s)
- Shima Rajaei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | | | - Somayeh Shateri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Somayeh Mahmoodi Baram
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran.,HealthWeX Clinical Research Co., Ltd., Toronto, ON, Canada
| | - Meisam Mahdavi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Farzad Mokhtari
- HealthWeX Clinical Research Co., Ltd., Toronto, ON, Canada.,Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Alimohammad Alimohammadi
- Research Center of Tehran Forensic Medicine Organization, Forensic Medicine, Legal Medicine Organization Research Center, Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological research, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Amiri
- Research Center of Tehran Forensic Medicine Organization, Forensic Medicine, Legal Medicine Organization Research Center, Tehran, Iran
| | - Vajiheh Aghamollaii
- Neurology Department, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Fatemi
- HealthWeX Clinical Research Co., Ltd., Toronto, ON, Canada
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Farzad Kobarfard
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Ali Gorji
- Department of Neurology and Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
14
|
Xia Y, Sorrentino ZA, Kim JD, Strang KH, Riffe CJ, Giasson BI. Impaired tau-microtubule interactions are prevalent among pathogenic tau variants arising from missense mutations. J Biol Chem 2019; 294:18488-18503. [PMID: 31653695 DOI: 10.1074/jbc.ra119.010178] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/17/2019] [Indexed: 01/22/2023] Open
Abstract
tau is a microtubule (MT)-associated protein that promotes tubulin assembly and stabilizes MTs by binding longitudinally along the MT surface. tau can aberrantly aggregate into pathological inclusions that define Alzheimer's disease, frontotemporal dementias, and other tauopathies. A spectrum of missense mutations in the tau-encoding gene microtubule-associated protein tau (MAPT) can cause frontotemporal dementias. tau aggregation is postulated to spread by a prion-like mechanism. Using a cell-based inclusion seeding assay, we recently reported that only a few tau variants are intrinsically prone to this type of aggregation. Here, we extended these studies to additional tau mutants and investigated their MT binding properties in mammalian cell-based assays. A limited number of tau variants exhibited modest aggregation propensity in vivo, but most tau mutants did not aggregate. Reduced MT binding appeared to be the most common dysfunction for the majority of tau variants due to missense mutations, implying that MT-targeting therapies could potentially be effective in the management of tauopathies.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida 32610; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Zachary A Sorrentino
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida 32610; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Justin D Kim
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida 32610; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Kevin H Strang
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida 32610; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Cara J Riffe
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida 32610; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, Florida 32610; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610.
| |
Collapse
|
15
|
Beyrent E, Gomez G. Oxidative stress differentially induces tau dissociation from neuronal microtubules in neurites of neurons cultured from different regions of the embryonic Gallus domesticus brain. J Neurosci Res 2019; 98:734-747. [PMID: 31621106 DOI: 10.1002/jnr.24541] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/27/2022]
Abstract
Abnormal phosphorylation of microtubule-associated proteins such as tau has been shown to play a role in neurodegenerative disorders. It is hypothesized that oxidative stress-induced aggregates of hyperphosphorylated tau could lead to the microtubule network degradation commonly associated with neurodegeneration. We investigated whether oxidative stress induced tau hyperphosphorylation and focused on neurite degradation using cultured neurons isolated from the embryonic chick brain as a model system. Cells were isolated from the cerebrum, cerebellum, and tectum of 14-day-old chicks, grown separately in culture, and treated with tert-Butyl hydroperoxide (to simulate oxidative stress) for 48 hr. Relative expression and localization of tau or phospho-tau and β-tubulin III in neurites were determined using quantitative immunocytochemistry and confocal microscopy. In untreated cells, tau was tightly colocalized with β-tubulin III. Increasing levels of oxidative stress induced an increase in overall tau expression in neurites of cerebral and tectal but not the cerebellar neurons, coupled with a decrease in phospho-tau expression in tectal but not the cerebral or cerebellar neurons. In addition, oxidative stress induced the degeneration of the distal ends of the neurites and redistribution of phospho-tau toward the neuronal soma in the cerebral but not the tectal and cerebellar neurons. These results suggest that oxidative stress induces changes in tau protein that precede cytoskeletal degradation and neurite retraction. Additionally, there is a differential susceptibility of neuronal subpopulations to oxidative stress, which may offer potential avenues for investigation of the cellular mechanisms underlying the differential manifestations of neurodegenerative disorders in different regions of the brain.
Collapse
Affiliation(s)
- Erika Beyrent
- Biology Department, University of Scranton, Scranton, PA, USA
| | - George Gomez
- Biology Department, University of Scranton, Scranton, PA, USA
| |
Collapse
|
16
|
Barbier P, Zejneli O, Martinho M, Lasorsa A, Belle V, Smet-Nocca C, Tsvetkov PO, Devred F, Landrieu I. Role of Tau as a Microtubule-Associated Protein: Structural and Functional Aspects. Front Aging Neurosci 2019; 11:204. [PMID: 31447664 PMCID: PMC6692637 DOI: 10.3389/fnagi.2019.00204] [Citation(s) in RCA: 336] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/18/2019] [Indexed: 12/24/2022] Open
Abstract
Microtubules (MTs) play a fundamental role in many vital processes such as cell division and neuronal activity. They are key structural and functional elements in axons, supporting neurite differentiation and growth, as well as transporting motor proteins along the axons, which use MTs as support tracks. Tau is a stabilizing MT associated protein, whose functions are mainly regulated by phosphorylation. A disruption of the MT network, which might be caused by Tau loss of function, is observed in a group of related diseases called tauopathies, which includes Alzheimer’s disease (AD). Tau is found hyperphosphorylated in AD, which might account for its loss of MT stabilizing capacity. Since destabilization of MTs after dissociation of Tau could contribute to toxicity in neurodegenerative diseases, a molecular understanding of this interaction and its regulation is essential.
