1
|
Wang HP, Singh S, Wong LC, Hsu CJ, Li SC, Lee SJ, Lee CH, Lee WT. Lacosamide Is a Novel Drug That Improves AGTPBP1 Knockout-Mediated Impairment of Neuronal and Dopaminergic Function. Mol Neurobiol 2025:10.1007/s12035-025-05016-y. [PMID: 40347376 DOI: 10.1007/s12035-025-05016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
AGTPBP1 regulates microtubule stabilization through post-translational modification of alpha-tubulin. Mutations in the AGTPBP1 gene are associated with clinical phenotypes such as early postnatal cerebellar atrophy, ataxia, spasticity, and dystonia, highlighting its critical roles in both neurodevelopment and neurodegeneration. However, how AGTPBP1 affects neurite development and its function in dopaminergic neurons remains unclear. To investigate the role of AGTPBP1, we utilized both in vitro AGTPBP1 knockout (KO) cell models and zebrafish models. Our findings reveal that AGTPBP1 KO in cells leads to excessive neurite outgrowth and significantly increases expression of collapsin response mediator protein 2 (CRMP2). Additionally, AGTPBP1 KO results in mitochondrial dysfunction and a hyperdopaminergic state in differentiated neurons. In zebrafish, knockdown of AGTPBP1 caused reduced brain volume and impaired swimming behavior, indicating disrupted neurodevelopment and motor function. Given CRMP2's involvement in both cytoskeletal dynamics and mitochondrial activity, we tested lacosamide, a drug known to modulate CRMP2 expression and phosphorylation. Lacosamide treatment in vitro improved cell morphology and restored mitochondrial function, while in vivo, it rescued brain volume deficits and enhanced swimming performance in AGTPBP1-deficient zebrafish. In conclusion, AGTPBP1 knockout impairs neuronal differentiation, induces mitochondrial dysfunction, increases oxidative stress, and promotes a hyperdopaminergic state. Our study suggests that elevated CRMP2 expression may underlie the pathophysiology of cerebellar degeneration in AGTPBP1-related disorders. Targeting CRMP2 with lacosamide represents a promising therapeutic strategy for mitigating AGTPBP1-mediated neurodegeneration.
Collapse
Affiliation(s)
- Hsin-Pei Wang
- Department of Pediatrics, National Taiwan University Hospital, Yunlin Branch, Yunlin, 970, Taiwan
- National Taiwan University College of Medicine Graduate Institute of Clinical Medicine, Taipei, 100, Taiwan
| | - Shekhar Singh
- National Taiwan University College of Medicine, Taipei, 100, Taiwan
| | - Lee-Chin Wong
- National Taiwan University College of Medicine Graduate Institute of Clinical Medicine, Taipei, 100, Taiwan
- National Taiwan University College of Medicine, Taipei, 100, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, Taipei, 100, Taiwan
- Department of Pediatric Neurology, National Taiwan University Children's Hospital, 8, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Chia-Jui Hsu
- Department of Pediatrics, National Taiwan University Hospital, Hsinchu Branch, Hsinchu City, 300, Taiwan
| | - Shih-Chi Li
- Department of Life Science, National Taiwan University, Taipei, 100, Taiwan
| | - Shyh-Jye Lee
- Department of Life Science, National Taiwan University, Taipei, 100, Taiwan
| | - Chia-Hwa Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City, 23564, Taiwan
| | - Wang-Tso Lee
- National Taiwan University College of Medicine, Taipei, 100, Taiwan.
- Department of Pediatrics, National Taiwan University Hospital, Taipei, 100, Taiwan.
- Department of Pediatric Neurology, National Taiwan University Children's Hospital, 8, Chung-Shan South Road, Taipei, 100, Taiwan.
| |
Collapse
|
2
|
Lal S, Snape TJ. Tubulin targeting agents and their implications in non-cancer disease management. Drug Discov Today 2025; 30:104338. [PMID: 40118444 DOI: 10.1016/j.drudis.2025.104338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Microtubules act as molecular 'tracks' for the intracellular transport of accessory proteins, enabling them to assemble into various larger structures, such as spindle fibres formed during the cell cycle. Microtubules provide an organisational framework for the healthy functioning of various cellular processes that work through the process of dynamic instability, driven by the hydrolysis of GTP. In this role, tubulin proteins undergo various modifications, and in doing so modulate various healthy or pathogenic physiological processes within cells. In this review, we provide a detailed update of small molecule chemical agents that interact with tubulin, along with their implications, specifically in non-cancer disease management.
