1
|
Girão H, Macário-Monteiro J, Figueiredo AC, Silva E Sousa R, Doria E, Demidov V, Osório H, Jacome A, Meraldi P, Grishchuk EL, Maiato H. α-tubulin detyrosination fine-tunes kinetochore-microtubule attachments. Nat Commun 2024; 15:9720. [PMID: 39521805 PMCID: PMC11550433 DOI: 10.1038/s41467-024-54155-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Post-translational cycles of α-tubulin detyrosination and tyrosination generate microtubule diversity, the cellular functions of which remain largely unknown. Here we show that α-tubulin detyrosination regulates kinetochore-microtubule attachments to ensure normal chromosome oscillations and timely anaphase onset during mitosis. Remarkably, detyrosinated α-tubulin levels near kinetochore microtubule plus-ends depend on the direction of chromosome motion during metaphase. Proteomic analyses unveil that the KNL-1/MIS12/NDC80 (KMN) network that forms the core microtubule-binding site at kinetochores and the microtubule-rescue protein CLASP2 are enriched on tyrosinated and detyrosinated microtubules during mitosis, respectively. α-tubulin detyrosination enhances CLASP2 binding and NDC80 complex diffusion along the microtubule lattice in vitro. Rescue experiments overexpressing NDC80, including variants with slower microtubule diffusion, suggest a functional interplay with α-tubulin detyrosination for the establishment of a labile kinetochore-microtubule interface. These results offer a mechanistic explanation for how different detyrosinated α-tubulin levels near kinetochore microtubule plus-ends fine-tune load-bearing attachments to both growing and shrinking microtubules.
Collapse
Affiliation(s)
- Hugo Girão
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- IBMC-Institute for Molecular and Cell Biology, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Joana Macário-Monteiro
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- IBMC-Institute for Molecular and Cell Biology, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Ana C Figueiredo
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- IBMC-Institute for Molecular and Cell Biology, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Ricardo Silva E Sousa
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- IBMC-Institute for Molecular and Cell Biology, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Elena Doria
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Geneva, Switzerland
- Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Geneva, Switzerland
| | - Vladimir Demidov
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hugo Osório
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, 4200-135, Porto, Portugal
| | - Ariana Jacome
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- IBMC-Institute for Molecular and Cell Biology, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Geneva, Switzerland
- Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Geneva, Switzerland
| | - Ekaterina L Grishchuk
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Helder Maiato
- i3S-Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.
- IBMC-Institute for Molecular and Cell Biology, University of Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.
- Cell Division Group, Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
2
|
Bak J, Brummelkamp TR, Perrakis A. Decoding microtubule detyrosination: enzyme families, structures, and functional implications. FEBS Lett 2024; 598:1453-1464. [PMID: 38811347 DOI: 10.1002/1873-3468.14940] [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: 03/04/2024] [Revised: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024]
Abstract
Microtubules are a major component of the cytoskeleton and can accumulate a plethora of modifications. The microtubule detyrosination cycle is one of these modifications; it involves the enzymatic removal of the C-terminal tyrosine of α-tubulin on assembled microtubules and the re-ligation of tyrosine on detyrosinated tubulin dimers. This modification cycle has been implicated in cardiac disease, neuronal development, and mitotic defects. The vasohibin and microtubule-associated tyrosine carboxypeptidase enzyme families are responsible for microtubule detyrosination. Their long-sought discovery allows to review and summarise differences and similarities between the two enzymes families and discuss how they interplay with other modifications and functions of the tubulin code.
Collapse
Affiliation(s)
- Jitske Bak
- Oncode Institute and Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thijn R Brummelkamp
- Oncode Institute and Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Anastassis Perrakis
- Oncode Institute and Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Viar GA, Pigino G. Tubulin posttranslational modifications through the lens of new technologies. Curr Opin Cell Biol 2024; 88:102362. [PMID: 38701611 DOI: 10.1016/j.ceb.2024.102362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024]
Abstract
The Tubulin Code revolutionizes our understanding of microtubule dynamics and functions, proposing a nuanced system governed by tubulin isotypes, posttranslational modifications (PTMs) and microtubule-associated proteins (MAPs). Tubulin isotypes, diverse across species, contribute structural complexity, and are thought to influence microtubule functions. PTMs encode dynamic information on microtubules, which are read by several microtubule interacting proteins and impact on cellular processes. Here we discuss recent technological and methodological advances, such as in genome engineering, live cell imaging, expansion microscopy, and cryo-electron microscopy that reveal new elements and levels of complexity of the tubulin code, including new modifying enzymes and nanopatterns of PTMs on individual microtubules. The Tubulin Code's exploration holds transformative potential, guiding therapeutic strategies and illuminating connections to diseases like cancer and neurodegenerative disorders, underscoring its relevance in decoding fundamental cellular language.
Collapse
Affiliation(s)
| | - Gaia Pigino
- Human Technopole, via Rita Levi Montalcini 1, Milan, Italy.
| |
Collapse
|
4
|
Nasilli G, de Waal TM, Marchal GA, Bertoli G, Veldkamp MW, Rothenberg E, Casini S, Remme CA. Decreasing microtubule detyrosination modulates Nav1.5 subcellular distribution and restores sodium current in mdx cardiomyocytes. Cardiovasc Res 2024; 120:723-734. [PMID: 38395031 PMCID: PMC11135645 DOI: 10.1093/cvr/cvae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/28/2023] [Accepted: 01/08/2024] [Indexed: 02/25/2024] Open
Abstract
AIMS The microtubule (MT) network plays a major role in the transport of the cardiac sodium channel Nav1.5 to the membrane, where the latter associates with interacting proteins such as dystrophin. Alterations in MT dynamics are known to impact on ion channel trafficking. Duchenne muscular dystrophy (DMD), caused by dystrophin deficiency, is associated with an increase in MT detyrosination, decreased sodium current (INa), and arrhythmias. Parthenolide (PTL), a compound that decreases MT detyrosination, has shown beneficial effects on cardiac function in DMD. We here investigated its impact on INa and Nav1.5 subcellular distribution. METHODS AND RESULTS Ventricular cardiomyocytes (CMs) from wild-type (WT) and mdx (DMD) mice were incubated with either 10 µM PTL, 20 µM EpoY, or dimethylsulfoxide (DMSO) for 3-5 h, followed by patch-clamp analysis to assess INa and action potential (AP) characteristics in addition to immunofluorescence and stochastic optical reconstruction microscopy (STORM) to investigate MT detyrosination and Nav1.5 cluster size and density, respectively. In accordance with previous studies, we observed increased MT detyrosination, decreased INa and reduced AP upstroke velocity (Vmax) in mdx CMs compared to WT. PTL decreased MT detyrosination and significantly increased INa magnitude (without affecting INa gating properties) and AP Vmax in mdx CMs, but had no effect in WT CMs. Moreover, STORM analysis showed that in mdx CMs, Nav1.5 clusters were decreased not only in the grooves of the lateral membrane (LM; where dystrophin is localized) but also at the LM crests. PTL restored Nav1.5 clusters at the LM crests (but not at the grooves), indicating a dystrophin-independent trafficking route to this subcellular domain. Interestingly, Nav1.5 cluster density was also reduced at the intercalated disc (ID) region of mdx CMs, which was restored to WT levels by PTL. Treatment of mdx CMs with EpoY, a specific MT detyrosination inhibitor, also increased INa density, while decreasing the amount of detyrosinated MTs, confirming a direct mechanistic link. CONCLUSION Attenuating MT detyrosination in mdx CMs restored INa and enhanced Nav1.5 localization at the LM crest and ID. Hence, the reduced whole-cell INa density characteristic of mdx CMs is not only the consequence of the lack of dystrophin within the LM grooves but is also due to reduced Nav1.5 at the LM crest and ID secondary to increased baseline MT detyrosination. Overall, our findings identify MT detyrosination as a potential therapeutic target for modulating INa and subcellular Nav1.5 distribution in pathophysiological conditions.
Collapse
Affiliation(s)
- Giovanna Nasilli
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Division of Cardiology, NYU Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Tanja M de Waal
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Giorgia Bertoli
- Division of Cardiology, NYU Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Marieke W Veldkamp
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Eli Rothenberg
- Department of Biochemistry and Pharmacology, NYU Grossman School of Medicine, 450 E 29TH ST Alexandria Center for Life Science, New York, NY 10016, USA
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam University Medical Center, University of Amsterdam, Heart Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Liutkute A, Prosser BL, Voigt N. Microtubules: highway to … arrhythmia? Cardiovasc Res 2024; 120:671-672. [PMID: 38637305 DOI: 10.1093/cvr/cvae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Affiliation(s)
- Aiste Liutkute
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Germany
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Germany
| |
Collapse
|
6
|
Konietzny A, Han Y, Popp Y, van Bommel B, Sharma A, Delagrange P, Arbez N, Moutin MJ, Peris L, Mikhaylova M. Efficient axonal transport of endolysosomes relies on the balanced ratio of microtubule tyrosination and detyrosination. J Cell Sci 2024; 137:jcs261737. [PMID: 38525600 PMCID: PMC11112122 DOI: 10.1242/jcs.261737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
In neurons, the microtubule (MT) cytoskeleton forms the basis for long-distance protein transport from the cell body into and out of dendrites and axons. To maintain neuronal polarity, the axon initial segment (AIS) serves as a physical barrier, separating the axon from the somatodendritic compartment and acting as a filter for axonal cargo. Selective trafficking is further instructed by axonal enrichment of MT post-translational modifications, which affect MT dynamics and the activity of motor proteins. Here, we compared two knockout mouse lines lacking the respective enzymes for MT tyrosination and detyrosination, and found that both knockouts led to a shortening of the AIS. Neurons from both lines also showed an increased immobile fraction of endolysosomes present in the axon, whereas mobile organelles displayed shortened run distances in the retrograde direction. Overall, our results highlight the importance of maintaining the balance of tyrosinated and detyrosinated MTs for proper AIS length and axonal transport processes.