Collapse
Affiliation(s)
- Pascale Barbier
- Fac Pharm, Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), Inst Neurophysiopathol (INP), Fac Pharm, Marseille, France
| | - Orgeta Zejneli
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France.,Univ. Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU-Lille, UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT (JPArc), Lille, France
| | - Marlène Martinho
- Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), UMR 7281, Bioénergétique et Ingénierie des Protéines (BIP), Marseille, France
| | - Alessia Lasorsa
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France
| | - Valérie Belle
- Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), UMR 7281, Bioénergétique et Ingénierie des Protéines (BIP), Marseille, France
| | - Caroline Smet-Nocca
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France
| | - Philipp O Tsvetkov
- Fac Pharm, Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), Inst Neurophysiopathol (INP), Fac Pharm, Marseille, France
| | - François Devred
- Fac Pharm, Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), Inst Neurophysiopathol (INP), Fac Pharm, Marseille, France
| | - Isabelle Landrieu
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France
| |
Collapse
|
17
|
Ukmar-Godec T, Wegmann S, Zweckstetter M. Biomolecular condensation of the microtubule-associated protein tau. Semin Cell Dev Biol 2019; 99:202-214. [PMID: 31260737 DOI: 10.1016/j.semcdb.2019.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
Cells contain multiple compartments dedicated to the regulation and control of biochemical reactions. Cellular compartments that are not surrounded by membranes can rapidly form and dissolve in response to changes in the cellular environment. The physicochemical processes that underlie the formation of non-membrane-bound compartments in vivo are connected to liquid-liquid phase separation of proteins and nucleic acids in vitro. Recent evidence suggests that the protein tau, which plays an important role in Alzheimer's disease and other neurodegenerative disorders, phase separates in solution, forms tau phases with microtubules, and associates with phase-separated RNA-binding protein granules in cells. Here we review the experimental evidence that supports the ability of tau to phase separate in solution and form biomolecular condensates in cells. As for other disease-relevant proteins, the physiological and pathological functions of tau are tightly connected - through loss of normal function or gain of toxic function - and we therefore discuss how tau phase separation plays a role for both, and with respect to different cellular functions of tau.
Collapse
Affiliation(s)
- Tina Ukmar-Godec
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany; Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Chariteplatz 1, 10117, Berlin, Germany.
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany; Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany; Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
18
|
Best RL, LaPointe NE, Liang J, Ruan K, Shade MF, Wilson L, Feinstein SC. Tau isoform-specific stabilization of intermediate states during microtubule assembly and disassembly. J Biol Chem 2019; 294:12265-12280. [PMID: 31266806 DOI: 10.1074/jbc.ra119.009124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/24/2019] [Indexed: 01/27/2023] Open
Abstract
The microtubule (MT)-associated protein tau regulates the critical growing and shortening behaviors of MTs, and its normal activity is essential for neuronal development and maintenance. Accordingly, aberrant tau action is tightly associated with Alzheimer's disease and is genetically linked to several additional neurodegenerative diseases known as tauopathies. Although tau is known to promote net MT growth and stability, the precise mechanistic details governing its regulation of MT dynamics remain unclear. Here, we have used the slowly-hydrolyzable GTP analog, guanylyl-(α,β)-methylene-diphosphonate (GMPCPP), to examine the structural effects of tau at MT ends that may otherwise be too transient to observe. The addition of both four-repeat (4R) and three-repeat (3R) tau isoforms to pre-formed GMPCPP MTs resulted in the formation of extended, multiprotofilament-wide projections at MT ends. Furthermore, at temperatures too low for assembly of bona fide MTs, both tau isoforms promoted the formation of long spiral ribbons from GMPCPP tubulin heterodimers. In addition, GMPCPP MTs undergoing cold-induced disassembly in the presence of 4R tau (and to a much lesser extent 3R tau) also formed spirals. Finally, three pathological tau mutations known to cause neurodegeneration and dementia were differentially compromised in their abilities to stabilize MT disassembly intermediates. Taken together, we propose that tau promotes the formation/stabilization of intermediate states in MT assembly and disassembly by promoting both longitudinal and lateral tubulin-tubulin contacts. We hypothesize that these activities represent fundamental aspects of tau action that normally occur at the GTP-rich ends of GTP/GDP MTs and that may be compromised in neurodegeneration-causing tau variants.
Collapse
Affiliation(s)
- Rebecca L Best
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Nichole E LaPointe
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Jiahao Liang
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Kevin Ruan
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Madeleine F Shade
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Leslie Wilson
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106
| | - Stuart C Feinstein
- Neuroscience Research Institute and Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, California 93106.
| |
Collapse
|
19
|
Moslehi M, Ng DC, Bogoyevitch MA. Pathogenic E2K mutation of doublecortin X (DCX) alters microtubule stabilisation and actin filament association. Biochem Biophys Res Commun 2019; 513:540-545. [DOI: 10.1016/j.bbrc.2019.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 04/01/2019] [Indexed: 10/27/2022]
|
20
|
Tang Y, Liu HL, Min LX, Yuan HS, Guo L, Han PB, Lu YX, Zhong JF, Wang DL. Serum and cerebrospinal fluid tau protein level as biomarkers for evaluating acute spinal cord injury severity and motor function outcome. Neural Regen Res 2019; 14:896-902. [PMID: 30688276 PMCID: PMC6375043 DOI: 10.4103/1673-5374.249238] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/28/2018] [Indexed: 12/19/2022] Open
Abstract
Tau protein, a microtubule-associated protein, has a high specific expression in neurons and axons. Because traumatic spinal cord injury mainly affects neurons and axons, we speculated that tau protein may be a promising biomarker to reflect the degree of spinal cord injury and prognosis of motor function. In this study, 160 female Sprague-Dawley rats were randomly divided into a sham group, and mild, moderate, and severe spinal cord injury groups. A laminectomy was performed at the T8 level to expose the spinal cord in all groups. A contusion lesion was made with the NYU-MASCIS impactor by dropping a 10 g rod from heights of 12.5 mm (mild), 25 mm (moderate) and 50 mm (severe) upon the exposed dorsal surface of the spinal cord. Tau protein levels were measured in serum and cerebrospinal fluid samples at 1, 6, 12, 24 hours, 3, 7, 14 and 28 days after operation. Locomotor function of all rats was assessed using the Basso, Beattie and Bresnahan locomotor rating scale. Tau protein concentration in the three spinal cord injury groups (both in serum and cerebrospinal fluid) rapidly increased and peaked at 12 hours after spinal cord injury. Statistically significant positive linear correlations were found between tau protein level and spinal cord injury severity in the three spinal cord injury groups, and between the tau protein level and Basso, Beattie, and Bresnahan locomotor rating scale scores. The tau protein level at 12 hours in the three spinal cord injury groups was negatively correlated with Basso, Beattie, and Bresnahan locomotor rating scale scores at 28 days (serum: r = -0.94; cerebrospinal fluid: r = -0.95). Our data suggest that tau protein levels in serum and cerebrospinal fluid might be a promising biomarker for predicting the severity and functional outcome of traumatic spinal cord injury.