Collapse
Affiliation(s)
- Samridhi Lal
- Amity Institute of Pharmacy, Amity University, Gurugram 122413 Haryana, India.
| | - Timothy J Snape
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| |
Collapse
|
3
|
Teoh J, Bartolini F. Emerging roles for tubulin PTMs in neuronal function and neurodegenerative disease. Curr Opin Neurobiol 2025; 90:102971. [PMID: 39862522 PMCID: PMC11839326 DOI: 10.1016/j.conb.2025.102971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
Neurons are equipped with microtubules of different stability with stable and dynamic domains often coexisting on the same microtubule. While dynamic microtubules undergo random transitions between disassembly and assembly, stable ones persist long enough to serve as platforms for tubulin-modifying enzymes (known as writers) that attach molecular components to the α- or β-tubulin subunits. The combination of these posttranslational modifications (PTMs) results in a "tubulin code," dictating the behavior of selected proteins (known as readers), some of which were shown to be crucial for neuronal function. Recent research has further highlighted that disturbances in tubulin PTMs can lead to neurodegeneration, sparking an emerging field of investigation with numerous questions such as whether and how tubulin PTMs can affect neurotransmission and synaptic plasticity and whether restoring balanced tubulin PTM levels could effectively prevent or mitigate neurodegenerative disease.
Collapse
Affiliation(s)
- JiaJie Teoh
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, 10032, New York, NY, USA
| | - Francesca Bartolini
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, 10032, New York, NY, USA.
| |
Collapse
|
4
|
Pérez-Revuelta L, Pérez-Boyero D, Pérez-Martín E, Cabedo VL, Téllez de Meneses PG, Weruaga E, Díaz D, Alonso JR. Neuroprotective Effects of VEGF-B in a Murine Model of Aggressive Neuronal Loss with Childhood Onset. Int J Mol Sci 2025; 26:538. [PMID: 39859255 PMCID: PMC11765331 DOI: 10.3390/ijms26020538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
In recent decades, the scientific community has faced a major challenge in the search for new therapies that can slow down or alleviate the process of neuronal death that accompanies neurodegenerative diseases. This study aimed to identify an effective therapy using neurotrophic factors to delay the rapid and aggressive cerebellar degeneration experienced by the Purkinje Cell Degeneration (PCD) mouse, a model of childhood-onset neurodegeneration with cerebellar atrophy (CONDCA). Initially, we analyzed the changes in the expression of several neurotrophic factors related to the degenerative process itself, identifying changes in insulin-like growth factor 1 (IGF-1) and Vascular Endothelial Growth Factor B (VEGF-B) in the affected animals. Then, we administered pharmacological treatments using human recombinant IGF-1 (rhIGF-1) or VEGF-B (rhVEGF-B) proteins, considering their temporal variations during the degenerative process. The effects of these treatments on motor, cognitive, and social behavior, as well as on cerebellar destructuration were analyzed. Whereas treatment with rhIGF-1 did not demonstrate any neuroprotective effect, rhVEGF-B administration at moderate dosages stopped the process of neuronal death and restored motor, cognitive, and social functions altered in PCD mice (and CONDCA patients). However, increasing the frequency of rhVEGF-B administration had a detrimental effect on Purkinje cell survival, suggesting an inverted U-shaped dose-response curve of this substance. Additionally, we demonstrate that this neuroprotective effect was achieved through a partial inhibition or delay of apoptosis. These findings provide strong evidence supporting the use of rhVEGF-B as a pharmacological agent to limit severe cerebellar neurodegenerative processes.
Collapse
Affiliation(s)
- Laura Pérez-Revuelta
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Pérez-Boyero
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Ester Pérez-Martín
- Neuroscience Innovative Technologies, Neurostech, 33428 Llanera, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Intervenciones Traslacionales para la Salud, 33011 Oviedo, Spain
| | - Valeria Lorena Cabedo
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Pablo González Téllez de Meneses
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Eduardo Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José Ramón Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain; (L.P.-R.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
5
|
Mercey O, Gadadhar S, Magiera MM, Lebrun L, Kostic C, Moulin A, Arsenijevic Y, Janke C, Guichard P, Hamel V. Glutamylation imbalance impairs the molecular architecture of the photoreceptor cilium. EMBO J 2024; 43:6679-6704. [PMID: 39528655 PMCID: PMC11649768 DOI: 10.1038/s44318-024-00284-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Microtubules, composed of conserved α/β-tubulin dimers, undergo complex post-translational modifications (PTMs) that fine-tune their properties and interactions with other proteins. Cilia exhibit several tubulin PTMs, such as polyglutamylation, polyglycylation, detyrosination, and acetylation, with functions that are not fully understood. Mutations in AGBL5, which encodes the deglutamylating enzyme CCP5, have been linked to retinitis pigmentosa, suggesting that altered polyglutamylation may cause photoreceptor cell degeneration, though the underlying mechanisms are unclear. Using super-resolution ultrastructure expansion microscopy (U-ExM) in mouse and human photoreceptor cells, we observed that most tubulin PTMs accumulate at the connecting cilium that links outer and inner photoreceptor segments. Mouse models with increased glutamylation (Ccp5-/- and Ccp1-/-) or loss of tubulin acetylation (Atat1-/-) showed that aberrant glutamylation, but not acetylation loss, disrupts outer segment architecture. This disruption includes exacerbation of the connecting cilium, loss of the bulge region, and destabilization of the distal axoneme. Additionally, we found significant impairment in tubulin glycylation, as well as reduced levels of intraflagellar transport proteins and of retinitis pigmentosa-associated protein RPGR. Our findings indicate that proper glutamylation levels are crucial for maintaining the molecular architecture of the photoreceptor cilium.