Collapse
Affiliation(s)
- Anja Konietzny
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan
| | - Yuhao Han
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Centre for Structural Systems Biology, Hamburg 22607, Germany
- Structural Cell Biology of Viruses, Leibniz Institute of Virology (LIV), Hamburg 20251, Germany
| | - Yannes Popp
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Charité – Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany
| | - Bas van Bommel
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin 14195, Germany
| | - Aditi Sharma
- University Grenoble Alpes, Inserm U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | | | - Nicolas Arbez
- Institut de Recherche Servier, Croissy 78290, France
| | - Marie-Jo Moutin
- University Grenoble Alpes, Inserm U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Leticia Peris
- University Grenoble Alpes, Inserm U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Marina Mikhaylova
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin 10115, Germany
- Guest Group ‘Neuronal Protein Transport’, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| |
Collapse
|
7
|
Bonet-Ponce L, Tegicho T, Beilina A, Kluss JH, Li Y, Cookson MR. Opposing actions of JIP4 and RILPL1 provide antagonistic motor force to dynamically regulate membrane reformation during lysosomal tubulation/sorting driven by LRRK2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587808. [PMID: 38903076 PMCID: PMC11188082 DOI: 10.1101/2024.04.02.587808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Lysosomes are dynamic cellular structures that adaptively remodel their membrane in response to stimuli, including membrane damage. We previously uncovered a process we term LYTL (LYsosomal Tubulation/sorting driven by Leucine-Rich Repeat Kinase 2 [LRRK2]), wherein damaged lysosomes generate tubules sorted into mobile vesicles. LYTL is orchestrated by the Parkinson's disease-associated kinase LRRK2 that recruits the motor adaptor protein and RHD family member JIP4 to lysosomes via phosphorylated RAB proteins. To identify new players involved in LYTL, we performed unbiased proteomics on isolated lysosomes after LRRK2 kinase inhibition. Our results demonstrate that there is recruitment of RILPL1 to ruptured lysosomes via LRRK2 activity to promote phosphorylation of RAB proteins at the lysosomal surface. RILPL1, which is also a member of the RHD family, enhances the clustering of LRRK2-positive lysosomes in the perinuclear area and causes retraction of LYTL tubules, in contrast to JIP4 which promotes LYTL tubule extension. Mechanistically, RILPL1 binds to p150Glued, a dynactin subunit, facilitating the transport of lysosomes and tubules to the minus end of microtubules. Further characterization of the tubulation process revealed that LYTL tubules move along tyrosinated microtubules, with tubulin tyrosination proving essential for tubule elongation. In summary, our findings emphasize the dynamic regulation of LYTL tubules by two distinct RHD proteins and pRAB effectors, serving as opposing motor adaptor proteins: JIP4, promoting tubulation via kinesin, and RILPL1, facilitating tubule retraction through dynein/dynactin. We infer that the two opposing processes generate a metastable lysosomal membrane deformation that facilitates dynamic tubulation events.
Collapse
Affiliation(s)
- Luis Bonet-Ponce
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, Ohio, 43210, USA
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Tsion Tegicho
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Alexandra Beilina
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Jillian H. Kluss
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Yan Li
- Proteomic Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892, USA
| |
Collapse
|
8
|
Pietsch N, Chen CY, Kupsch S, Bacmeister L, Geertz B, Herera-Rivero M, Voß H, Krämer E, Braren I, Westermann D, Schlüter H, Mearini G, Schlossarek S, van der Velden J, Caporizzo MA, Lindner D, Prosser BL, Carrier L. Chronic activation of tubulin tyrosination in HCM mice and human iPSC-engineered heart tissues improves heart function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.25.542365. [PMID: 37292763 PMCID: PMC10245930 DOI: 10.1101/2023.05.25.542365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Rationale: Hypertrophic cardiomyopathy (HCM) is the most common cardiac genetic disorder caused by sarcomeric gene variants and associated with left ventricular (LV) hypertrophy and diastolic dysfunction. The role of the microtubule network has recently gained interest with the findings that -α-tubulin detyrosination (dTyr-tub) is markedly elevated in heart failure. Acute reduction of dTyr-tub by inhibition of the detyrosinase (VASH/SVBP complex) or activation of the tyrosinase (tubulin tyrosine ligase, TTL) markedly improved contractility and reduced stiffness in human failing cardiomyocytes, and thus poses a new perspective for HCM treatment. Objective: In this study, we tested the impact of chronic tubulin tyrosination in a HCM mouse model ( Mybpc3 -knock-in; KI), in human HCM cardiomyocytes and in SVBP-deficient human engineered heart tissues (EHTs). Methods and Results: AAV9-mediated TTL transfer was applied in neonatal wild-type (WT) rodents and 3-week-old KI mice and in HCM human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes. We show that i) TTL for 6 weeks dose-dependently reduced dTyr-tub and improved contractility without affecting cytosolic calcium transients in WT cardiomyocytes; ii) TTL for 12 weeks improved diastolic filling, cardiac output and stroke volume and reduced stiffness in KI mice; iii) TTL for 10 days normalized cell hypertrophy in HCM hiPSC-cardiomyocytes; iv) TTL induced a marked transcription and translation of several tubulins and modulated mRNA or protein levels of components of mitochondria, Z-disc, ribosome, intercalated disc, lysosome and cytoskeleton in KI mice; v) SVBP-deficient EHTs exhibited reduced dTyr-tub levels, higher force and faster relaxation than TTL-deficient and WT EHTs. RNA-seq and mass spectrometry analysis revealed distinct enrichment of cardiomyocyte components and pathways in SVBP-KO vs. TTL-KO EHTs. Conclusion: This study provides the first proof-of-concept that chronic activation of tubulin tyrosination in HCM mice and in human EHTs improves heart function and holds promise for targeting the non-sarcomeric cytoskeleton in heart disease.
Collapse
|
9
|
Tang Q, Sensale S, Bond C, Xing J, Qiao A, Hugelier S, Arab A, Arya G, Lakadamyali M. Interplay between stochastic enzyme activity and microtubule stability drives detyrosination enrichment on microtubule subsets. Curr Biol 2023; 33:5169-5184.e8. [PMID: 37979580 PMCID: PMC10843832 DOI: 10.1016/j.cub.2023.10.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/03/2023] [Accepted: 10/30/2023] [Indexed: 11/20/2023]
Abstract
Microtubules in cells consist of functionally diverse subpopulations carrying distinct post-translational modifications (PTMs). Akin to the histone code, the tubulin code regulates a myriad of microtubule functions, ranging from intracellular transport to chromosome segregation. However, how individual PTMs only occur on subsets of microtubules to contribute to microtubule specialization is not well understood. In particular, microtubule detyrosination, the removal of the C-terminal tyrosine on α-tubulin subunits, marks the stable population of microtubules and modifies how microtubules interact with other microtubule-associated proteins to regulate a wide range of cellular processes. Previously, we found that in certain cell types, only ∼30% of microtubules are highly enriched with the detyrosination mark and that detyrosination spans most of the length of a microtubule, often adjacent to a completely tyrosinated microtubule. How the activity of a cytosolic detyrosinase, vasohibin (VASH), leads to only a small subpopulation of highly detyrosinated microtubules is unclear. Here, using quantitative super-resolution microscopy, we visualized nascent microtubule detyrosination events in cells consisting of 1-3 detyrosinated α-tubulin subunits after nocodazole washout. Microtubule detyrosination accumulates slowly and in a dispersed pattern across the microtubule length. By visualizing single molecules of VASH in live cells, we found that VASH engages with microtubules stochastically on a short timescale, suggesting limited removal of tyrosine per interaction, consistent with the super-resolution results. Combining these quantitative imaging results with simulations incorporating parameters from our experiments, we provide evidence for a stochastic model for cells to establish a subset of detyrosinated microtubules via a detyrosination-stabilization feedback mechanism.
Collapse
Affiliation(s)
- Qing Tang
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sebastian Sensale
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA; Department of Physics, Cleveland State University, Cleveland, OH 44115-2214, USA.
| | - Charles Bond
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiazheng Xing
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andy Qiao
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Siewert Hugelier
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arian Arab
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gaurav Arya
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Melike Lakadamyali
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Cason SE, Holzbaur EL. Axonal transport of autophagosomes is regulated by dynein activators JIP3/JIP4 and ARF/RAB GTPases. J Cell Biol 2023; 222:e202301084. [PMID: 37909920 PMCID: PMC10620608 DOI: 10.1083/jcb.202301084] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/28/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Neuronal autophagosomes form and engulf cargos at presynaptic sites in the axon and are then transported to the soma to recycle their cargo. Autophagic vacuoles (AVs) mature en route via fusion with lysosomes to become degradatively competent organelles; transport is driven by the microtubule motor protein cytoplasmic dynein, with motor activity regulated by a sequential series of adaptors. Using lysate-based single-molecule motility assays and live-cell imaging in primary neurons, we show that JNK-interacting proteins 3 (JIP3) and 4 (JIP4) are activating adaptors for dynein that are regulated on autophagosomes and lysosomes by the small GTPases ARF6 and RAB10. GTP-bound ARF6 promotes formation of the JIP3/4-dynein-dynactin complex. Either knockdown or overexpression of RAB10 stalls transport, suggesting that this GTPase is also required to coordinate the opposing activities of bound dynein and kinesin motors. These findings highlight the complex coordination of motor regulation during organelle transport in neurons.
Collapse
Affiliation(s)
- Sydney E. Cason
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Erika L.F. Holzbaur
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Zhong Y, Wang C, Wang Y, Wu Y, Wang H, Qiu S, Hao M, Wang Z, Wang X, Jin H, Zhou J. Suppression of alpha-tubulin acetylation potentiates therapeutic efficacy of Eribulin in liver cancer. Am J Cancer Res 2023; 13:5698-5718. [PMID: 38058833 PMCID: PMC10695797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/25/2023] [Indexed: 12/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent cancer with limited effective treatments. Eribulin mesylate is a novel chemotherapy drug that inhibits microtubule elongation and may impact the tumor microenvironment and immune pathway. This study aims to investigate the impact of changes in microtubule acetylation levels on HCC development and treatment outcomes. Clinical and molecular data were aggregated from databases, with survival analysis conducted to evaluate the relevance of microtubule acetylation. In vitro experiments using HCC cell lines and a tumor cell transplantation model in C57BL/c mice were performed to investigate the effects of microtubule acetylation on Eribulin treatment. A significant correlation was found between the level of lysine 40 acetylation of α-tubulin (acetyl-α-tubulin-lys40) and overall survival of HCC patients, with a better prognosis associated with a lower level of acetyl-α-tubulin-lys40. Knocking down ATAT1 or overexpressing HDAC6 reduced the level of acetyl-α-tubulin-lys40 and sensitized Eribulin treatment both in vitro and in vivo. In summary, acetyl-α-tubulin-lys40 was increased in HCC and was associated with a shorter overall survival of HCC patients. Reducing the level of acetyl-α-tubulin-lys40 can enhance sensitivity to Eribulin treatment both in vitro and in vivo, thereby establishing acetyl-α-tubulin-lys40 as a potential prognostic marker and predictive indicator for Eribulin treatment in HCC patients.
Collapse
Affiliation(s)
- Yiming Zhong
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang Province, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Chaoqun Wang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical UniversityDongyang 322100, Zhejiang, China
| | - Yanmei Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Yan Wu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang Province, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Hanying Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Shuying Qiu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Minyan Hao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Zhuo Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang Province, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| | - Jia Zhou
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang Province, Cancer Center of Zhejiang University, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou 310016, Zhejiang, China
| |
Collapse
|
12
|
McKenna ED, Sarbanes SL, Cummings SW, Roll-Mecak A. The Tubulin Code, from Molecules to Health and Disease. Annu Rev Cell Dev Biol 2023; 39:331-361. [PMID: 37843925 DOI: 10.1146/annurev-cellbio-030123-032748] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Microtubules are essential dynamic polymers composed of α/β-tubulin heterodimers. They support intracellular trafficking, cell division, cellular motility, and other essential cellular processes. In many species, both α-tubulin and β-tubulin are encoded by multiple genes with distinct expression profiles and functionality. Microtubules are further diversified through abundant posttranslational modifications, which are added and removed by a suite of enzymes to form complex, stereotyped cellular arrays. The genetic and chemical diversity of tubulin constitute a tubulin code that regulates intrinsic microtubule properties and is read by cellular effectors, such as molecular motors and microtubule-associated proteins, to provide spatial and temporal specificity to microtubules in cells. In this review, we synthesize the rapidly expanding tubulin code literature and highlight limitations and opportunities for the field. As complex microtubule arrays underlie essential physiological processes, a better understanding of how cells employ the tubulin code has important implications for human disease ranging from cancer to neurological disorders.