Collapse
Affiliation(s)
- Ying Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, China
| | - Hong-Liang Liu
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ling-Xia Min
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hao-Shi Yuan
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Lei Guo
- Department of Orthopedics, Chinese PLA Beijing Army General Hospital, Beijing, China
| | - Peng-Bo Han
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Yu-Xin Lu
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Jian-Feng Zhong
- Department of Spine Surgery, Second Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Dong-Lin Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
21
|
Breuzard G, Pagano A, Bastonero S, Malesinski S, Parat F, Barbier P, Peyrot V, Kovacic H. Tau regulates the microtubule-dependent migration of glioblastoma cells via the Rho-ROCK signaling pathway. J Cell Sci 2019; 132:jcs.222851. [PMID: 30659115 DOI: 10.1242/jcs.222851] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/08/2019] [Indexed: 12/24/2022] Open
Abstract
The pathological significance of Tau (encoded by MAPT) in mechanisms driving cell migration in glioblastoma is unclear. By using an shRNA approach to deplete microtubule-stabilizing Tau in U87 cells, we determined its impact on cytoskeletal coordination during migration. We demonstrated here that the motility of these Tau-knockdown cells (shTau cells) was significantly (36%) lower than that of control cells. The shTau cells displayed a slightly changed motility in the presence of nocodazole, which inhibits microtubule formation. Such reduced motility of shTau cells was characterized by a 28% lower number of microtubule bundles at the non-adhesive edges of the tails. In accordance with Tau-stabilized microtubules being required for cell movement, measurements of the front, body and rear section displacements of cells showed inefficient tail retraction in shTau cells. The tail retraction was restored by treatment with Y27632, an inhibitor of Rho-ROCK signaling. Moreover, we clearly identified that shTau cells displayed relocation of the active phosphorylated form of p190-RhoGAP (also known as ARHGAP35), which inhibits Rho-ROCK signaling, and focal adhesion kinase (FAK, also known as PTK2) in cell bodies. In conclusion, our findings indicate that Tau governs the remodeling of microtubule and actin networks for the retraction of the tail of cells, which is necessary for effective migration.
Collapse
Affiliation(s)
- Gilles Breuzard
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| | - Alessandra Pagano
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| | - Sonia Bastonero
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| | - Soazig Malesinski
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| | - Fabrice Parat
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| | - Pascale Barbier
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| | - Vincent Peyrot
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| | - Hervé Kovacic
- Aix-Marseille University, CNRS, Institute of Neurophysiopathology (INP), 13385 Marseille, France
| |
Collapse
|
22
|
Siano G, Varisco M, Caiazza MC, Quercioli V, Mainardi M, Ippolito C, Cattaneo A, Di Primio C. Tau Modulates VGluT1 Expression. J Mol Biol 2019; 431:873-884. [PMID: 30664870 DOI: 10.1016/j.jmb.2019.01.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 02/07/2023]
Abstract
Tau displacement from microtubules is the first step in the onset of tauopathies and is followed by toxic protein aggregation. However, other non-canonical functions of Tau might have a role in these pathologies. Here, we demonstrate that a small amount of Tau localizes in the nuclear compartment and accumulates in both the soluble and chromatin-bound fractions. We show that favoring Tau nuclear translocation and accumulation, by Tau overexpression or detachment from MTs, increases the expression of VGluT1, a disease-relevant gene directly involved in glutamatergic synaptic transmission. Remarkably, the P301L mutation, related to frontotemporal dementia FTDP-17, impairs this mechanism leading to a loss of function. Altogether, our results provide the demonstration of a direct physiological role of Tau on gene expression. Alterations of this mechanism may be at the basis of the onset of neurodegeneration.
Collapse
Affiliation(s)
- Giacomo Siano
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Martina Varisco
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | | | | | - Marco Mainardi
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Chiara Ippolito
- Unit of Histology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Antonino Cattaneo
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | | |
Collapse
|
23
|
Moslehi M, Ng DCH, Bogoyevitch MA. Doublecortin X (DCX) serine 28 phosphorylation is a regulatory switch, modulating association of DCX with microtubules and actin filaments. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:638-649. [PMID: 30625347 DOI: 10.1016/j.bbamcr.2019.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/17/2018] [Accepted: 01/04/2019] [Indexed: 11/18/2022]
Abstract
Doublecortin X (DCX) plays essential roles in neuronal development via its regulation of cytoskeleton dynamics. This is mediated through direct interactions between its doublecortin (DC) domains (DC1 and DC2) with microtubules (MTs) and indirect association with actin filaments (F-ACT). While the regulatory role of the DCX C-terminus following DC2 (i.e. DCX residues 275-366) has been established, less is known of the possible contributions made by the DCX N-terminus preceding DC1 (i.e. DCX residues 1-44). Here, we assessed the influence of DCX Ser28 within the DCX N-terminus, on the association of DCX with MTs and F-ACT. We compared the cytoskeletal interactions of the DCX S28E phosphomimetic and DCX S28A phospho-resistant mutants and wild-type DCX. Immunoprecipitation and colocalisation analyses indicated increased association of DCX S28E with F-ACT but decreased interaction with MTs, and conversely enhanced DCX S28A association with MTs but decreased association with F-ACT. To evaluate the impact of DCX mutants on cytoskeletal filaments we performed fluorescence recovery after photobleaching (FRAP) studies on SiR-tubulin and β-actin-mCherry and observed comparable tubulin and actin exchange rates in the presence of DCX WT and DCX S28A. However, we observed faster tubulin exchange rates but slower actin exchange rates in the presence of DCX S28E. Moreover, DCX S28E enhanced the association with the actin-binding protein spinophilin (Spn) suggesting the shift to favour association with both F-ACT and Spn in the presence of DCX S28E. Taken together, our results highlight a new role for DCX S28 as a regulatory switch for cytoskeletal organisation.