Collapse
Affiliation(s)
- Olivier Mercey
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Sudarshan Gadadhar
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Orsay, France
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Post, Bellary Road, Bangalore, India
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Laura Lebrun
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Corinne Kostic
- Group for Retinal Disorder Research, Department of Ophthalmology, University Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Alexandre Moulin
- Department of Ophthalmology, University Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Yvan Arsenijevic
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, Orsay, France.
- Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| | - Paul Guichard
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| | - Virginie Hamel
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
6
|
Jang EH, Choi H, Hur EM. Microtubule function and dysfunction in the nervous system. Mol Cells 2024; 47:100111. [PMID: 39265797 PMCID: PMC11474369 DOI: 10.1016/j.mocell.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
Microtubules are core components of the neuronal cytoskeleton, providing structural support for the complex cytoarchitecture of neurons and serving as tracks for long-distance transport. The properties and functions of neuronal microtubules are controlled by tubulin isoforms and a variety of post-translational modifications, collectively known as the "tubulin code." The tubulin code exerts direct control over the intrinsic properties of neuronal microtubules and regulates the repertoire of proteins that read the code, which in turn, has a significant impact on microtubule stability and dynamics. Here, we review progress in the understanding of the tubulin code in the nervous system, with a particular focus on tubulin post-translational modifications that have been proposed as potential contributors to the development and maintenance of the mammalian nervous system. Furthermore, we also discuss the potential links between disruptions in the tubulin code and neurological disorders, including neurodevelopmental abnormalities and neurodegenerative diseases.
Collapse
Affiliation(s)
- Eun-Hae Jang
- Laboratory of Neuroscience, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea
| | - Harryn Choi
- Laboratory of Neuroscience, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; BK21 Four Future Veterinary Medicine Leading Education & Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea
| | - Eun-Mi Hur
- Laboratory of Neuroscience, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea; BK21 Four Future Veterinary Medicine Leading Education & Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, South Korea; Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
7
|
Kolinko Y, Kralickova M, Cendelin J. Reduction of Microvessel Number and Length in the Cerebellum of Purkinje Cell Degeneration Mice. CEREBELLUM (LONDON, ENGLAND) 2024; 23:471-478. [PMID: 37071329 DOI: 10.1007/s12311-023-01556-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 04/19/2023]
Abstract
Degenerative effects of nerve tissues are often accompanied by changes in vascularization. In this regard, knowledge about hereditary cerebellar degeneration is limited. In this study, we compared the vascularity of the individual cerebellar components of 3-month-old wild-type mice (n = 8) and Purkinje cell degeneration (pcd) mutant mice, which represent a model of hereditary cerebellar degeneration (n = 8). Systematic random samples of tissue sections were processed, and laminin was immunostained to visualize microvessels. A computer-assisted stereology system was used to quantify microvessel parameters including total number, total length, and associated densities in cerebellar layers. Our results in pcd mice revealed a 45% (p < 0.01) reduction in the total volume of the cerebellum, a 28% (p < 0.05) reduction in the total number of vessels and a lower total length, approaching 50% (p < 0.001), compared to the control mice. In pcd mutants, cerebellar degeneration is accompanied by significant reduction in the microvascular network that is proportional to the cerebellar volume reduction therefore does not change density of in the cerebellar gray matter of pcd mice.
Collapse
Affiliation(s)
- Yaroslav Kolinko
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | - Milena Kralickova
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jan Cendelin
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
8
|
Lin Y, Wang Y, Lai T, Teng J, Lin C, Ke C, Yu I, Lee H, Chan C, Tung C, Conrad DF, O'Bryan MK, Lin Y. Deleterious genetic changes in AGTPBP1 result in teratozoospermia with sperm head and flagella defects. J Cell Mol Med 2024; 28:e18031. [PMID: 37937809 PMCID: PMC10826451 DOI: 10.1111/jcmm.18031] [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: 04/20/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/09/2023] Open
Abstract
Approximately 10%-15% of couples worldwide are infertile, and male factors account for approximately half of these cases. Teratozoospermia is a major cause of male infertility. Although various mutations have been identified in teratozoospermia, these can vary among ethnic groups. In this study, we performed whole-exome sequencing to identify genetic changes potentially causative of teratozoospermia. Out of seven genes identified, one, ATP/GTP Binding Protein 1 (AGTPBP1), was characterized, and three missense changes were identified in two patients (Affected A: p.Glu423Asp and p.Pro631Leu; Affected B: p.Arg811His). In those two cases, severe sperm head and tail defects were observed. Moreover, AGTPBP1 localization showed a fragmented pattern compared to control participants, with specific localization in the neck and annulus regions. Using murine models, we found that AGTPBP1 is localized in the manchette structure, which is essential for sperm structure formation. Additionally, in Agtpbp1-null mice, we observed sperm head and tail defects similar to those in sperm from AGTPBP1-mutated cases, along with abnormal polyglutamylation tubulin and decreasing △-2 tubulin levels. In this study, we established a link between genetic changes in AGTPBP1 and human teratozoospermia for the first time and identified the role of AGTPBP1 in deglutamination, which is crucial for sperm formation.