Collapse
Affiliation(s)
- Elizabeth D McKenna
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Stephanie L Sarbanes
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Steven W Cummings
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Nicot S, Gillard G, Impheng H, Joachimiak E, Urbach S, Mochizuki K, Wloga D, Juge F, Rogowski K. A family of carboxypeptidases catalyzing α- and β-tubulin tail processing and deglutamylation. SCIENCE ADVANCES 2023; 9:eadi7838. [PMID: 37703372 PMCID: PMC10499314 DOI: 10.1126/sciadv.adi7838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
Tubulin posttranslational modifications represent an important mechanism involved in the regulation of microtubule functions. The most widespread among them are detyrosination, α∆2-tubulin, and polyglutamylation. Here, we describe a family of tubulin-modifying enzymes composed of two closely related proteins, KIAA0895L and KIAA0895, which have tubulin metallocarboxypeptidase activity and thus were termed TMCP1 and TMCP2, respectively. We show that TMCP1 (also known as MATCAP) acts as α-tubulin detyrosinase that also catalyzes α∆2-tubulin. In contrast, TMCP2 preferentially modifies βI-tubulin by removing three amino acids from its C terminus, generating previously unknown βI∆3 modification. We show that βI∆3-tubulin is mostly found on centrioles and mitotic spindles and in cilia. Moreover, we demonstrate that TMCPs also remove posttranslational polyglutamylation and thus act as tubulin deglutamylases. Together, our study describes the identification and comprehensive biochemical analysis of a previously unknown type of tubulin-modifying enzymes involved in the processing of α- and β-tubulin C-terminal tails and deglutamylation.
Collapse
Affiliation(s)
- Simon Nicot
- Tubulin Code team, Institute of Human Genetics, Université Montpellier, CNRS, Montpellier, France
| | - Ghislain Gillard
- Tubulin Code team, Institute of Human Genetics, Université Montpellier, CNRS, Montpellier, France
| | - Hathaichanok Impheng
- Department of Physiology, Faculty of Medical science, Naresuan University, Phitsanulok 65000, Thailand
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Serge Urbach
- Functional Proteomics Platform (FPP), IGF, Université Montpellier, CNRS, INSERM, Montpellier, France
| | - Kazufumi Mochizuki
- Epigenetic Chromatin Regulation team, Institute of Human Genetics, Université Montpellier, CNRS, Montpellier, France
| | - Dorota Wloga
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - François Juge
- Tubulin Code team, Institute of Human Genetics, Université Montpellier, CNRS, Montpellier, France
| | - Krzysztof Rogowski
- Tubulin Code team, Institute of Human Genetics, Université Montpellier, CNRS, Montpellier, France
| |
Collapse
|
14
|
Iwanski MK, Kapitein LC. Cellular cartography: Towards an atlas of the neuronal microtubule cytoskeleton. Front Cell Dev Biol 2023; 11:1052245. [PMID: 37035244 PMCID: PMC10073685 DOI: 10.3389/fcell.2023.1052245] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Microtubules, one of the major components of the cytoskeleton, play a crucial role during many aspects of neuronal development and function, such as neuronal polarization and axon outgrowth. Consequently, the microtubule cytoskeleton has been implicated in many neurodevelopmental and neurodegenerative disorders. The polar nature of microtubules is quintessential for their function, allowing them to serve as tracks for long-distance, directed intracellular transport by kinesin and dynein motors. Most of these motors move exclusively towards either the plus- or minus-end of a microtubule and some have been shown to have a preference for either dynamic or stable microtubules, those bearing a particular post-translational modification or those decorated by a specific microtubule-associated protein. Thus, it becomes important to consider the interplay of these features and their combinatorial effects on transport, as well as how different types of microtubules are organized in the cell. Here, we discuss microtubule subsets in terms of tubulin isotypes, tubulin post-translational modifications, microtubule-associated proteins, microtubule stability or dynamicity, and microtubule orientation. We highlight techniques used to study these features of the microtubule cytoskeleton and, using the information from these studies, try to define the composition, role, and organization of some of these subsets in neurons.
Collapse
Affiliation(s)
| | - Lukas C. Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
15
|
Akera T. Tubulin post-translational modifications in meiosis. Semin Cell Dev Biol 2023; 137:38-45. [PMID: 34836784 PMCID: PMC9124733 DOI: 10.1016/j.semcdb.2021.11.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/22/2021] [Accepted: 11/14/2021] [Indexed: 11/18/2022]
Abstract
Haploid gametes are produced from diploid parents through meiosis, a process inherent to all sexually reproducing eukaryotes. Faithful chromosome segregation in meiosis is essential for reproductive success, although it is less clear how the meiotic spindle achieves this compared to the mitotic spindle. It is becoming increasingly clear that tubulin post-translational modifications (PTMs) play critical roles in regulating microtubule functions in many biological processes, and meiosis is no exception. Here, I review recent advances in the understanding of tubulin PTMs in meiotic spindles, especially focusing on their roles in spindle integrity, oocyte aging, and non-Mendelian transmission.
Collapse
Affiliation(s)
- Takashi Akera
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda 20892, MD, USA.
| |
Collapse
|
16
|
Zocchi R, Compagnucci C, Bertini E, Sferra A. Deciphering the Tubulin Language: Molecular Determinants and Readout Mechanisms of the Tubulin Code in Neurons. Int J Mol Sci 2023; 24:ijms24032781. [PMID: 36769099 PMCID: PMC9917122 DOI: 10.3390/ijms24032781] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Microtubules (MTs) are dynamic components of the cell cytoskeleton involved in several cellular functions, such as structural support, migration and intracellular trafficking. Despite their high similarity, MTs have functional heterogeneity that is generated by the incorporation into the MT lattice of different tubulin gene products and by their post-translational modifications (PTMs). Such regulations, besides modulating the tubulin composition of MTs, create on their surface a "biochemical code" that is translated, through the action of protein effectors, into specific MT-based functions. This code, known as "tubulin code", plays an important role in neuronal cells, whose highly specialized morphologies and activities depend on the correct functioning of the MT cytoskeleton and on its interplay with a myriad of MT-interacting proteins. In recent years, a growing number of mutations in genes encoding for tubulins, MT-interacting proteins and enzymes that post-translationally modify MTs, which are the main players of the tubulin code, have been linked to neurodegenerative processes or abnormalities in neural migration, differentiation and connectivity. Nevertheless, the exact molecular mechanisms through which the cell writes and, downstream, MT-interacting proteins decipher the tubulin code are still largely uncharted. The purpose of this review is to describe the molecular determinants and the readout mechanisms of the tubulin code, and briefly elucidate how they coordinate MT behavior during critical neuronal events, such as neuron migration, maturation and axonal transport.
Collapse
Affiliation(s)
- Riccardo Zocchi
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| | - Claudia Compagnucci
- Molecular Genetics and Functional Genomics, Bambino Gesù Children’s Research Hospital, IRCCS, 00146 Rome, Italy
| | - Enrico Bertini
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence: (E.B.); or (A.S.); Tel.: +39-06-6859-2104 (E.B. & A.S.)
| | - Antonella Sferra
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
- Correspondence: (E.B.); or (A.S.); Tel.: +39-06-6859-2104 (E.B. & A.S.)
| |
Collapse
|
17
|
Pero ME, Chowdhury F, Bartolini F. Role of tubulin post-translational modifications in peripheral neuropathy. Exp Neurol 2023; 360:114274. [PMID: 36379274 PMCID: PMC11320756 DOI: 10.1016/j.expneurol.2022.114274] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Peripheral neuropathy is a common disorder that results from nerve damage in the periphery. The degeneration of sensory axon terminals leads to changes or loss of sensory functions, often manifesting as debilitating pain, weakness, numbness, tingling, and disability. The pathogenesis of most peripheral neuropathies remains to be fully elucidated. Cumulative evidence from both early and recent studies indicates that tubulin damage may provide a common underlying mechanism of axonal injury in various peripheral neuropathies. In particular, tubulin post-translational modifications have been recently implicated in both toxic and inherited forms of peripheral neuropathy through regulation of axonal transport and mitochondria dynamics. This knowledge forms a new area of investigation with the potential for developing therapeutic strategies to prevent or delay peripheral neuropathy by restoring tubulin homeostasis.
Collapse
Affiliation(s)
- Maria Elena Pero
- Department of Pathology and Cell Biology, Columbia University, New York, USA; Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Italy
| | - Farihah Chowdhury
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, USA.
| |
Collapse
|
18
|
Cason SE, Holzbaur EL. Axonal transport of autophagosomes is regulated by dynein activators JIP3/JIP4 and ARF/RAB GTPases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.28.526044. [PMID: 36747648 PMCID: PMC9901177 DOI: 10.1101/2023.01.28.526044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Neuronal autophagosomes, "self-eating" degradative organelles, form at presynaptic sites in the distal axon and are transported to the soma to recycle their cargo. During transit, autophagic vacuoles (AVs) mature through fusion with lysosomes to acquire the enzymes necessary to breakdown their cargo. AV transport is driven primarily by the microtubule motor cytoplasmic dynein in concert with dynactin and a series of activating adaptors that change depending on organelle maturation state. The transport of mature AVs is regulated by the scaffolding proteins JIP3 and JIP4, both of which activate dynein motility in vitro. AV transport is also regulated by ARF6 in a GTP-dependent fashion. While GTP-bound ARF6 promotes the formation of the JIP3/4-dynein-dynactin complex, RAB10 competes with the activity of this complex by increasing kinesin recruitment to axonal AVs and lysosomes. These interactions highlight the complex coordination of motors regulating organelle transport in neurons.