Collapse
Affiliation(s)
- Maryam Moslehi
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Dominic C H Ng
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
24
|
The regulatory effect of Tau protein on polymerization of MCF7 microtubules in vitro. Biochem Biophys Rep 2019; 17:151-156. [PMID: 30671547 PMCID: PMC6327910 DOI: 10.1016/j.bbrep.2018.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/23/2018] [Accepted: 12/24/2018] [Indexed: 11/23/2022] Open
Abstract
Growing evidence continues to point toward the critical role of beta tubulin isotypes in regulating some intracellular functions. Changes that were observed in the microtubules’ intrinsic dynamics, the way they interact with some chemotherapeutic agents, or differences on translocation specifications of some molecular motors along microtubules, were associated to their structural uniqueness in terms of beta tubulin isotype distributions. These findings suggest that the effects of microtubule associated proteins (MAPs) may also vary on structurally different microtubules. Among different microtubule associated proteins, Tau proteins, which are known as neuronal MAPs, bind to beta tubulin, stabilize microtubules, and consequently promote their polymerizations. In this study, in a set of well controlled experiments, the direct effect of Tau proteins on the polymerization of two structurally different microtubules, porcine brain and breast cancer (MCF7), were tested and compared. Remarkably, we found that in contrast with the promoted effect of Tau proteins on brain microtubules’ polymerization, MCF7 expressed a demoted polymerization while interacting with Tau proteins. This finding can potentially be a novel insight into the mechanism of drug resistance in some breast cancer cells. It has been reported that microtubules show destabilizing behavior in some MCF7 cells with overexpression of Tau protein when treated with a microtubules’ stabilizing agent, Taxol. This behavior has been classified by others as drug resistance, but it may instead be potentially caused by a competition between the destabilizing effect of the Tau protein and the stabilizing effect of the drug on MCF7 microtubules. Also, we quantified the polarization coefficient of MCF7 microtubules in the presence and absence of Tau proteins by the electro-orientation method and compared the values. The two significantly different values obtained can possibly be one factor considered to explain the effect of Tau proteins on the polymerization of MCF7 microtubules. MCF7 microtubules express slow and stable polymerization behavior. Tau-MCF7 microtubules express demoted polymerization behavior. Tau-MCF7 polymerization can possibly be explained by electrostatic specifications.
Collapse
|
25
|
Design, synthesis and molecular modeling of new 4-phenylcoumarin derivatives as tubulin polymerization inhibitors targeting MCF-7 breast cancer cells. Bioorg Med Chem 2018; 26:3474-3490. [DOI: 10.1016/j.bmc.2018.05.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/13/2018] [Accepted: 05/15/2018] [Indexed: 11/21/2022]
|
26
|
Terciolo C, Dobric A, Ouaissi M, Siret C, Breuzard G, Silvy F, Marchiori B, Germain S, Bonier R, Hama A, Owens R, Lombardo D, Rigot V, André F. Saccharomyces boulardii CNCM I-745 Restores intestinal Barrier Integrity by Regulation of E-cadherin Recycling. J Crohns Colitis 2017; 11:999-1010. [PMID: 28333335 DOI: 10.1093/ecco-jcc/jjx030] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/21/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Alteration in intestinal permeability is the main factor underlying the pathogenesis of many diseases affecting the gut, such as inflammatory bowel disease [IBD]. Characterization of molecules targeting the restoration of intestinal barrier integrity is therefore vital for the development of alternative therapies. The yeast Saccharomyces boulardii CNCM I-745 [Sb], used to prevent and treat antibiotic-associated infectious and functional diarrhea, may have a beneficial effect in the treatment of IBD. METHODS We analyzed the impact of Sb supernatant on tissue integrity and components of adherens junctions using cultured explants of colon from both IBD and healthy patients. To evaluate the pathways by which Sb regulates the expression of E-cadherin at the cell surface, we developed in vitro assays using human colonic cell lines, including cell aggregation, a calcium switch assay, real-time measurement of transepithelial electrical resistance [TEER] and pulse-chase experiments. RESULTS We showed that Sb supernatant treatment of colonic explants protects the epithelial morphology and maintains E-cadherin expression at the cell surface. In vitro experiments revealed that Sb supernatant enhances E-cadherin delivery to the cell surface by re-routing endocytosed E-cadherin back to the plasma membrane. This process, involving Rab11A-dependent recycling endosome, leads to restoration of enterocyte adherens junctions, in addition to the overall restoration and strengthening of intestinal barrier function. CONCLUSION These findings open new possibilities of discovering novel options for prevention and therapy of diseases that affect intestinal permeability.
Collapse
Affiliation(s)
- Chloé Terciolo
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Aurélie Dobric
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Mehdi Ouaissi
- Aix-Marseille Université, Inserm, AP-HM, UMR 911, CRO2, Marseille, France
| | - Carole Siret
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Gilles Breuzard
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Françoise Silvy
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | | | | | - Renaté Bonier
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Adel Hama
- Ecole des Mines de Saint Etienne, BEL, Gardanne, France
| | - Roisin Owens
- Ecole des Mines de Saint Etienne, BEL, Gardanne, France
| | | | - Véronique Rigot
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Frédéric André
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| |
Collapse
|
27
|
Dynamic microtubule association of Doublecortin X (DCX) is regulated by its C-terminus. Sci Rep 2017; 7:5245. [PMID: 28701724 PMCID: PMC5507856 DOI: 10.1038/s41598-017-05340-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022] Open
Abstract
Doublecortin X (DCX), known to be essential for neuronal migration and cortical layering in the developing brain, is a 40 kDa microtubule (MT)-associated protein. DCX directly interacts with MTs via its two structured doublecortin (DC) domains, but the dynamics of this association and the possible regulatory roles played by the flanking unstructured regions remain poorly defined. Here, we employ quantitative fluorescence recovery after photobleaching (FRAP) protocols in living cells to reveal that DCX shows remarkably rapid and complete exchange within the MT network but that the removal of the C-terminal region significantly slows this exchange. We further probed how MT organization or external stimuli could additionally modulate DCX exchange dynamics. MT depolymerisation (nocodazole treatment) or stabilization (taxol treatment) further enhanced DCX exchange rates, however the exchange rates for the C-terminal truncated DCX protein were resistant to the impact of taxol-induced stabilization. Furthermore, in response to a hyperosmotic stress stimulus, DCX exchange dynamics were slowed, and again the C-terminal truncated DCX protein was resistant to the stimulus. Thus, the DCX dynamically associates with MTs in living cells and its C-terminal region plays important roles in the MT-DCX association.
Collapse
|
28
|
Di Primio C, Quercioli V, Siano G, Rovere M, Kovacech B, Novak M, Cattaneo A. The Distance between N and C Termini of Tau and of FTDP-17 Mutants Is Modulated by Microtubule Interactions in Living Cells. Front Mol Neurosci 2017; 10:210. [PMID: 28713242 PMCID: PMC5492851 DOI: 10.3389/fnmol.2017.00210] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/14/2017] [Indexed: 11/22/2022] Open
Abstract
The microtubule (MT)-associated protein Tau is a natively unfolded protein, involved in a number of neurodegenerative disorders, collectively called tauopathies, aggregating in neurofibrillary tangles (NFT). It is an open question how the conversion from a MT bound molecule to an aggregation-prone Tau species occurs and, also, if and how tauopathy-related mutations affect its behavior in the cell. To address these points, we exploited a genetically encoded FRET sensor based on the full length Tau protein, to monitor in real time Tau conformational changes in different conditions in live cells. By studying the FRET signal we found that soluble Tau molecules, detached from MTs, display an unfolded structure. On the contrary, we observed an increased FRET signal generated by Tau monomers bound to MT, indicating that the association with MTs induced a folding of Tau protein, decreasing the distance between its N and C termini. We exploited the FRET sensor to investigate the impact of FTDP-17 mutations and of phosphorylation-site mutations on Tau folding and mobility in live cells. We demonstrated that the FTDP-17 Tau mutations weaken the interaction of Tau with cellular MTs, shifting the equilibrium towards the soluble pool while, conversely, phosphorylation site mutations shift the equilibrium of Tau towards the MT-bound state and a more closed conformation.