Collapse
Affiliation(s)
- Yu‐Hua Lin
- Division of Urology, Department of SurgeryCardinal Tien HospitalNew TaipeiTaiwan
- Department of ChemistryFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Ya‐Yun Wang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Tsung‐Hsuan Lai
- Department of Obstetrics and GynecologyCathay General HospitalTaipeiTaiwan
- School of Medicine, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Jih‐Lung Teng
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Chi‐Wei Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Chih‐Chun Ke
- Department of UrologyEn Chu Kong HospitalNew Taipei CityTaiwan
| | - I‐Shing Yu
- Laboratory Animal CenterCollege of Medicine, National Taiwan UniversityTaipeiTaiwan
| | - Hui‐Ling Lee
- Department of ChemistryFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Chying‐Chyuan Chan
- Department of Obstetrics and GynecologyTaipei City Hospital, Zhongxing Branch and Branch for Women and ChildrenTaipeiTaiwan
| | - Chi‐Hua Tung
- Program of Artificial Intelligence & Information SecurityFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Donald F. Conrad
- Division of Genetics, Oregon National Primate Research CenterBeavertonOregonUSA
| | - Moira K. O'Bryan
- School of BioSciences and Bio21 Institute, The University of MelbourneParkvilleVictoriaAustralia
| | - Ying‐Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic UniversityNew Taipei CityTaiwan
| |
Collapse
|
9
|
Zhang X, Li X, Chen W, Wang Y, Diao L, Gao Y, Wang H, Bao L, Liang X, Wu HY. The distinct initiation sites and processing activities of TTLL4 and TTLL7 in glutamylation of brain tubulin. J Biol Chem 2023; 299:104923. [PMID: 37321451 PMCID: PMC10404701 DOI: 10.1016/j.jbc.2023.104923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023] Open
Abstract
Mammalian brain tubulins undergo a reversible posttranslational modification-polyglutamylation-which attaches a secondary polyglutamate chain to the primary sequence of proteins. Loss of its erasers can disrupt polyglutamylation homeostasis and cause neurodegeneration. Tubulin tyrosine ligase like 4 (TTLL4) and TTLL7 were known to modify tubulins, both with preference for the β-isoform, but differently contribute to neurodegeneration. However, differences in their biochemical properties and functions remain largely unknown. Here, using an antibody-based method, we characterized the properties of a purified recombinant TTLL4 and confirmed its sole role as an initiator, unlike TTLL7, which both initiates and elongates the side chains. Unexpectedly, TTLL4 produced stronger glutamylation immunosignals for α-isoform than β-isoform in brain tubulins. Contrarily, the recombinant TTLL7 raised comparable glutamylation immunoreactivity for two isoforms. Given the site selectivity of the glutamylation antibody, we analyzed modification sites of two enzymes. Tandem mass spectrometry analysis revealed their incompatible site selectivity on synthetic peptides mimicking carboxyl termini of α1- and β2-tubulins and a recombinant tubulin. Particularly, in the recombinant α1A-tubulin, a novel region was found glutamylated by TTLL4 and TTLL7, that again at distinct sites. These results pinpoint different site specificities between two enzymes. Moreover, TTLL7 exhibits less efficiency to elongate microtubules premodified by TTLL4, suggesting possible regulation of TTLL7 elongation activity by TTLL4-initiated sites. Finally, we showed that kinesin behaves differentially on microtubules modified by two enzymes. This study underpins the different reactivity, site selectivity, and function of TTLL4 and TTLL7 on brain tubulins and sheds light on their distinct role in vivo.
Collapse
Affiliation(s)
- Xinyue Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xiangxiao Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Wei Chen
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yujuan Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lei Diao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Heyi Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lan Bao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xin Liang
- IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hui-Yuan Wu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
| |
Collapse
|
10
|
Pérez-Martín E, Pérez-Revuelta L, Barahona-López C, Pérez-Boyero D, Alonso JR, Díaz D, Weruaga E. Oleoylethanolamide Treatment Modulates Both Neuroinflammation and Microgliosis, and Prevents Massive Leukocyte Infiltration to the Cerebellum in a Mouse Model of Neuronal Degeneration. Int J Mol Sci 2023; 24:ijms24119691. [PMID: 37298639 DOI: 10.3390/ijms24119691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Neurodegenerative diseases involve an exacerbated neuroinflammatory response led by microglia that triggers cytokine storm and leukocyte infiltration into the brain. PPARα agonists partially dampen this neuroinflammation in some models of brain insult, but neuronal loss was not the triggering cause in any of them. This study examines the anti-inflammatory and immunomodulatory properties of the PPARα agonist oleoylethanolamide (OEA) in the Purkinje Cell Degeneration (PCD) mouse, which exhibits striking neuroinflammation caused by aggressive loss of cerebellar Purkinje neurons. Using real-time quantitative polymerase chain reaction and immunostaining, we quantified changes in pro- and anti-inflammatory markers, microglial density and marker-based phenotype, and overall leukocyte recruitment at different time points after OEA administration. OEA was found to modulate cerebellar neuroinflammation by increasing the gene expression of proinflammatory mediators at the onset of neurodegeneration and decreasing it over time. OEA also enhanced the expression of anti-inflammatory and neuroprotective factors and the Pparα gene. Regarding microgliosis, OEA reduced microglial density-especially in regions where it is preferentially located in PCD mice-and shifted the microglial phenotype towards an anti-inflammatory state. Finally, OEA prevented massive leukocyte infiltration into the cerebellum. Overall, our findings suggest that OEA may change the environment to protect neurons from degeneration caused by exacerbated inflammation.