Collapse
Affiliation(s)
- Sydney E. Cason
- Department of Physiology, University of Pennsylvania
- Neuroscience Graduate Group, University of Pennsylvania
- Pennsylvania Muscle Institute, University of Pennsylvania
| | - Erika L.F. Holzbaur
- Department of Physiology, University of Pennsylvania
- Neuroscience Graduate Group, University of Pennsylvania
- Pennsylvania Muscle Institute, University of Pennsylvania
| |
Collapse
|
19
|
Ramirez-Rios S, Choi SR, Sanyal C, Blum TB, Bosc C, Krichen F, Denarier E, Soleilhac JM, Blot B, Janke C, Stoppin-Mellet V, Magiera MM, Arnal I, Steinmetz MO, Moutin MJ. VASH1-SVBP and VASH2-SVBP generate different detyrosination profiles on microtubules. J Cell Biol 2022; 222:213744. [PMID: 36512346 PMCID: PMC9750192 DOI: 10.1083/jcb.202205096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022] Open
Abstract
The detyrosination/tyrosination cycle of α-tubulin is critical for proper cell functioning. VASH1-SVBP and VASH2-SVBP are ubiquitous enzymes involved in microtubule detyrosination, whose mode of action is little known. Here, we show in reconstituted systems and cells that VASH1-SVBP and VASH2-SVBP drive the global and local detyrosination of microtubules, respectively. We solved the cryo-electron microscopy structure of VASH2-SVBP bound to microtubules, revealing a different microtubule-binding configuration of its central catalytic region compared to VASH1-SVBP. We show that the divergent mode of detyrosination between the two enzymes is correlated with the microtubule-binding properties of their disordered N- and C-terminal regions. Specifically, the N-terminal region is responsible for a significantly longer residence time of VASH2-SVBP on microtubules compared to VASH1-SVBP. We suggest that this VASH region is critical for microtubule detachment and diffusion of VASH-SVBP enzymes on lattices. Our results suggest a mechanism by which VASH1-SVBP and VASH2-SVBP could generate distinct microtubule subpopulations and confined areas of detyrosinated lattices to drive various microtubule-based cellular functions.
Collapse
Affiliation(s)
- Sacnicte Ramirez-Rios
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Sung Ryul Choi
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Chadni Sanyal
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Thorsten B. Blum
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Christophe Bosc
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Fatma Krichen
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Eric Denarier
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Jean-Marc Soleilhac
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Béatrice Blot
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Carsten Janke
- Institut Curie, Université Paris Sciences et Lettres, Centre National de la Recherche Scientifique UMR3348, Orsay, France,Université Paris-Saclay, Centre National de la Recherche Scientifique UMR3348, Orsay, France
| | - Virginie Stoppin-Mellet
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Maria M. Magiera
- Institut Curie, Université Paris Sciences et Lettres, Centre National de la Recherche Scientifique UMR3348, Orsay, France,Université Paris-Saclay, Centre National de la Recherche Scientifique UMR3348, Orsay, France
| | - Isabelle Arnal
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Michel O. Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland,Biozentrum, University of Basel, Basel, Switzerland
| | - Marie-Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France,Correspondence to Marie-Jo Moutin:
| |
Collapse
|
20
|
Szczesna E, Zehr EA, Cummings SW, Szyk A, Mahalingan KK, Li Y, Roll-Mecak A. Combinatorial and antagonistic effects of tubulin glutamylation and glycylation on katanin microtubule severing. Dev Cell 2022; 57:2497-2513.e6. [PMID: 36347241 PMCID: PMC9665884 DOI: 10.1016/j.devcel.2022.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/17/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
Microtubules have spatiotemporally complex posttranslational modification patterns. How cells interpret this tubulin modification code is largely unknown. We show that C. elegans katanin, a microtubule severing AAA ATPase mutated in microcephaly and critical for cell division, axonal elongation, and cilia biogenesis, responds precisely, differentially, and combinatorially to three chemically distinct tubulin modifications-glycylation, glutamylation, and tyrosination-but is insensitive to acetylation. Glutamylation and glycylation are antagonistic rheostats with glycylation protecting microtubules from severing. Katanin exhibits graded and divergent responses to glutamylation on the α- and β-tubulin tails, and these act combinatorially. The katanin hexamer central pore constrains the polyglutamate chain patterns on β-tails recognized productively. Elements distal to the katanin AAA core sense α-tubulin tyrosination, and detyrosination downregulates severing. The multivalent microtubule recognition that enables katanin to read multiple tubulin modification inputs explains in vivo observations and illustrates how effectors can integrate tubulin code signals to produce diverse functional outcomes.
Collapse
Affiliation(s)
- Ewa Szczesna
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Elena A Zehr
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Steven W Cummings
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Agnieszka Szyk
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Kishore K Mahalingan
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Yan Li
- Proteomic Core Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA; Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
21
|
Hauguel C, Ducellier S, Provot O, Ibrahim N, Lamaa D, Balcerowiak C, Letribot B, Nascimento M, Blanchard V, Askenatzis L, Levaique H, Bignon J, Baschieri F, Bauvais C, Bollot G, Renko D, Deroussent A, Prost B, Laisne MC, Michallet S, Lafanechère L, Papot S, Montagnac G, Tran C, Alami M, Apcher S, Hamze A. Design, synthesis and biological evaluation of quinoline-2-carbonitrile-based hydroxamic acids as dual tubulin polymerization and histone deacetylases inhibitors. Eur J Med Chem 2022; 240:114573. [DOI: 10.1016/j.ejmech.2022.114573] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/04/2022]
|
22
|
Lafanechère L. The microtubule cytoskeleton: An old validated target for novel therapeutic drugs. Front Pharmacol 2022; 13:969183. [PMID: 36188585 PMCID: PMC9521402 DOI: 10.3389/fphar.2022.969183] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Compounds targeting microtubules are widely used in cancer therapy with a proven efficacy. However, because they also target non-cancerous cells, their administration leads to numerous adverse effects. With the advancement of knowledge on the structure of tubulin, the regulation of microtubule dynamics and their deregulation in pathological processes, new therapeutic strategies are emerging, both for the treatment of cancer and for other diseases, such as neuronal or even heart diseases and parasite infections. In addition, a better understanding of the mechanism of action of well-known drugs such as colchicine or certain kinase inhibitors contributes to the development of these new therapeutic approaches. Nowadays, chemists and biologists are working jointly to select drugs which target the microtubule cytoskeleton and have improved properties. On the basis of a few examples this review attempts to depict the panorama of these recent advances.
Collapse
|
23
|
Fourest-Lieuvin A, Vinit A, Blot B, Perrot A, Denarier E, Saudou F, Arnal I. Controlled Tau Cleavage in Cells Reveals Abnormal Localizations of Tau Fragments. Neuroscience 2022; 518:162-177. [PMID: 35995336 DOI: 10.1016/j.neuroscience.2022.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/08/2022] [Accepted: 08/16/2022] [Indexed: 11/15/2022]
Abstract
In several forms of dementia, such as Alzheimer's disease, the cytoskeleton-associated protein tau undergoes proteolysis, giving rise to fragments that have a toxic impact on neuronal homeostasis. How these fragments interact with cellular structures, in particular with the cytoskeleton, is currently incompletely understood. Here, we developed a method, derived from a Tobacco Etch Virus (TEV) protease system, to induce controlled cleavage of tau at specific sites. Five tau proteins containing specific TEV recognition sites corresponding to pathological proteolytic sites were engineered, and tagged with GFP at one end and mCherry at the other. Following controlled cleavage to produce GFP-N-terminal and C-terminal-mCherry fragments, we followed the fate of tau fragments in cells. Our results showed that whole engineered tau proteins associate with the cytoskeleton similarly to the non-modified tau, whereas tau fragments adopted different localizations with respect to the actin and microtubule cytoskeletons. These distinct localizations were confirmed by expressing each separate fragment in cells. Some cleavages - in particular cleavages at amino-acid positions 124 or 256 - displayed a certain level of cellular toxicity, with an unusual relocalization of the N-terminal fragments to the nucleus. Based on the data presented here, inducible cleavage of tau by the TEV protease appears to be a valuable tool to reproduce tau fragmentation in cells and study the resulting consequences on cell physiology.
Collapse
Affiliation(s)
- Anne Fourest-Lieuvin
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| | - Angélique Vinit
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Béatrice Blot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Anthime Perrot
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eric Denarier
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frédéric Saudou
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Isabelle Arnal
- Université Grenoble Alpes, INSERM, U1216, CEA, CNRS, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
24
|
Isoda R, Morita I, Isida A, Mikami Y, Monobe Y, Sato Y, Moriya T. Pathological Study on the Expression of Vasohibins in Peripheral Artery Disease. TOHOKU J EXP MED 2022; 258:121-128. [PMID: 35922907 DOI: 10.1620/tjem.2022.j063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ryutaro Isoda
- Department of Pathology, Kawasaki Medical School.,Department of Surgery, Kawasaki Medical School General Medical Center
| | - Ichiro Morita
- Department of Surgery, Kawasaki Medical School General Medical Center
| | - Atsuhisa Isida
- Department of Surgery, Kawasaki Medical School General Medical Center
| | - Yuka Mikami
- Department of Pathology, Kawasaki Medical School
| | | | - Yasufumi Sato
- New Industry Creation Hatchery Center, Tohoku University
| | | |
Collapse
|
25
|
Müller M, Gorek L, Kamm N, Jacob R. Manipulation of the Tubulin Code Alters Directional Cell Migration and Ciliogenesis. Front Cell Dev Biol 2022; 10:901999. [PMID: 35903547 PMCID: PMC9315229 DOI: 10.3389/fcell.2022.901999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/15/2022] [Indexed: 11/23/2022] Open
Abstract
Conjunction of epithelial cells into monolayer sheets implies the ability to migrate and to undergo apicobasal polarization. Both processes comprise reorganization of cytoskeletal elements and rearrangements of structural protein interactions. We modulated expression of tubulin tyrosin ligase (TTL), the enzyme that adds tyrosine to the carboxy terminus of detyrosinated α-tubulin, to study the role of tubulin detyrosination/-tyrosination in the orientation of cell motility and in epithelial morphogenesis. Oriented cell migration and the organization of focal adhesions significantly lose directionality with diminishing amounts of microtubules enriched in detyrosinated tubulin. On the other hand, increasing quantities of detyrosinated tubulin results in faster plus end elongation of microtubules in migrating and in polarized epithelial cells. These plus ends are decorated by the plus end binding protein 1 (EB1), which mediates interaction between microtubules enriched in detyrosinated tubulin and the integrin-ILK complex at focal adhesions. EB1 accumulates at the apical cell pole at the base of the primary cilium following apicobasal polarization. Polarized cells almost devoid of detyrosinated tubulin form stunted primary cilia and multiluminal cysts in 3D-matrices. We conclude that the balance between detyrosinated and tyrosinated tubulin alters microtubule dynamics, affects the orientation of focal adhesions and determines the organization of primary cilia on epithelial cells.
Collapse
Affiliation(s)
- Manuel Müller
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodelling, GRK 2213, Philipps-Universität Marburg, Marburg, Germany
| | - Lena Gorek
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg, Germany
| | - Natalia Kamm
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg, Germany
| | - Ralf Jacob
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodelling, GRK 2213, Philipps-Universität Marburg, Marburg, Germany
- *Correspondence: Ralf Jacob,
| |
Collapse
|
26
|
Liu C, Chen Y, Xie Y, Xiang M. Tubulin Post-translational Modifications: Potential Therapeutic Approaches to Heart Failure. Front Cell Dev Biol 2022; 10:872058. [PMID: 35493101 PMCID: PMC9039000 DOI: 10.3389/fcell.2022.872058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
In recent decades, advancing insights into the mechanisms of cardiac dysfunction have focused on the involvement of microtubule network. A variety of tubulin post-translational modifications have been discovered to fine-tune the microtubules’ properties and functions. Given the limits of therapies based on conserved structures of the skeleton, targeting tubulin modifications appears to be a potentially promising therapeutic strategy. Here we review the current understanding of tubulin post-translational modifications in regulating microtubule functions in the cardiac system. We also discussed how altered modifications may lead to a range of cardiac dysfunctions, many of which are linked to heart failure.