Collapse
Affiliation(s)
| | | | - Giacomo Siano
- Bio@SNS Laboratory, Scuola Normale SuperiorePisa, Italy
| | - Matteo Rovere
- Bio@SNS Laboratory, Scuola Normale SuperiorePisa, Italy
| | - Branislav Kovacech
- Institute of Neuroimmunology, Slovak Academy of Sciences, Axon Neuroscience SEBratislava, Slovakia
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Axon Neuroscience SEBratislava, Slovakia
| | | |
Collapse
|
29
|
Duan AR, Jonasson EM, Alberico EO, Li C, Scripture JP, Miller RA, Alber MS, Goodson HV. Interactions between Tau and Different Conformations of Tubulin: Implications for Tau Function and Mechanism. J Mol Biol 2017; 429:1424-1438. [PMID: 28322917 DOI: 10.1016/j.jmb.2017.03.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/22/2017] [Accepted: 03/12/2017] [Indexed: 11/16/2022]
Abstract
Tau is a multifaceted neuronal protein that stabilizes microtubules (MTs), but the mechanism of this activity remains poorly understood. Questions include whether Tau binds MTs laterally or longitudinally and whether Tau's binding affinity depends on the nucleotide state of tubulin. We observed that Tau binds tightly to Dolastatin-10 tubulin rings and promotes the formation of Dolastatin-10 ring stacks, implying that Tau can crosslink MT protofilaments laterally. In addition, we found that Tau prefers GDP-like tubulin conformations, which implies that Tau binding to the MT surface is biased away from the dynamic GTP-rich MT tip. To investigate the potential impact of these Tau activities on MT stabilization, we incorporated them into our previously developed dimer-scale computational model of MT dynamics. We found that lateral crosslinking activities have a much greater effect on MT stability than do longitudinal crosslinking activities, and that introducing a bias toward GDP tubulin has little impact on the observed MT stabilization. To address the question of why Tau is GDP-tubulin-biased, we tested whether Tau might affect MT binding of the +TIP EB1. We confirmed recent reports that Tau binds directly to EB1 and that Tau competes with EB1 for MT binding. Our results lead to a conceptual model where Tau stabilizes the MT lattice by strengthening lateral interactions between protofilaments. We propose that Tau's GDP preference allows the cell to independently regulate the dynamics of the MT tip and the stability of the lattice.
Collapse
Affiliation(s)
- Aranda R Duan
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Erin M Jonasson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Emily O Alberico
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Chunlei Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jared P Scripture
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel A Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Mark S Alber
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Mathematics, University of California, Riverside, CA 92521, USA
| | - Holly V Goodson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
30
|
Mutai P, Breuzard G, Pagano A, Allegro D, Peyrot V, Chibale K. Synthesis and biological evaluation of 4 arylcoumarin analogues as tubulin-targeting antitumor agents. Bioorg Med Chem 2017; 25:1652-1665. [DOI: 10.1016/j.bmc.2017.01.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 10/20/2022]
|
31
|
De Bessa T, Breuzard G, Allegro D, Devred F, Peyrot V, Barbier P. Tau Interaction with Tubulin and Microtubules: From Purified Proteins to Cells. Methods Mol Biol 2017; 1523:61-85. [PMID: 27975244 DOI: 10.1007/978-1-4939-6598-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Microtubules (MTs) play an important role in many cellular processes and are dynamic structures regulated by an important network of microtubules-associated proteins, MAPs, such as Tau. Tau has been discovered as an essential factor for MTs formation in vitro, and its region implicated in binding to MTs has been identified. By contrast, the affinity, the stoichiometry, and the topology of Tau-MTs interaction remain controversial. Indeed, depending on the experiment conditions a wide range of values have been obtained. In this chapter, we focus on three biophysical methods, turbidimetry, cosedimentation assay, and Förster Resonance Energy Transfer to study Tau-tubulin interaction both in vitro and in cell. We highlight precautions that must be taken in order to avoid pitfalls and we detail the nature of the conclusions that can be drawn from these methods about Tau-tubulin interaction.
Collapse
Affiliation(s)
- Tiphany De Bessa
- Aix-Marseille Université, INSERM, CRO2, UMR_S 911, 13385, Marseille, France
| | - Gilles Breuzard
- Aix-Marseille Université, INSERM, CRO2, UMR_S 911, 13385, Marseille, France
| | - Diane Allegro
- Aix-Marseille Université, INSERM, CRO2, UMR_S 911, 13385, Marseille, France
| | - François Devred
- Aix-Marseille Université, INSERM, CRO2, UMR_S 911, 13385, Marseille, France
| | - Vincent Peyrot
- Aix-Marseille Université, INSERM, CRO2, UMR_S 911, 13385, Marseille, France
| | - Pascale Barbier
- Aix-Marseille Université, INSERM, CRO2, UMR_S 911, 13385, Marseille, France.
| |
Collapse
|
32
|
Smit FX, Luiken JA, Bolhuis PG. Primary Fibril Nucleation of Aggregation Prone Tau Fragments PHF6 and PHF6. J Phys Chem B 2016; 121:3250-3261. [PMID: 27776213 DOI: 10.1021/acs.jpcb.6b07045] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We performed replica exchange molecular dynamics and forward flux sampling simulations of hexapeptide VQIINK and VQIVYK systems, also known as, respectively, fragments PHF6* and PHF6 from the tau protein. Being a part of the microtubule binding region, these fragments are known to be aggregation prone, and at least one of them is a prerequisite for fibril formation of the tau protein. Using a coarse-grained force field, we establish the phase behavior of both fragments, and investigate the nucleation kinetics for the conversion into a β-sheet fibril. As the conversion is, in principle, a reversible process, we predict the rate constants for both the fibril formation and melting, and examine the corresponding mechanisms. Our simulations indicate that, while both fragments form disordered aggregates, only PHF6 is able to form β-sheet fibrils. This observation provides a possible explanation for the lack of available steric zipper crystal structures for PHF6*.