Collapse
Affiliation(s)
- Ester Pérez-Martín
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Laura Pérez-Revuelta
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Cristina Barahona-López
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
| | - David Pérez-Boyero
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José R Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Eduardo Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
11
|
Wang Y, Zhang Y, Guo X, Zheng Y, Zhang X, Feng S, Wu HY. CCP5 and CCP6 retain CP110 and negatively regulate ciliogenesis. BMC Biol 2023; 21:124. [PMID: 37226238 DOI: 10.1186/s12915-023-01622-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/09/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND The axonemal microtubules of primary cilium undergo a conserved protein posttranslational modification (PTM) - polyglutamylation. This reversible procedure is processed by tubulin tyrosine ligase-like polyglutamylases to form secondary polyglutamate side chains, which are metabolized by the 6-member cytosolic carboxypeptidase (CCP) family. Although polyglutamylation modifying enzymes have been linked to ciliary architecture and motility, it was unknown whether they also play a role in ciliogenesis. RESULTS In this study, we found that CCP5 expression is transiently downregulated upon the initiation of ciliogenesis, but recovered after cilia are formed. Overexpression of CCP5 inhibited ciliogenesis, suggesting that a transient downregulation of CCP5 expression is required for ciliation initiation. Interestingly, the inhibitory effect of CCP5 on ciliogenesis does not rely on its enzyme activity. Among other 3 CCP members tested, only CCP6 can similarly suppress ciliogenesis. Using CoIP-MS analysis, we identified a protein that potentially interacts with CCP - CP110, a known negative regulator of ciliogenesis, whose degradation at the distal end of mother centriole permits cilia assembly. We found that both CCP5 and CCP6 can modulate CP110 level. Particularly, CCP5 interacts with CP110 through its N-terminus. Loss of CCP5 or CCP6 led to the disappearance of CP110 at the mother centriole and abnormally increased ciliation in cycling RPE-1 cells. Co-depletion of CCP5 and CCP6 synergized this abnormal ciliation, suggesting their partially overlapped function in suppressing cilia formation in cycling cells. In contrast, co-depletion of the two enzymes did not further increase the length of cilia, although CCP5 and CCP6 differentially regulate polyglutamate side-chain length of ciliary axoneme and both contribute to limiting cilia length, suggesting that they may share a common pathway in cilia length control. Through inducing the overexpression of CCP5 or CCP6 at different stages of ciliogenesis, we further demonstrated that CCP5 or CCP6 inhibited cilia formation before ciliogenesis, while shortened the length of cilia after cilia formation. CONCLUSION These findings reveal the dual role of CCP5 and CCP6. In addition to regulating cilia length, they also retain CP110 level to suppress cilia formation in cycling cells, pointing to a novel regulatory mechanism for ciliogenesis mediated by demodifying enzymes of a conserved ciliary PTM, polyglutamylation.
Collapse
Affiliation(s)
- Yujuan Wang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Building 24, Room 417-8, Tianjin, 300072, China
| | - Yuan Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Building 24, Room 417-8, Tianjin, 300072, China
| | - Xinyu Guo
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Building 24, Room 417-8, Tianjin, 300072, China
| | - Yiqiang Zheng
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Building 24, Room 417-8, Tianjin, 300072, China
| | - Xinjie Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Building 24, Room 417-8, Tianjin, 300072, China
| | - Shanshan Feng
- Key Laboratory of Regenerative Medicine, Ministry of Education, Department of Developmental and Regenerative Biology, Jinan University, Guangzhou, 51063, China
| | - Hui-Yuan Wu
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Building 24, Room 417-8, Tianjin, 300072, China.