Collapse
Affiliation(s)
- Chang Liu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuwen Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Lu YM, Zheng C. The Expression and Function of Tubulin Isotypes in Caenorhabditis elegans. Front Cell Dev Biol 2022; 10:860065. [PMID: 35399537 PMCID: PMC8987236 DOI: 10.3389/fcell.2022.860065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Microtubules, made from the polymerization of the highly conserved α/β-tubulin heterodimers, serve as important components of the cytoskeleton in all eukaryotic cells. The existence of multiple tubulin isotypes in metazoan genomes and a dazzling variety of tubulin posttranslational modifications (PTMs) prompted the “tubulin code” hypothesis, which proposed that microtubule structure and functions are determined by the tubulin composition and PTMs. Evidence for the tubulin code has emerged from studies in several organisms with the characterization of specific tubulins for their expression and functions. The studies of tubulin PTMs are accelerated by the discovery of the enzymes that add or remove the PTMs. In tubulin research, the use of simple organisms, such as Caenorhabditis elegans, has been instrumental for understanding the expression and functional specialization of tubulin isotypes and the effects of their PTMs. In this review, we summarize the current understanding of the expression patterns and cellular functions of the nine α-tubulin and six β-tubulin isotypes. Expression studies are greatly facilitated by the CRISPR/Cas9-mediated endogenous GFP knock-in reporters and the organism-wide single cell transcriptomic studies. Meanwhile, functional studies benefit from the ease of genetic manipulation and precise gene replacement in C. elegans. These studies identified both ubiquitously expressed tubulin isotypes and tissue-specific isotypes. The isotypes showed functional redundancy, as well as functional specificity, which is likely caused by the subtle differences in their amino acid sequences. Many of these differences concentrate at the C-terminal tails that are subjected to several PTMs. Indeed, tubulin PTM, such as polyglutamylation, is shown to modulate microtubule organization and properties in both ciliated and non-ciliated neurons. Overall, studies from C. elegans support the distinct expression and function patterns of tubulin isotypes and the importance of their PTMs and offer the promise of cracking the tubulin code at the whole-genome and the whole-organism level.
Collapse
|
28
|
Bär J, Popp Y, Bucher M, Mikhaylova M. Direct and indirect effects of tubulin post-translational modifications on microtubule stability: Insights and regulations. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119241. [PMID: 35181405 DOI: 10.1016/j.bbamcr.2022.119241] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/17/2022]
Abstract
Microtubules (MTs) mediate various cellular functions such as structural support, chromosome segregation, and intracellular transport. To achieve this, the pivotal properties of MTs have to be changeable and tightly controlled. This is enabled by a high variety of tubulin posttranslational modifications, which influence MT properties directly, via altering the MT lattice structurally, or indirectly by changing MT interaction partners. Here, the distinction between these direct and indirect effects of MT PTMs are exemplified by acetylation of the luminal α-tubulin K40 resulting in decreased rigidity of MTs, and by MT detyrosination which decreases interaction with depolymerizing proteins, thus causing more stable MTs. We discuss how these PTMs are reversed and regulated, e.g. on the level of enzyme transcription, localization, and activity via various signalling pathways including the conventional calcium-dependent proteases calpains and how advances in microscopy techniques and development of live-sensors facilitate the understanding of MT PTM interaction and effects.
Collapse
Affiliation(s)
- Julia Bär
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| | - Yannes Popp
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| | - Michael Bucher
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| | - Marina Mikhaylova
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| |
Collapse
|
29
|
Saunders HAJ, Johnson-Schlitz DM, Jenkins BV, Volkert PJ, Yang SZ, Wildonger J. Acetylated α-tubulin K394 regulates microtubule stability to shape the growth of axon terminals. Curr Biol 2022; 32:614-630.e5. [PMID: 35081332 PMCID: PMC8843987 DOI: 10.1016/j.cub.2021.12.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/19/2021] [Accepted: 12/07/2021] [Indexed: 02/09/2023]
Abstract
Microtubules are essential to neuron shape and function. Acetylation of tubulin has the potential to directly tune the behavior and function of microtubules in cells. Although proteomic studies have identified several acetylation sites in α-tubulin, the effects of acetylation at these sites remains largely unknown. This includes the highly conserved residue lysine 394 (K394), which is located at the αβ-tubulin dimer interface. Using a fly model, we show that α-tubulin K394 is acetylated in the nervous system and is an essential residue. We found that an acetylation-blocking mutation in endogenous α-tubulin, K394R, perturbs the synaptic morphogenesis of motoneurons and reduces microtubule stability. Intriguingly, the K394R mutation has opposite effects on the growth of two functionally and morphologically distinct motoneurons, revealing neuron-type-specific responses when microtubule stability is altered. Eliminating the deacetylase HDAC6 increases K394 acetylation, and the over-expression of HDAC6 reduces microtubule stability similar to the K394R mutant. Thus, our findings implicate α-tubulin K394 and its acetylation in the regulation of microtubule stability and suggest that HDAC6 regulates K394 acetylation during synaptic morphogenesis.
Collapse
Affiliation(s)
- Harriet A. J. Saunders
- Integrated Program in Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Dena M. Johnson-Schlitz
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Brian V. Jenkins
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Peter J. Volkert
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Biochemistry Scholars Program, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA
| | - Sihui Z. Yang
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Cellular & Molecular Biology Graduate Program, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA
| | - Jill Wildonger
- Department of Biochemistry, University of Wisconsin-Madison, 440 Henry Mall, Madison, WI, 53706, USA,Current address: Pediatrics Department and Biological Sciences Division, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA,Lead and author for correspondence:
| |
Collapse
|
30
|
JIP3 links lysosome transport to regulation of multiple components of the axonal cytoskeleton. Commun Biol 2022; 5:5. [PMID: 35013510 PMCID: PMC8748971 DOI: 10.1038/s42003-021-02945-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
Lysosome axonal transport is important for the clearance of cargoes sequestered by the endocytic and autophagic pathways. Building on observations that mutations in the JIP3 (MAPK8IP3) gene result in lysosome-filled axonal swellings, we analyzed the impact of JIP3 depletion on the cytoskeleton of human neurons. Dynamic focal lysosome accumulations were accompanied by disruption of the axonal periodic scaffold (spectrin, F-actin and myosin II) throughout each affected axon. Additionally, axonal microtubule organization was locally disrupted at each lysosome-filled swelling. This local axonal microtubule disorganization was accompanied by accumulations of both F-actin and myosin II. These results indicate that transport of axonal lysosomes is functionally interconnected with mechanisms that control the organization and maintenance of the axonal cytoskeleton. They have potential relevance to human neurological disease arising from JIP3 mutations as well as for neurodegenerative diseases associated with the focal accumulations of lysosomes within axonal swellings such as Alzheimer’s disease. Rafiq et al. report that disruption of JIP3-dependent control of axonal lysosome transport in human neurons results in unexpected changes to the organization of multiple cytoskeletal proteins. This study provides new insights that improve our understanding of intellectual disabilities caused by mutations in JIP3, and are relevant for neurodegenerative diseases associated with accumulations of lysosomes such as the Alzheimer’s disease
Collapse
|
31
|
Peris L, Parato J, Qu X, Soleilhac JM, Lanté F, Kumar A, Pero ME, Martínez-Hernández J, Corrao C, Falivelli G, Payet F, Gory-Fauré S, Bosc C, Blanca Ramirez M, Sproul A, Brocard J, Di Cara B, Delagrange P, Buisson A, Goldberg Y, Moutin MJ, Bartolini F, Andrieux A. OUP accepted manuscript. Brain 2022; 145:2486-2506. [PMID: 35148384 PMCID: PMC9337816 DOI: 10.1093/brain/awab436] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/04/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022] Open
Abstract
Microtubules play fundamental roles in the maintenance of neuronal processes and in synaptic function and plasticity. While dynamic microtubules are mainly composed of tyrosinated tubulin, long-lived microtubules contain detyrosinated tubulin, suggesting that the tubulin tyrosination/detyrosination cycle is a key player in the maintenance of microtubule dynamics and neuronal homeostasis, conditions that go awry in neurodegenerative diseases. In the tyrosination/detyrosination cycle, the C-terminal tyrosine of α-tubulin is removed by tubulin carboxypeptidases and re-added by tubulin tyrosine ligase (TTL). Here we show that TTL heterozygous mice exhibit decreased tyrosinated microtubules, reduced dendritic spine density and both synaptic plasticity and memory deficits. We further report decreased TTL expression in sporadic and familial Alzheimer’s disease, and reduced microtubule dynamics in human neurons harbouring the familial APP-V717I mutation. Finally, we show that synapses visited by dynamic microtubules are more resistant to oligomeric amyloid-β peptide toxicity and that expression of TTL, by restoring microtubule entry into spines, suppresses the loss of synapses induced by amyloid-β peptide. Together, our results demonstrate that a balanced tyrosination/detyrosination tubulin cycle is necessary for the maintenance of synaptic plasticity, is protective against amyloid-β peptide-induced synaptic damage and that this balance is lost in Alzheimer’s disease, providing evidence that defective tubulin retyrosination may contribute to circuit dysfunction during neurodegeneration in Alzheimer’s disease.
Collapse
Affiliation(s)
- Leticia Peris
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julie Parato
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Natural Sciences, SUNY ESC, Brooklyn, NY 11201, USA
| | - Xiaoyi Qu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jean Marc Soleilhac
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Fabien Lanté
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Atul Kumar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Maria Elena Pero
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - José Martínez-Hernández
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Charlotte Corrao
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Giulia Falivelli
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Floriane Payet
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Sylvie Gory-Fauré
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Christophe Bosc
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Marian Blanca Ramirez
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrew Sproul
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jacques Brocard
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | | | | | - Alain Buisson
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Yves Goldberg
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Marie Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Annie Andrieux
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| |
Collapse
|
32
|
Desmin intermediate filaments and tubulin detyrosination stabilize growing microtubules in the cardiomyocyte. Basic Res Cardiol 2022; 117:53. [PMID: 36326891 PMCID: PMC9633452 DOI: 10.1007/s00395-022-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/26/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
In heart failure, an increased abundance of post-translationally detyrosinated microtubules stiffens the cardiomyocyte and impedes its contractile function. Detyrosination promotes interactions between microtubules, desmin intermediate filaments, and the sarcomere to increase cytoskeletal stiffness, yet the mechanism by which this occurs is unknown. We hypothesized that detyrosination may regulate the growth and shrinkage of dynamic microtubules to facilitate interactions with desmin and the sarcomere. Through a combination of biochemical assays and direct observation of growing microtubule plus-ends in adult cardiomyocytes, we find that desmin is required to stabilize growing microtubules at the level of the sarcomere Z-disk, where desmin also rescues shrinking microtubules from continued depolymerization. Further, reducing detyrosination (i.e. tyrosination) below basal levels promotes frequent depolymerization and less efficient growth of microtubules. This is concomitant with tyrosination promoting the interaction of microtubules with the depolymerizing protein complex of end-binding protein 1 (EB1) and CAP-Gly domain-containing linker protein 1 (CLIP1/CLIP170). The dynamic growth and shrinkage of tyrosinated microtubules reduce their opportunity for stabilizing interactions at the Z-disk region, coincident with tyrosination globally reducing microtubule stability. These data provide a model for how intermediate filaments and tubulin detyrosination establish long-lived and physically reinforced microtubules that stiffen the cardiomyocyte and inform both the mechanism of action and therapeutic index for strategies aimed at restoring tyrosination for the treatment of cardiac disease.