Collapse
Affiliation(s)
- Florent X Smit
- van't Hoff Institute for Molecular Sciences, University of Amsterdam , PO Box 94157, 1090 GD Amsterdam, The Netherlands
| | - Jurriaan A Luiken
- van't Hoff Institute for Molecular Sciences, University of Amsterdam , PO Box 94157, 1090 GD Amsterdam, The Netherlands
| | - Peter G Bolhuis
- van't Hoff Institute for Molecular Sciences, University of Amsterdam , PO Box 94157, 1090 GD Amsterdam, The Netherlands
| |
Collapse
|
33
|
Méphon-Gaspard A, Boca M, Pioche-Durieu C, Desforges B, Burgo A, Hamon L, Piétrement O, Pastré D. Role of tau in the spatial organization of axonal microtubules: keeping parallel microtubules evenly distributed despite macromolecular crowding. Cell Mol Life Sci 2016; 73:3745-60. [PMID: 27076215 PMCID: PMC5002045 DOI: 10.1007/s00018-016-2216-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/24/2016] [Accepted: 04/01/2016] [Indexed: 02/07/2023]
Abstract
Opposing views have been proposed regarding the role of tau, the principal microtubule-associated protein in axons. On the one hand, tau forms cross-bridges at the interface between microtubules and induces microtubule bundling in neurons. On the other hand, tau is also considered a polymer brush which efficiently separates microtubules. In mature axons, microtubules are indeed arranged in parallel arrays and are well separated from each other. To reconcile these views, we developed a mechanistic model based on in vitro and cellular approaches combined to analytical and numerical analyses. The results indicate that tau forms long-range cross-bridges between microtubules under macromolecular crowding conditions. Tau cross-bridges prevent the redistribution of tau away from the interface between microtubules, which would have occurred in the polymer brush model. Consequently, the short-range attractive force between microtubules induced by macromolecular crowding is avoided and thus microtubules remain well separated from each other. Interestingly, in this unified model, tau diffusion on microtubules enables to keep microtubules evenly distributed in axonal sections at low tau levels.
Collapse
Affiliation(s)
- Alix Méphon-Gaspard
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Mirela Boca
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Catherine Pioche-Durieu
- UMR 8126, CNRS, Gustave Roussy Université Paris Sud, Université Paris-Saclay, Villejuif, 94805, France
| | - Bénédicte Desforges
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Andrea Burgo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Loic Hamon
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Olivier Piétrement
- UMR 8126, CNRS, Gustave Roussy Université Paris Sud, Université Paris-Saclay, Villejuif, 94805, France
| | - David Pastré
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France.
| |
Collapse
|
34
|
Nouar R, Breuzard G, Bastonero S, Gorokhova S, Barbier P, Devred F, Kovacic H, Peyrot V. Direct evidence for the interaction of stathmin along the length and the plus end of microtubules in cells. FASEB J 2016; 30:3202-15. [DOI: 10.1096/fj.201500125r] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/31/2015] [Indexed: 02/04/2023]
Affiliation(s)
- Roqiya Nouar
- Aix Marseille Université Mixte de Recherche (UMR) 911Center for Research in Oncobiology and Oncopharmacology (CRO2)Faculté de Pharmacie Marseille France
| | - Gilles Breuzard
- Aix Marseille Université Mixte de Recherche (UMR) 911Center for Research in Oncobiology and Oncopharmacology (CRO2)Faculté de Pharmacie Marseille France
| | - Sonia Bastonero
- Aix Marseille Université Mixte de Recherche (UMR) 911Center for Research in Oncobiology and Oncopharmacology (CRO2)Faculté de Pharmacie Marseille France
| | - Svetlana Gorokhova
- Aix Marseille Université, INSERM UMR 910Génétique Médicale et Génomique Fonctionnelle (GMGF)Faculté de Médecine Marseille France
| | - Pascale Barbier
- Aix Marseille Université Mixte de Recherche (UMR) 911Center for Research in Oncobiology and Oncopharmacology (CRO2)Faculté de Pharmacie Marseille France
| | - François Devred
- Aix Marseille Université Mixte de Recherche (UMR) 911Center for Research in Oncobiology and Oncopharmacology (CRO2)Faculté de Pharmacie Marseille France
| | - Hervé Kovacic
- Aix Marseille Université Mixte de Recherche (UMR) 911Center for Research in Oncobiology and Oncopharmacology (CRO2)Faculté de Pharmacie Marseille France
| | - Vincent Peyrot
- Aix Marseille Université Mixte de Recherche (UMR) 911Center for Research in Oncobiology and Oncopharmacology (CRO2)Faculté de Pharmacie Marseille France
| |
Collapse
|
35
|
Feinstein HE, Benbow SJ, LaPointe NE, Patel N, Ramachandran S, Do TD, Gaylord MR, Huskey NE, Dressler N, Korff M, Quon B, Cantrell KL, Bowers MT, Lal R, Feinstein SC. Oligomerization of the microtubule-associated protein tau is mediated by its N-terminal sequences: implications for normal and pathological tau action. J Neurochem 2016; 137:939-54. [PMID: 26953146 PMCID: PMC4899250 DOI: 10.1111/jnc.13604] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/10/2016] [Accepted: 03/06/2016] [Indexed: 11/28/2022]
Abstract
Despite extensive structure-function analyses, the molecular mechanisms of normal and pathological tau action remain poorly understood. How does the C-terminal microtubule-binding region regulate microtubule dynamics and bundling? In what biophysical form does tau transfer trans-synaptically from one neuron to another, promoting neurodegeneration and dementia? Previous biochemical/biophysical work led to the hypothesis that tau can dimerize via electrostatic interactions between two N-terminal 'projection domains' aligned in an anti-parallel fashion, generating a multivalent complex capable of interacting with multiple tubulin subunits. We sought to test this dimerization model directly. Native gel analyses of full-length tau and deletion constructs demonstrate that the N-terminal region leads to multiple bands, consistent with oligomerization. Ferguson analyses of native gels indicate that an N-terminal fragment (tau(45-230) ) assembles into heptamers/octamers. Ferguson analyses of denaturing gels demonstrates that tau(45-230) can dimerize even in sodium dodecyl sulfate. Atomic force microscopy reveals multiple levels of oligomerization by both full-length tau and tau(45-230) . Finally, ion mobility-mass spectrometric analyses of tau(106-144) , a small peptide containing the core of the hypothesized dimerization region, also demonstrate oligomerization. Thus, multiple independent strategies demonstrate that the N-terminal region of tau can mediate higher order oligomerization, which may have important implications for both normal and pathological tau action. The microtubule-associated protein tau is essential for neuronal development and maintenance, but is also central to Alzheimer's and related dementias. Unfortunately, the molecular mechanisms underlying normal and pathological tau action remain poorly understood. Here, we demonstrate that tau can homo-oligomerize, providing novel mechanistic models for normal tau action (promoting microtubule growth and bundling, suppressing microtubule shortening) and pathological tau action (poisoning of oligomeric complexes).