| |
Collapse
|
12
|
Spencer WJ. Extracellular vesicles highlight many cases of photoreceptor degeneration. Front Mol Neurosci 2023; 16:1182573. [PMID: 37273908 PMCID: PMC10233141 DOI: 10.3389/fnmol.2023.1182573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
The release of extracellular vesicles is observed across numerous cell types and serves a range of biological functions including intercellular communication and waste disposal. One cell type which stands out for its robust capacity to release extracellular vesicles is the vertebrate photoreceptor cell. For decades, the release of extracellular vesicles by photoreceptors has been documented in many different animal models of photoreceptor degeneration and, more recently, in wild type photoreceptors. Here, I review all studies describing extracellular vesicle release by photoreceptors and discuss the most unifying theme among them-a photoreceptor cell fully, or partially, diverts its light sensitive membrane material to extracellular vesicles when it has defects in the delivery or morphing of this material into the photoreceptor's highly organized light sensing organelle. Because photoreceptors generate an enormous amount of light sensitive membrane every day, the diversion of this material to extracellular vesicles can cause a massive accumulation of these membranes within the retina. Little is known about the uptake of photoreceptor derived extracellular vesicles, although in some cases the retinal pigment epithelial cells, microglia, Müller glia, and/or photoreceptor cells themselves have been shown to phagocytize them.
Collapse
|
13
|
CCP1, a Regulator of Tubulin Post-Translational Modifications, Potentially Plays an Essential Role in Cerebellar Development. Int J Mol Sci 2023; 24:ijms24065335. [PMID: 36982413 PMCID: PMC10049023 DOI: 10.3390/ijms24065335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
The cytosolic carboxypeptidase (CCP) 1 protein, encoded by CCP1, is expressed in cerebellar Purkinje cells (PCs). The dysfunction of CCP1 protein (caused by CCP1 point mutation) and the deletion of CCP1 protein (caused by CCP1 gene knockout) all lead to the degeneration of cerebellar PCs, which leads to cerebellar ataxia. Thus, two CCP1 mutants (i.e., Ataxia and Male Sterility [AMS] mice and Nna1 knockout [KO] mice) are used as disease models. We investigated the cerebellar CCP1 distribution in wild-type (WT), AMS and Nna1 KO mice on postnatal days (P) 7–28 to investigate the differential effects of CCP protein deficiency and disorder on cerebellar development. Immunohistochemical and immunofluorescence studies revealed significant differences in the cerebellar CCP1 expression in WT and mutant mice of P7 and P15, but no significant difference between AMS and Nna1 KO mice. Electron microscopy showed slight abnormality in the nuclear membrane structure of PCs in the AMS and Nna1 KO mice at P15 and significant abnormality with depolymerization and fragmentation of microtubule structure at P21. Using two CCP1 mutant mice strains, we revealed the morphological changes of PCs at postnatal stages and indicated that CCP1 played an important role in cerebellar development, most likely via polyglutamylation.
Collapse
|
14
|
Hotta T, Plemmons A, Gebbie M, Ziehm TA, Blasius TL, Johnson C, Verhey KJ, Pearring JN, Ohi R. Mechanistic Analysis of CCP1 in Generating ΔC2 α-Tubulin in Mammalian Cells and Photoreceptor Neurons. Biomolecules 2023; 13:357. [PMID: 36830726 PMCID: PMC9952995 DOI: 10.3390/biom13020357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
An important post-translational modification (PTM) of α-tubulin is the removal of amino acids from its C-terminus. Removal of the C-terminal tyrosine residue yields detyrosinated α-tubulin, and subsequent removal of the penultimate glutamate residue produces ΔC2-α-tubulin. These PTMs alter the ability of the α-tubulin C-terminal tail to interact with effector proteins and are thereby thought to change microtubule dynamics, stability, and organization. The peptidase(s) that produces ΔC2-α-tubulin in a physiological context remains unclear. Here, we take advantage of the observation that ΔC2-α-tubulin accumulates to high levels in cells lacking tubulin tyrosine ligase (TTL) to screen for cytosolic carboxypeptidases (CCPs) that generate ΔC2-α-tubulin. We identify CCP1 as the sole peptidase that produces ΔC2-α-tubulin in TTLΔ HeLa cells. Interestingly, we find that the levels of ΔC2-α-tubulin are only modestly reduced in photoreceptors of ccp1-/- mice, indicating that other peptidases act synergistically with CCP1 to produce ΔC2-α-tubulin in post-mitotic cells. Moreover, the production of ΔC2-α-tubulin appears to be under tight spatial control in the photoreceptor cilium: ΔC2-α-tubulin persists in the connecting cilium of ccp1-/- but is depleted in the distal portion of the photoreceptor. This work establishes the groundwork to pinpoint the function of ΔC2-α-tubulin in proliferating and post-mitotic mammalian cells.