Collapse
|
33
|
Sanyal C, Pietsch N, Ramirez Rios S, Peris L, Carrier L, Moutin MJ. The detyrosination/re-tyrosination cycle of tubulin and its role and dysfunction in neurons and cardiomyocytes. Semin Cell Dev Biol 2021; 137:46-62. [PMID: 34924330 DOI: 10.1016/j.semcdb.2021.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022]
Abstract
Among the variety of post-translational modifications to which microtubules are subjected, the detyrosination/re-tyrosination cycle is specific to tubulin. It is conserved by evolution and characterized by the enzymatic removal and re-addition of a gene-encoded tyrosine residue at the C-terminus of α-tubulin. Detyrosinated tubulin can be further converted to Δ2-tubulin by the removal of an additional C-terminal glutamate residue. Detyrosinated and Δ2-tubulin are carried by stable microtubules whereas tyrosinated microtubules are present on dynamic polymers. The cycle regulates trafficking of many cargo transporting molecular motors and is linked to the microtubule dynamics via regulation of microtubule interactions with specific cellular effectors such as kinesin-13. Here, we give an historical overview of the general features discovered for the cycle. We highlight the recent progress toward structure and functioning of the enzymes that keep the levels of tyrosinated and detyrosinated tubulin in cells, the long-known tubulin tyrosine ligase and the recently discovered vasohibin-SVBP complexes. We further describe how the cycle controls microtubule functions in healthy neurons and cardiomyocytes and how deregulations of the cycle are involved in dysfunctions of these highly differentiated cells, leading to neurodegeneration and heart failure in humans.
Collapse
Affiliation(s)
- Chadni Sanyal
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Niels Pietsch
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sacnicte Ramirez Rios
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Leticia Peris
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Marie-Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
34
|
Wei Y, Wang G, Chen J, Xiao L, Wu Z, He J, Zhang N. Maternal deprivation induces cytoskeletal alterations and depressive-like behavior in adult male rats by regulating the AKT/GSK3β/CRMP2 signaling pathway. Physiol Behav 2021; 242:113625. [PMID: 34666114 DOI: 10.1016/j.physbeh.2021.113625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 08/21/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022]
Abstract
Early-life adverse events exert persistent effects on brain functions and may increase the risk of psychopathology in adulthood. However, the underlying mechanism remains unclear. The purpose of our study was to study the long-lasting effects of maternal deprivation (MD) on depression-related behaviors and microtubule dynamics, and to illuminate the underlying molecular mechanism. Rat pups were separated from the dams for 360 min (MD) or 15 min (brief maternal separation) each day from postnatal day 4 through 10. Rats with MD experience showed significant depressive-like behaviors in adulthood, while brief maternal separation did not alter the behaviors. Behavioral alterations in the MD group were accompanied by alterations in the AKT/GSK3β/CRMP2 signaling pathway and hyperphosphorylation of CRMP2. CRMP2 interacted and colocalized with the cytoskeleton in the hippocampus, and the overlap of CRMP2 and tubulin staining in the hippocampus of MD rats was decreased. In MD rats, the expression of the α-tubulin isoforms Acet-tubulin and Tyr-tubulin changed, and the ratio of Tyr/Acet-tubulin, which is an important marker of microtubule dynamics, was decreased, indicating decreased microtubule dynamics. Furthermore, regulation of the AKT/GSK3β/CRMP2 signaling pathway by an LY294002 microinjection in the hippocampus resulted in cytoskeletal alterations and depressive-like behaviors in rats. These findings suggest that early-life MD induces depressive-like behaviors and cytoskeletal alterations in adult male rats and that the effects may be partly mediated by the AKT/GSK3β/CRMP2 signaling pathway. An understanding of the mechanism underlying the effect of MD on behaviors is crucial for developing pharmacological and psychological interventions for childhood neglect.
Collapse
Affiliation(s)
- Yanyan Wei
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China.
| | - Jingxu Chen
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Zuotian Wu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Jing He
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Nan Zhang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| |
Collapse
|
35
|
Laisne MC, Michallet S, Lafanechère L. Characterization of Microtubule Destabilizing Drugs: A Quantitative Cell-Based Assay That Bridges the Gap between Tubulin Based- and Cytotoxicity Assays. Cancers (Basel) 2021; 13:cancers13205226. [PMID: 34680374 PMCID: PMC8533752 DOI: 10.3390/cancers13205226] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The characterization of new microtubule depolymerizing agents relies mainly on purified tubulin assays in vitro and on cytotoxicity tests. However, the relationship between the in vitro effects of drugs and their effect on cell viability may not be direct. Here, we have systematically compared the effect of four reference drugs on tubulin polymerization in vitro and in cells, using a recently-developed quantitative assay of the cellular microtubule content. By comparing these results with cell viability assays, we found that this new cellular microtubule content test better predicts cellular drug toxicity than the in vitro tubulin polymerization assay. This test can thus be easily implemented in the process of discovery and characterization of novel microtubule poisons. Abstract (1) Background: Microtubule depolymerizing agents (MDAs) are commonly used for cancer treatment. However, the therapeutic use of such microtubule inhibitors is limited by their toxicity and the emergence of resistance. Thus, there is still a sustained effort to develop new MDAs. During the characterization of such agents, mainly through in vitro analyses using purified tubulin and cytotoxicity assays, quantitative comparisons are mandatory. The relationship between the effect of the drugs on purified tubulin and on cell viability are not always direct. (2) Methods: We have recently developed a cell-based assay that quantifies the cellular microtubule content. In this study, we have conducted a systematic comparative analysis of the effect of four well-characterized MDAs on the kinetics of in vitro tubulin assembly, on the cellular microtubule content (using our recently developed assay) and on cell viability. (3) Conclusions: These assays gave complementary results. Additionally, we found that the drugs’ effect on in vitro tubulin polymerization is not completely predictive of their relative cytotoxicity. Their effect on the cellular microtubule content, however, is closely related to their effect on cell viability. In conclusion, the assay we have recently developed can bridge the gap between in vitro tubulin assays and cell viability assays.
Collapse
|
36
|
Lechler T, Mapelli M. Spindle positioning and its impact on vertebrate tissue architecture and cell fate. Nat Rev Mol Cell Biol 2021; 22:691-708. [PMID: 34158639 PMCID: PMC10544824 DOI: 10.1038/s41580-021-00384-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 12/18/2022]
Abstract
In multicellular systems, oriented cell divisions are essential for morphogenesis and homeostasis as they determine the position of daughter cells within the tissue and also, in many cases, their fate. Early studies in invertebrates led to the identification of conserved core mechanisms of mitotic spindle positioning centred on the Gαi-LGN-NuMA-dynein complex. In recent years, much has been learnt about the way this complex functions in vertebrate cells. In particular, studies addressed how the Gαi-LGN-NuMA-dynein complex dynamically crosstalks with astral microtubules and the actin cytoskeleton, and how it is regulated to orient the spindle according to cellular and tissue-wide cues. We have also begun to understand how dynein motors and actin regulators interact with mechanosensitive adhesion molecules sensing extracellular mechanical stimuli, such as cadherins and integrins, and with signalling pathways so as to respond to extracellular cues instructing the orientation of the division axis in vivo. In this Review, with the focus on epithelial tissues, we discuss the molecular mechanisms of mitotic spindle orientation in vertebrate cells, and how this machinery is regulated by epithelial cues and extracellular signals to maintain tissue cohesiveness during mitosis. We also outline recent knowledge of how spindle orientation impacts tissue architecture in epithelia and its emerging links to the regulation of cell fate decisions. Finally, we describe how defective spindle orientation can be corrected or its effects eliminated in tissues under physiological conditions, and the pathological implications associated with spindle misorientation.
Collapse
Affiliation(s)
- Terry Lechler
- Department of Dermatology, Duke University Medical Center, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
37
|
Ghasemizadeh A, Christin E, Guiraud A, Couturier N, Abitbol M, Risson V, Girard E, Jagla C, Soler C, Laddada L, Sanchez C, Jaque-Fernandez FI, Jacquemond V, Thomas JL, Lanfranchi M, Courchet J, Gondin J, Schaeffer L, Gache V. MACF1 controls skeletal muscle function through the microtubule-dependent localization of extra-synaptic myonuclei and mitochondria biogenesis. eLife 2021; 10:e70490. [PMID: 34448452 PMCID: PMC8500715 DOI: 10.7554/elife.70490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/10/2021] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.
Collapse
Affiliation(s)
- Alireza Ghasemizadeh
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Emilie Christin
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Alexandre Guiraud
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Nathalie Couturier
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Marie Abitbol
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
- Université Marcy l’Etoile, VetAgro SupLyonFrance
| | - Valerie Risson
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Emmanuelle Girard
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Christophe Jagla
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Cedric Soler
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Lilia Laddada
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Colline Sanchez
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Francisco-Ignacio Jaque-Fernandez
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Vincent Jacquemond
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Jean-Luc Thomas
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Marine Lanfranchi
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Julien Courchet
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Julien Gondin
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Laurent Schaeffer
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Vincent Gache
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| |
Collapse
|
38
|
The Interaction of Possible Anti-AD ASA-NAP Peptide Conjugate with Tubulin: A Theoretical and Experimental Insight. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10267-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Chen J, Kholina E, Szyk A, Fedorov VA, Kovalenko I, Gudimchuk N, Roll-Mecak A. α-tubulin tail modifications regulate microtubule stability through selective effector recruitment, not changes in intrinsic polymer dynamics. Dev Cell 2021; 56:2016-2028.e4. [PMID: 34022132 PMCID: PMC8476856 DOI: 10.1016/j.devcel.2021.05.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 10/21/2022]
Abstract
Microtubules are non-covalent polymers of αβ-tubulin dimers. Posttranslational processing of the intrinsically disordered C-terminal α-tubulin tail produces detyrosinated and Δ2-tubulin. Although these are widely employed as proxies for stable cellular microtubules, their effect (and of the α-tail) on microtubule dynamics remains uncharacterized. Using recombinant, engineered human tubulins, we now find that neither detyrosinated nor Δ2-tubulin affect microtubule dynamics, while the α-tubulin tail is an inhibitor of microtubule growth. Consistent with the latter, molecular dynamics simulations show the α-tubulin tail transiently occluding the longitudinal microtubule polymerization interface. The marked differential in vivo stabilities of the modified microtubule subpopulations, therefore, must result exclusively from selective effector recruitment. We find that tyrosination quantitatively tunes CLIP-170 density at the growing plus end and that CLIP170 and EB1 synergize to selectively upregulate the dynamicity of tyrosinated microtubules. Modification-dependent recruitment of regulators thereby results in microtubule subpopulations with distinct dynamics, a tenet of the tubulin code hypothesis.