Collapse
Affiliation(s)
- H Eric Feinstein
- Neuroscience Research Institute, University of California, Santa Barbara, California, USA
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Sarah J Benbow
- Neuroscience Research Institute, University of California, Santa Barbara, California, USA
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Nichole E LaPointe
- Neuroscience Research Institute, University of California, Santa Barbara, California, USA
| | - Nirav Patel
- Department of Bioengineering, Department of Mechanical Engineering and Materials Science Graduate Program, University of California, San Diego, California, USA
| | - Srinivasan Ramachandran
- Department of Bioengineering, Department of Mechanical Engineering and Materials Science Graduate Program, University of California, San Diego, California, USA
| | - Thanh D Do
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California, USA
| | - Michelle R Gaylord
- Neuroscience Research Institute, University of California, Santa Barbara, California, USA
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Noelle E Huskey
- Neuroscience Research Institute, University of California, Santa Barbara, California, USA
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| | - Nicolette Dressler
- Department of Chemistry, Westmont College, Santa Barbara, California, USA
| | - Megan Korff
- Department of Chemistry, Westmont College, Santa Barbara, California, USA
| | - Brady Quon
- Department of Chemistry, Westmont College, Santa Barbara, California, USA
| | | | - Michael T Bowers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California, USA
| | - Ratnesh Lal
- Department of Bioengineering, Department of Mechanical Engineering and Materials Science Graduate Program, University of California, San Diego, California, USA
| | - Stuart C Feinstein
- Neuroscience Research Institute, University of California, Santa Barbara, California, USA
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California, USA
| |
Collapse
|
36
|
Regulation of Microtubule Assembly by Tau and not by Pin1. J Mol Biol 2016; 428:1742-59. [PMID: 26996940 DOI: 10.1016/j.jmb.2016.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/08/2016] [Accepted: 03/10/2016] [Indexed: 11/21/2022]
Abstract
The molecular mechanism by which the microtubule-associated protein (MAP) tau regulates the formation of microtubules (MTs) is poorly understood. The activity of tau is controlled via phosphorylation at specific Ser/Thr sites. Of those phosphorylation sites, 17 precede a proline, making them potential recognition sites for the peptidyl-prolyl isomerase Pin1. Pin1 binding and catalysis of phosphorylated tau at the AT180 epitope, which was implicated in Alzheimer's disease, has been reported to be crucial for restoring tau's ability to promote MT polymerization in vitro and in vivo [1]. Surprisingly, we discover that Pin1 does not promote phosphorylated tau-induced MT formation in vitro, refuting the commonly accepted model in which Pin1 binding and catalysis on the A180 epitope restores the function of the Alzheimer's associated phosphorylated tau in tubulin assembly [1, 2]. Using turbidity assays, time-resolved small angle X-ray scattering (SAXS), and time-resolved negative stain electron microscopy (EM), we investigate the mechanism of tau-mediated MT assembly and the role of the Thr231 and Ser235 phosphorylation on this process. We discover novel GTP-tubulin ring-shaped species, which are detectable in the earliest stage of tau-induced polymerization and may play a crucial role in the early nucleation phase of MT assembly. Finally, by NMR and SAXS experiments, we show that the tau molecules must be located on the surface of MTs and tubulin rings during the polymerization reaction. The interaction between tau and tubulin is multipartite, with a high affinity interaction of the four tubulin-binding repeats, and a weaker interaction with the proline-rich sequence and the termini of tau.
Collapse
|
37
|
Fontaine SN, Martin MD, Akoury E, Assimon VA, Borysov S, Nordhues BA, Sabbagh JJ, Cockman M, Gestwicki JE, Zweckstetter M, Dickey CA. The active Hsc70/tau complex can be exploited to enhance tau turnover without damaging microtubule dynamics. Hum Mol Genet 2015; 24:3971-81. [PMID: 25882706 DOI: 10.1093/hmg/ddv135] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/13/2015] [Indexed: 11/12/2022] Open
Abstract
The pathological accumulation of abnormally hyperphosphorylated and aggregated tau, a neuronal microtubule (MT)-associated protein that functions to maintain MT stability, is implicated in a number of hereditary and sporadic neurodegenerative diseases including frontotemporal dementia and Alzheimer's disease. Targeting tau for the treatment of these diseases is an area of intense interest and toward that end, modulation of cellular molecular chaperones is a potential therapeutic target. In particular, the constitutive Hsp70 isoform, Hsc70, seems highly interconnected with tau, preserving tau protein levels and synergizing with it to assemble MTs. But the relationship between tau and Hsc70, as well as the impact of this interaction in neurons and its therapeutic implications remain unknown. Using a human dominant negative Hsc70 that resembles isoform selective inhibition of this important chaperone, we found for the first time that Hsc70 activity is required to stimulate MT assembly in cells and brain. However, surprisingly, active Hsc70 also requires active tau to regulate MT assembly in vivo, suggesting that tau acts in some ways as a co-chaperone for Hsc70 to coordinate MT assembly. This was despite tau binding to Hsc70 as substrate, as determined biochemically. Moreover, we show that while chronic Hsc70 inhibition damaged MT dynamics, intermittent treatment with a small molecule Hsp70 inhibitor lowered tau in brain tissue without disrupting MT integrity. Thus, in tauopathies, where MT injury would be detrimental to neurons, the unique relationship of tau with the Hsc70 machinery can be exploited to deplete tau levels without damaging MT networks.