Collapse
Affiliation(s)
- Takashi Hotta
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexandra Plemmons
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margo Gebbie
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Trevor A. Ziehm
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Teresa Lynne Blasius
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kristen J. Verhey
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jillian N. Pearring
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryoma Ohi
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
15
|
Kumar G, Ma CHE. Toward a cerebello-thalamo-cortical computational model of spinocerebellar ataxia. Neural Netw 2023; 162:541-556. [PMID: 37023628 DOI: 10.1016/j.neunet.2023.01.045] [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: 05/04/2022] [Revised: 12/07/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Computational neural network modelling is an emerging approach for optimization of drug treatment of neurological disorders and fine-tuning of rehabilitation strategies. In the current study, we constructed a cerebello-thalamo-cortical computational neural network model to simulate a mouse model of cerebellar ataxia (pcd5J mice) by manipulating cerebellar bursts through reduction of GABAergic inhibitory input. Cerebellar output neurons were projected to the thalamus and bidirectionally connected with the cortical network. Our results showed that reduction of inhibitory input in the cerebellum orchestrated the cortical local field potential (LFP) dynamics to generate specific motor outputs of oscillations of the theta, alpha, and beta bands in the computational model as well as in mouse motor cortical neurons. The therapeutic potential of deep brain stimulation (DBS) was tested in the computational model by increasing the sensory input to restore cortical output. Ataxia mice showed normalization of the motor cortex LFP after cerebellum DBS. We provide a novel approach to computational modelling to investigate the effect of DBS by mimicking cerebellar ataxia involving degeneration of Purkinje cells. Simulated neural activity coincides with findings from neural recordings of ataxia mice. Our computational model could thus represent cerebellar pathologies and provide insight into how to improve disease symptoms by restoring neuronal electrophysiological properties using DBS.
Collapse
Affiliation(s)
- Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong Special Administrative Region.
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong Special Administrative Region.
| |
Collapse
|
16
|
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: 1.0] [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
|
17
|
Rodriguez-Calado S, Van Damme P, Avilés FX, Candiota AP, Tanco S, Lorenzo J. Proximity Mapping of CCP6 Reveals Its Association with Centrosome Organization and Cilium Assembly. Int J Mol Sci 2023; 24:ijms24021273. [PMID: 36674791 PMCID: PMC9867282 DOI: 10.3390/ijms24021273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
The cytosolic carboxypeptidase 6 (CCP6) catalyzes the deglutamylation of polyglutamate side chains, a post-translational modification that affects proteins such as tubulins or nucleosome assembly proteins. CCP6 is involved in several cell processes, such as spermatogenesis, antiviral activity, embryonic development, and pathologies like renal adenocarcinoma. In the present work, the cellular role of CCP6 has been assessed by BioID, a proximity labeling approach for mapping physiologically relevant protein-protein interactions (PPIs) and bait proximal proteins by mass spectrometry. We used HEK 293 cells stably expressing CCP6-BirA* to identify 37 putative interactors of this enzyme. This list of CCP6 proximal proteins displayed enrichment of proteins associated with the centrosome and centriolar satellites, indicating that CCP6 could be present in the pericentriolar material. In addition, we identified cilium assembly-related proteins as putative interactors of CCP6. In addition, the CCP6 proximal partner list included five proteins associated with the Joubert syndrome, a ciliopathy linked to defects in polyglutamylation. Using the proximity ligation assay (PLA), we show that PCM1, PIBF1, and NudC are true CCP6 physical interactors. Therefore, the BioID methodology confirms the location and possible functional role of CCP6 in centrosomes and centrioles, as well as in the formation and maintenance of primary cilia.
Collapse
Affiliation(s)
- Sergi Rodriguez-Calado
- Institut de Biotecnologia i Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Petra Van Damme
- iRIP Unit, Laboratory of Microbiology, Department of Biochemistry and Microbiology, Ghent University, K. L. Ledeganckstraat 35, 9000 Ghent, Belgium
| | - Francesc Xavier Avilés
- Institut de Biotecnologia i Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Ana Paula Candiota
- Institut de Biotecnologia i Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Sebastian Tanco
- Institut de Biotecnologia i Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
- Correspondence: (S.T.); (J.L.); Tel.: +34-93-586-8938 (S.T.); +34-93-586-8957 (J.L.)
| | - Julia Lorenzo
- Institut de Biotecnologia i Biomedicina, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
- Correspondence: (S.T.); (J.L.); Tel.: +34-93-586-8938 (S.T.); +34-93-586-8957 (J.L.)
| |
Collapse
|
18
|
Preclinical Models of Retinitis Pigmentosa. Methods Mol Biol 2022; 2560:181-215. [PMID: 36481897 DOI: 10.1007/978-1-0716-2651-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Retinitis pigmentosa (RP) is the name for a group of phenotypically-related heritable retinal degenerative disorders. Many genes have been implicated as causing variants of RP, and while the clinical phenotypes are remarkably similar, they may differ in age of onset, progression, and severity. Common inheritance patterns for specific genes connected with the development of the disorder include autosomal dominant, autosomal recessive, and X-linked. Modeling the disease in animals and other preclinical systems offers a cost-conscious, ethical, and time-efficient method for studying the disease subtypes. The history of RP models is briefly examined, and both naturally occurring and transgenic preclinical models of RP in many different organisms are discussed. Syndromic forms of RP and models thereof are reviewed as well.