Collapse
Affiliation(s)
- Jiayi Chen
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Ekaterina Kholina
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Agnieszka Szyk
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Vladimir A Fedorov
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Ilya Kovalenko
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia; Astrakhan State University, Astrakhan 414056, Russia; Sechenov University, Moscow 119991, Russia
| | - Nikita Gudimchuk
- Department of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA; Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
40
|
Rasamizafy SF, Delsert C, Rabeharivelo G, Cau J, Morin N, van Dijk J. Mitotic Acetylation of Microtubules Promotes Centrosomal PLK1 Recruitment and Is Required to Maintain Bipolar Spindle Homeostasis. Cells 2021; 10:1859. [PMID: 34440628 PMCID: PMC8394630 DOI: 10.3390/cells10081859] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Tubulin post-translational modifications regulate microtubule properties and functions. Mitotic spindle microtubules are highly modified. While tubulin detyrosination promotes proper mitotic progression by recruiting specific microtubule-associated proteins motors, tubulin acetylation that occurs on specific microtubule subsets during mitosis is less well understood. Here, we show that siRNA-mediated depletion of the tubulin acetyltransferase ATAT1 in epithelial cells leads to a prolonged prometaphase arrest and the formation of monopolar spindles. This results from collapse of bipolar spindles, as previously described in cells deficient for the mitotic kinase PLK1. ATAT1-depleted mitotic cells have defective recruitment of PLK1 to centrosomes, defects in centrosome maturation and thus microtubule nucleation, as well as labile microtubule-kinetochore attachments. Spindle bipolarity could be restored, in the absence of ATAT1, by stabilizing microtubule plus-ends or by increasing PLK1 activity at centrosomes, demonstrating that the phenotype is not just a consequence of lack of K-fiber stability. We propose that microtubule acetylation of K-fibers is required for a recently evidenced cross talk between centrosomes and kinetochores.
Collapse
Affiliation(s)
- Sylvia Fenosoa Rasamizafy
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Claude Delsert
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
- Institut Français de Recherche pour l’Exploitation de la mer, L3AS, 34250 Palavas-les-Flots, France
| | - Gabriel Rabeharivelo
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Julien Cau
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- IGH, CNRS UMR 9002, 141, rue de la Cardonille, 34396 Montpellier, France
- Montpellier Rio Imaging, 34293 Montpellier, France
| | - Nathalie Morin
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Juliette van Dijk
- Université de Montpellier, 34293 Montpellier, France; (S.F.R.); (C.D.); (G.R.); (J.C.)
- Centre National de la Recherche Scientifique (CNRS) UMR5237, 1919 Route de Mende, 34293 Montpellier, France
| |
Collapse
|
41
|
MacTaggart B, Kashina A. Posttranslational modifications of the cytoskeleton. Cytoskeleton (Hoboken) 2021; 78:142-173. [PMID: 34152688 DOI: 10.1002/cm.21679] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
The cytoskeleton plays important roles in many essential processes at the cellular and organismal levels, including cell migration and motility, cell division, and the establishment and maintenance of cell and tissue architecture. In order to facilitate these varied functions, the main cytoskeletal components-microtubules, actin filaments, and intermediate filaments-must form highly diverse intracellular arrays in different subcellular areas and cell types. The question of how this diversity is conferred has been the focus of research for decades. One key mechanism is the addition of posttranslational modifications (PTMs) to the major cytoskeletal proteins. This posttranslational addition of various chemical groups dramatically increases the complexity of the cytoskeletal proteome and helps facilitate major global and local cytoskeletal functions. Cytoskeletal proteins undergo many PTMs, most of which are not well understood. Recent technological advances in proteomics and cell biology have allowed for the in-depth study of individual PTMs and their functions in the cytoskeleton. Here, we provide an overview of the major PTMs that occur on the main structural components of the three cytoskeletal systems-tubulin, actin, and intermediate filament proteins-and highlight the cellular function of these modifications.
Collapse
Affiliation(s)
- Brittany MacTaggart
- School of Veterinary Medicine, Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna Kashina
- School of Veterinary Medicine, Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Ramirez Rios S, Torres A, Diemer H, Collin-Faure V, Cianférani S, Lafanechère L, Rabilloud T. A proteomic-informed view of the changes induced by loss of cellular adherence: The example of mouse macrophages. PLoS One 2021; 16:e0252450. [PMID: 34048472 PMCID: PMC8162644 DOI: 10.1371/journal.pone.0252450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/14/2021] [Indexed: 11/19/2022] Open
Abstract
Except cells circulating in the bloodstream, most cells in vertebrates are adherent. Studying the repercussions of adherence per se in cell physiology is thus very difficult to carry out, although it plays an important role in cancer biology, e.g. in the metastasis process. In order to study how adherence impacts major cell functions, we used a murine macrophage cell line. Opposite to the monocyte/macrophage system, where adherence is associated with the acquisition of differentiated functions, these cells can be grown in both adherent or suspension conditions without altering their differentiated functions (phagocytosis and inflammation signaling). We used a proteomic approach to cover a large panel of proteins potentially modified by the adherence status. Targeted experiments were carried out to validate the proteomic results, e.g. on metabolic enzymes, mitochondrial and cytoskeletal proteins. The mitochondrial activity was increased in non-adherent cells compared with adherent cells, without differences in glucose consumption. Concerning the cytoskeleton, a rearrangement of the actin organization (filopodia vs sub-cortical network) and of the microtubule network were observed between adherent and non-adherent cells. Taken together, these data show the mechanisms at play for the modification of the cytoskeleton and also modifications of the metabolic activity between adherent and non-adherent cells.
Collapse
Affiliation(s)
- Sacnite Ramirez Rios
- Institute for Advanced Biosciences, Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Grenoble, France
| | - Anaelle Torres
- Chemistry and Biology of Metals, Univ. Grenoble Alpes, CNRS UMR5249, CEA, IRIG-DIESE-CBM-ProMD, Grenoble, France
| | - Hélène Diemer
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
- Infrastructure Nationale de Protéomique, FR2048 ProFI, Strasbourg, France
| | - Véronique Collin-Faure
- Chemistry and Biology of Metals, Univ. Grenoble Alpes, CNRS UMR5249, CEA, IRIG-DIESE-CBM-ProMD, Grenoble, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
- Infrastructure Nationale de Protéomique, FR2048 ProFI, Strasbourg, France
| | - Laurence Lafanechère
- Institute for Advanced Biosciences, Univ. Grenoble Alpes, CNRS UMR 5309, INSERM U1209, Grenoble, France
| | - Thierry Rabilloud
- Chemistry and Biology of Metals, Univ. Grenoble Alpes, CNRS UMR5249, CEA, IRIG-DIESE-CBM-ProMD, Grenoble, France
- * E-mail:
| |
Collapse
|
43
|
Bouvrais H, Chesneau L, Le Cunff Y, Fairbrass D, Soler N, Pastezeur S, Pécot T, Kervrann C, Pécréaux J. The coordination of spindle-positioning forces during the asymmetric division of the Caenorhabditis elegans zygote. EMBO Rep 2021; 22:e50770. [PMID: 33900015 DOI: 10.15252/embr.202050770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022] Open
Abstract
In Caenorhabditis elegans zygote, astral microtubules generate forces essential to position the mitotic spindle, by pushing against and pulling from the cortex. Measuring microtubule dynamics there, we revealed the presence of two populations, corresponding to pulling and pushing events. It offers a unique opportunity to study, under physiological conditions, the variations of both spindle-positioning forces along space and time. We propose a threefold control of pulling force, by polarity, spindle position and mitotic progression. We showed that the sole anteroposterior asymmetry in dynein on-rate, encoding pulling force imbalance, is sufficient to cause posterior spindle displacement. The positional regulation, reflecting the number of microtubule contacts in the posterior-most region, reinforces this imbalance only in late anaphase. Furthermore, we exhibited the first direct proof that dynein processivity increases along mitosis. It reflects the temporal control of pulling forces, which strengthens at anaphase onset following mitotic progression and independently from chromatid separation. In contrast, the pushing force remains constant and symmetric and contributes to maintaining the spindle at the cell centre during metaphase.
Collapse
Affiliation(s)
| | | | - Yann Le Cunff
- CNRS, IGDR - UMR 6290, University of Rennes, Rennes, France
| | | | - Nina Soler
- CNRS, IGDR - UMR 6290, University of Rennes, Rennes, France
| | | | - Thierry Pécot
- INRIA, Centre Rennes - Bretagne Atlantique, Rennes, France
| | | | | |
Collapse
|
44
|
Waites C, Qu X, Bartolini F. The synaptic life of microtubules. Curr Opin Neurobiol 2021; 69:113-123. [PMID: 33873059 DOI: 10.1016/j.conb.2021.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022]
Abstract
In neurons, control of microtubule dynamics is required for multiple homeostatic and regulated activities. Over the past few decades, a great deal has been learned about the role of the microtubule cytoskeleton in axonal and dendritic transport, with a broad impact on neuronal health and disease. However, significantly less attention has been paid to the importance of microtubule dynamics in directly regulating synaptic function. Here, we review emerging literature demonstrating that microtubules enter synapses and control central aspects of synaptic activity, including neurotransmitter release and synaptic plasticity. The pleiotropic effects caused by a dysfunctional synaptic microtubule cytoskeleton may thus represent a key point of vulnerability for neurons and a primary driver of neurological disease.