Collapse
Affiliation(s)
- Sarah N Fontaine
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA, James A. Haley Veteran's Hospital, 13000 Bruce B. Downs Blvd, Tampa, FL 33612, USA
| | - Mackenzie D Martin
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - Elias Akoury
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany, German Center for Neurodegenerative Diseases (DZNE), Göttingen 37077, Germany, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center, Göttingen 37073, Germany and
| | - Victoria A Assimon
- Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Sergiy Borysov
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - Bryce A Nordhues
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA, James A. Haley Veteran's Hospital, 13000 Bruce B. Downs Blvd, Tampa, FL 33612, USA
| | - Jonathan J Sabbagh
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA, James A. Haley Veteran's Hospital, 13000 Bruce B. Downs Blvd, Tampa, FL 33612, USA
| | - Matt Cockman
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - Jason E Gestwicki
- Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Markus Zweckstetter
- Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany, German Center for Neurodegenerative Diseases (DZNE), Göttingen 37077, Germany, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center, Göttingen 37073, Germany and
| | - Chad A Dickey
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA, James A. Haley Veteran's Hospital, 13000 Bruce B. Downs Blvd, Tampa, FL 33612, USA,
| |
Collapse
|
38
|
Fontaine SN, Sabbagh JJ, Baker J, Martinez-Licha CR, Darling A, Dickey CA. Cellular factors modulating the mechanism of tau protein aggregation. Cell Mol Life Sci 2015; 72:1863-79. [PMID: 25666877 DOI: 10.1007/s00018-015-1839-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/18/2014] [Accepted: 01/13/2015] [Indexed: 01/12/2023]
Abstract
Pathological accumulation of the microtubule-associated protein tau, in the form of neurofibrillary tangles, is a major hallmark of Alzheimer's disease, the most prevalent neurodegenerative condition worldwide. In addition to Alzheimer's disease, a number of neurodegenerative diseases, called tauopathies, are characterized by the accumulation of aggregated tau in a variety of brain regions. While tau normally plays an important role in stabilizing the microtubule network of the cytoskeleton, its dissociation from microtubules and eventual aggregation into pathological deposits is an area of intense focus for therapeutic development. Here we discuss the known cellular factors that affect tau aggregation, from post-translational modifications to molecular chaperones.
Collapse
Affiliation(s)
- Sarah N Fontaine
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
| | | | | | | | | | | |
Collapse
|
39
|
Kanaan NM, Himmelstein DS, Ward SM, Combs B, Binder LI. Tau Protein. Mov Disord 2015. [DOI: 10.1016/b978-0-12-405195-9.00056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
40
|
A refined reaction-diffusion model of tau-microtubule dynamics and its application in FDAP analysis. Biophys J 2014; 107:2567-78. [PMID: 25468336 DOI: 10.1016/j.bpj.2014.09.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/22/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022] Open
Abstract
Fluorescence decay after photoactivation (FDAP) and fluorescence recovery after photobleaching (FRAP) are well established approaches for studying the interaction of the microtubule (MT)-associated protein tau with MTs in neuronal cells. Previous interpretations of FDAP/FRAP data have revealed dwell times of tau on MTs in the range of several seconds. However, this is difficult to reconcile with a dwell time recently measured by single-molecule analysis in neuronal processes that was shorter by two orders of magnitude. Questioning the validity of previously used phenomenological interpretations of FDAP/FRAP data, we have generalized the standard two-state reaction-diffusion equations by 1), accounting for the parallel and discrete arrangement of MTs in cell processes (i.e., homogeneous versus heterogeneous distribution of tau-binding sites); and 2), explicitly considering both active (diffusion upon MTs) and passive (piggybacking upon MTs at rates of slow axonal transport) motion of bound tau. For some idealized cases, analytical solutions were derived. By comparing them with the full numerical solution and Monte Carlo simulations, the respective validity domains were mapped. Interpretation of our FDAP data (from processes of neuronally differentiated PC12 cells) in light of the heterogeneous formalism yielded independent estimates for the association (∼2 ms) and dwell (∼100 ms) times of tau to/on a single MT rather than in an MT array. The dwell time was shorter by orders of magnitude than that in a previous report where a homogeneous topology of MTs was assumed. We found that the diffusion of bound tau was negligible in vivo, in contrast to an earlier report that tau diffuses along the MT lattice in vitro. Methodologically, our results demonstrate that the heterogeneity of binding sites cannot be ignored when dealing with reaction-diffusion of cytoskeleton-associated proteins. Physiologically, the results reveal the behavior of tau in cellular processes, which is noticeably different from that in vitro.
Collapse
|
41
|
Werner ME, Mitchell JW, Putzbach W, Bacon E, Kim SK, Mitchell BJ. Radial intercalation is regulated by the Par complex and the microtubule-stabilizing protein CLAMP/Spef1. J Cell Biol 2014; 206:367-76. [PMID: 25070955 PMCID: PMC4121976 DOI: 10.1083/jcb.201312045] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 06/25/2014] [Indexed: 01/28/2023] Open
Abstract
The directed movement of cells is critical for numerous developmental and disease processes. A developmentally reiterated form of migration is radial intercalation; the process by which cells move in a direction orthogonal to the plane of the tissue from an inner layer to an outer layer. We use the radial intercalation of cells into the skin of Xenopus laevis embryos as a model to study directed cell migration within an epithelial tissue. We identify a novel function for both the microtubule-binding protein CLAMP and members of the microtubule-regulating Par complex during intercalation. Specifically, we show that Par3 and aPKC promote the apical positioning of centrioles, whereas CLAMP stabilizes microtubules along the axis of migration. We propose a model in which the Par complex defines the orientation of apical migration during intercalation and in which subcellular localization of CLAMP promotes the establishment of an axis of microtubule stability required for the active migration of cells into the outer epithelium.
Collapse
Affiliation(s)
- Michael E Werner
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Jennifer W Mitchell
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - William Putzbach
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Elizabeth Bacon
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Sun K Kim
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Brian J Mitchell
- Department of Cell and Molecular Biology and Driskill Graduate Program in Life Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
42
|
Cook C, Stankowski JN, Carlomagno Y, Stetler C, Petrucelli L. Acetylation: a new key to unlock tau's role in neurodegeneration. ALZHEIMERS RESEARCH & THERAPY 2014; 6:29. [PMID: 25031639 PMCID: PMC4075151 DOI: 10.1186/alzrt259] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The identification of tau protein as a major constituent of neurofibrillary tangles spurred considerable effort devoted to identifying and validating pathways through which therapeutics may alleviate tau burden in Alzheimer's disease and related tauopathies, including chronic traumatic encephalopathy associated with sport- and military-related injuries. Most tau-based therapeutic strategies have previously focused on modulating tau phosphorylation, given that tau species present within neurofibrillary tangles are hyperphosphorylated on a number of different residues. However, the recent discovery that tau is modified by acetylation necessitates additional research to provide greater mechanistic insight into the spectrum of physiological consequences of tau acetylation, which may hold promise as a novel therapeutic target. In this review, we discuss recent findings evaluating tau acetylation in the context of previously accepted notions regarding tau biology and pathophysiology. We also examine the evidence demonstrating the neuroprotective and beneficial consequences of inhibiting histone deacetylase (HDAC)6, a tau deacetylase, including its effect on microtubule stabilization. We also discuss the rationale for pharmacologically modulating HDAC6 in tau-based pathologies as a novel therapeutic strategy.
Collapse
Affiliation(s)
- Casey Cook
- Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | - Yari Carlomagno
- Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | | | | |
Collapse
|