Collapse
Affiliation(s)
- Clarissa Waites
- Department of Neuroscience, Columbia University, 3227 Broadway, New York, NY 10027, USA
| | - Xiaoyi Qu
- Department of Pathology & Cell Biology, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032, USA
| | - Francesca Bartolini
- Department of Pathology & Cell Biology, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032, USA.
| |
Collapse
|
45
|
Xia Y, Nasif L, Giasson BI. Pathogenic MAPT mutations Q336H and Q336R have isoform-dependent differences in aggregation propensity and microtubule dysfunction. J Neurochem 2021; 158:455-466. [PMID: 33772783 DOI: 10.1111/jnc.15358] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/24/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022]
Abstract
Tauopathies are a group of heterogeneous neurodegenerative disorders characterized by brain deposition of tau inclusions. These insidious disorders include Alzheimer's disease and frontotemporal dementia, the two leading causes of dementia. Mutations in the microtubule-associated protein tau (MAPT) gene lead to familial forms of frontotemporal dementia. Previously, we used cell-based assays to screen over 20 missense tau mutations and found that decreased microtubule (MT) binding affinity was the most shared property. As a break from this trend, the MAPT mutations Q336H and Q336R are thought to promote MT assembly rather than inhibit it based on in vitro studies. Q336H and Q336R MAPT mutations also cause early onset frontotemporal dementia with Pick bodies, which are mostly composed of 3R tau isoforms. To provide further insights on the pathobiology of these mutations, we assessed Q336H and Q336R tau mutants for aggregation propensity and MT binding in cell-based assays in the context of both 0N3R and 0N4R tau isoforms. Q336R tau was prone to prion-like seeded aggregation but both Q336H and Q336R tau led to increased MT binding. Additionally, we found that different tau isoforms with these mutations heterogeneously regulate different MT subpopulations of tyrosinated and acetylated MTs, markers of newly formed MTs and stable MTs. The Q336H and Q336R tau mutations may exemplify an alternative mechanism where pathogenic tau can bind MTs with higher affinity and hyperstabilize MTs, which prevent proper MT regulation and homeostasis.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lith Nasif
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
46
|
Renda F, Khodjakov A. Role of spatial patterns and kinetochore architecture in spindle morphogenesis. Semin Cell Dev Biol 2021; 117:75-85. [PMID: 33836948 PMCID: PMC8762378 DOI: 10.1016/j.semcdb.2021.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/30/2022]
Abstract
Mitotic spindle is a self-assembling macromolecular machine responsible for the faithful segregation of chromosomes during cell division. Assembly of the spindle is believed to be governed by the 'Search & Capture' (S&C) principle in which dynamic microtubules explore space in search of kinetochores while the latter capture microtubules and thus connect chromosomes to the spindle. Due to the stochastic nature of the encounters between kinetochores and microtubules, the time required for incorporating all chromosomes into the spindle is profoundly affected by geometric constraints, such as the size and shape of kinetochores as well as their distribution in space at the onset of spindle assembly. In recent years, several molecular mechanisms that control these parameters have been discovered. It is now clear that stochastic S&C takes place in structured space, where components are optimally distributed and oriented to minimize steric hindrances. Nucleation of numerous non-centrosomal microtubules near kinetochores accelerates capture, while changes in the kinetochore architecture at various stages of spindle assembly promote proper connection of sister kinetochores to the opposite spindle poles. Here we discuss how the concerted action of multiple facilitating mechanisms ensure that the spindle assembles rapidly yet with a minimal number of errors.
Collapse
Affiliation(s)
- Fioranna Renda
- Biggs Laboratory, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12237, United States.
| | - Alexey Khodjakov
- Biggs Laboratory, Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12237, United States; Rensselaer Polytechnic Institute, Troy, NY 12180, United States.
| |
Collapse
|
47
|
Parato J, Bartolini F. The microtubule cytoskeleton at the synapse. Neurosci Lett 2021; 753:135850. [PMID: 33775740 DOI: 10.1016/j.neulet.2021.135850] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
In neurons, microtubules (MTs) provide routes for transport throughout the cell and structural support for dendrites and axons. Both stable and dynamic MTs are necessary for normal neuronal functions. Research in the last two decades has demonstrated that MTs play additional roles in synaptic structure and function in both pre- and postsynaptic elements. Here, we review current knowledge of the functions that MTs perform in excitatory and inhibitory synapses, as well as in the neuromuscular junction and other specialized synapses, and discuss the implications that this knowledge may have in neurological disease.
Collapse
Affiliation(s)
- Julie Parato
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States; SUNY Empire State College, Department of Natural Sciences, 177 Livingston Street, Brooklyn, NY, 11201, United States
| | - Francesca Bartolini
- Columbia University Medical Center, Department of Pathology & Cell Biology, 630 West 168(th)Street, P&S 15-421, NY, NY, 10032, United States.
| |
Collapse
|
48
|
Kesarwani S, Lama P, Chandra A, Reddy PP, Jijumon AS, Bodakuntla S, Rao BM, Janke C, Das R, Sirajuddin M. Genetically encoded live-cell sensor for tyrosinated microtubules. J Cell Biol 2021; 219:152071. [PMID: 32886100 PMCID: PMC7659708 DOI: 10.1083/jcb.201912107] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/16/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Microtubule cytoskeleton exists in various biochemical forms in different cells due to tubulin posttranslational modifications (PTMs). Tubulin PTMs are known to affect microtubule stability, dynamics, and interaction with MAPs and motors in a specific manner, widely known as tubulin code hypothesis. At present, there exists no tool that can specifically mark tubulin PTMs in living cells, thus severely limiting our understanding of their dynamics and cellular functions. Using a yeast display library, we identified a binder against terminal tyrosine of α-tubulin, a unique PTM site. Extensive characterization validates the robustness and nonperturbing nature of our binder as tyrosination sensor, a live-cell tubulin nanobody specific towards tyrosinated microtubules. Using this sensor, we followed nocodazole-, colchicine-, and vincristine-induced depolymerization events of tyrosinated microtubules in real time and found each distinctly perturbs the microtubule polymer. Together, our work describes a novel tyrosination sensor and its potential applications to study the dynamics of microtubule and their PTM processes in living cells.
Collapse
Affiliation(s)
- Shubham Kesarwani
- Centre for Cardiovascular Biology and Diseases, Institute for Stem Cell Science and Regenerative Medicine, Gandhi Krishi Vigyan Kendra Campus, Bangalore, India.,Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Prakash Lama
- Centre for Cardiovascular Biology and Diseases, Institute for Stem Cell Science and Regenerative Medicine, Gandhi Krishi Vigyan Kendra Campus, Bangalore, India.,Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anchal Chandra
- National Center for Biological Sciences, Tata Institute of Fundamental Research, Gandhi Krishi Vigyan Kendra Campus, Bangalore, India
| | - P Purushotam Reddy
- National Center for Biological Sciences, Tata Institute of Fundamental Research, Gandhi Krishi Vigyan Kendra Campus, Bangalore, India
| | - A S Jijumon
- Institut Curie, Paris Sciences et Lettres University, Centre National de la Recherche Scientifique UMR3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, Centre National de la Recherche Scientifique UMR3348, Orsay, France
| | - Satish Bodakuntla
- Institut Curie, Paris Sciences et Lettres University, Centre National de la Recherche Scientifique UMR3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, Centre National de la Recherche Scientifique UMR3348, Orsay, France
| | - Balaji M Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC
| | - Carsten Janke
- Institut Curie, Paris Sciences et Lettres University, Centre National de la Recherche Scientifique UMR3348, Orsay, France.,Université Paris Sud, Université Paris-Saclay, Centre National de la Recherche Scientifique UMR3348, Orsay, France
| | - Ranabir Das
- National Center for Biological Sciences, Tata Institute of Fundamental Research, Gandhi Krishi Vigyan Kendra Campus, Bangalore, India
| | - Minhajuddin Sirajuddin
- Centre for Cardiovascular Biology and Diseases, Institute for Stem Cell Science and Regenerative Medicine, Gandhi Krishi Vigyan Kendra Campus, Bangalore, India
| |
Collapse
|
49
|
Henrie H, Bakhos-Douaihy D, Cantaloube I, Pilon A, Talantikite M, Stoppin-Mellet V, Baillet A, Poüs C, Benoit B. Stress-induced phosphorylation of CLIP-170 by JNK promotes microtubule rescue. J Cell Biol 2021; 219:151834. [PMID: 32491151 PMCID: PMC7337496 DOI: 10.1083/jcb.201909093] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/17/2020] [Accepted: 04/20/2020] [Indexed: 01/01/2023] Open
Abstract
The stress-induced c-Jun N-terminal kinase (JNK) controls microtubule dynamics by enhancing both microtubule growth and rescues. Here, we show that upon cell stress, JNK directly phosphorylates the microtubule rescue factor CLIP-170 in its microtubule-binding domain to increase its rescue-promoting activity. Phosphomimetic versions of CLIP-170 enhance its ability to promote rescue events in vitro and in cells. Furthermore, while phosphomimetic mutations do not alter CLIP-170’s capability to form comets at growing microtubule ends, both phosphomimetic mutations and JNK activation increase the occurrence of CLIP-170 remnants on the microtubule lattice at the rear of comets. As the CLIP-170 remnants, which are potential sites of microtubule rescue, display a shorter lifetime when CLIP-170 is phosphorylated, we propose that instead of acting at the time of rescue occurrence, CLIP-170 would rather contribute in preparing the microtubule lattice for future rescues at these predetermined sites.
Collapse
Affiliation(s)
- Hélène Henrie
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France
| | - Dalal Bakhos-Douaihy
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France
| | - Isabelle Cantaloube
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France
| | - Antoine Pilon
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France.,Département de Biochimie, Hormonologie et Suivi Thérapeutique, Département Médico-Universitaire BioGeM, Assistance Publique - Hôpitaux de Paris Sorbonne Université, Paris, France
| | - Maya Talantikite
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France
| | - Virginie Stoppin-Mellet
- Grenoble Institut des Neurosciences, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1216, Université Grenoble Alpes, Grenoble, France
| | - Anita Baillet
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France
| | - Christian Poüs
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France.,Biochimie-Hormonologie, Assistance Publique - Hôpitaux de Paris Université Paris-Saclay, Clamart, France
| | - Béatrice Benoit
- Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1193, Châtenay-Malabry, France
| |
Collapse
|
50
|
Perez T, Bergès R, Maccario H, Oddoux S, Honoré S. Low concentrations of vorinostat decrease EB1 expression in GBM cells and affect microtubule dynamics, cell survival and migration. Oncotarget 2021; 12:304-315. [PMID: 33659042 PMCID: PMC7899546 DOI: 10.18632/oncotarget.27892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/01/2021] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiform (GBM) is the most frequent primitive brain tumor with a high recurrence and mortality. Histone deacetylase inhibitors (HDACi) have evoked great interest because they are able to change transcriptomic profiles to promote tumor cell death but also induce side effects due to the lack of selectivity. We show in this paper new anticancer properties and mechanisms of action of low concentrations of vorinostat on various GBM cells which acts by affecting microtubule cytoskeleton in a non-histone 3 (H3) manner. Indeed, vorinostat induces tubulin acetylation and detyrosination, affects EB stabilizing cap on microtubule plus ends and suppresses microtubule dynamic instability. We previously identified EB1 overexpression as a marker of bad prognostic in GBM. Interestingly, we show for the first time to our knowledge, a strong decrease of EB1 expression in GBM cells by a drug. Altogether, our results suggest that low dose vorinostat, which is more selective for HDAC6 inhibition, could therefore represent an interesting therapeutic option for GBM especially in patients with EB1 overexpressing tumor with lower expected side effects. A validation of our hypothesis is needed during future clinical trials with this drug in GBM.
Collapse
Affiliation(s)
- Thomas Perez
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Raphaël Bergès
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Hélène Maccario
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Sarah Oddoux
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Stéphane Honoré
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France
| |
Collapse
|