Collapse
|
19
|
Lewis TR, Phan S, Kim KY, Jha I, Castillo CM, Ding JD, Sajdak BS, Merriman DK, Ellisman MH, Arshavsky VY. Microvesicle release from inner segments of healthy photoreceptors is a conserved phenomenon in mammalian species. Dis Model Mech 2022; 15:dmm049871. [PMID: 36420970 PMCID: PMC9796728 DOI: 10.1242/dmm.049871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022] Open
Abstract
Many inherited visual diseases arise from mutations that affect the structure and function of photoreceptor cells. In some cases, the pathology is accompanied by a massive release of extracellular vesicles from affected photoreceptors. In this study, we addressed whether vesicular release is an exclusive response to ongoing pathology or a normal homeostatic phenomenon amplified in disease. We analyzed the ultrastructure of normal photoreceptors from both rod- and cone-dominant mammalian species and found that these cells release microvesicles budding from their inner segment compartment. Inner segment-derived microvesicles vary in their content, with some of them containing the visual pigment rhodopsin and others appearing to be interconnected with mitochondria. These data suggest the existence of a fundamental process whereby healthy mammalian photoreceptors release mistrafficked or damaged inner segment material as microvesicles into the interphotoreceptor space. This release may be greatly enhanced under pathological conditions associated with defects in protein targeting and trafficking. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tylor R. Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sebastien Phan
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Isha Jha
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Carson M. Castillo
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jin-Dong Ding
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | - Benjamin S. Sajdak
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, WI 54901, USA
- Fauna Bio Inc., Emeryville, CA 94608, USA
| | - Dana K. Merriman
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, WI 54901, USA
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Vadim Y. Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
20
|
Wu HY, Rong Y, Bansal PK, Wei P, Guo H, Morgan JI. TTLL1 and TTLL4 polyglutamylases are required for the neurodegenerative phenotypes in pcd mice. PLoS Genet 2022; 18:e1010144. [PMID: 35404950 PMCID: PMC9022812 DOI: 10.1371/journal.pgen.1010144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/21/2022] [Accepted: 03/14/2022] [Indexed: 02/01/2023] Open
Abstract
Polyglutamylation is a dynamic posttranslational modification where glutamate residues are added to substrate proteins by 8 tubulin tyrosine ligase-like (TTLL) family members (writers) and removed by the 6 member Nna1/CCP family of carboxypeptidases (erasers). Genetic disruption of polyglutamylation leading to hyperglutamylation causes neurodegenerative phenotypes in humans and animal models; the best characterized being the Purkinje cell degeneration (pcd) mouse, a mutant of the gene encoding Nna1/CCP1, the prototypic eraser. Emphasizing the functional importance of the balance between glutamate addition and elimination, loss of TTLL1 prevents Purkinje cell degeneration in pcd. However, whether Ttll1 loss protects other vulnerable neurons in pcd, or if elimination of other TTLLs provides protection is largely unknown. Here using a mouse genetic rescue strategy, we characterized the contribution of Ttll1, 4, 5, 7, or 11 to the degenerative phenotypes in cerebellum, olfactory bulb and retinae of pcd mutants. Ttll1 deficiency attenuates Purkinje cell loss and function and reduces olfactory bulb mitral cell death and retinal photoreceptor degeneration. Moreover, degeneration of photoreceptors in pcd is preceded by impaired rhodopsin trafficking to the rod outer segment and likely represents the causal defect leading to degeneration as this too is rescued by elimination of TTLL1. Although TTLLs have similar catalytic properties on model substrates and several are highly expressed in Purkinje cells (e.g. TTLL5 and 7), besides TTLL1 only TTLL4 deficiency attenuated degeneration of Purkinje and mitral cells in pcd. Additionally, TTLL4 loss partially rescued photoreceptor degeneration and impaired rhodopsin trafficking. Despite their common properties, the polyglutamylation profile changes promoted by TTLL1 and TTLL4 deficiencies in pcd mice are very different. We also report that loss of anabolic TTLL5 synergizes with loss of catabolic Nna1/CCP1 to promote photoreceptor degeneration. Finally, male infertility in pcd is not rescued by loss of any Ttll. These data provide insight into the complexity of polyglutamate homeostasis and function in vivo and potential routes to ameliorate disorders caused by disrupted polyglutamylation.
Collapse
Affiliation(s)
- Hui-Yuan Wu
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yongqi Rong
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Parmil K. Bansal
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Peng Wei
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hong Guo
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - James I. Morgan
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
21
|
Lalonde R, Strazielle C. The AGTPBP1 gene in neurobiology. Gene 2022; 809:146001. [PMID: 34637898 DOI: 10.1016/j.gene.2021.146001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/04/2022]
Abstract
The function of the Agtpbp1 gene has mainly been delineated by studying Agtpbp1pcd (pcd) mutant mice, characterized by losses in cerebellar Purkinje and granule cells along with degeneration of retinal photoreceptors, mitral cells of the olfactory bulb, thalamic neurons, and alpha-motoneurons. As a result of cerebellar degeneration, cerebellar GABA and glutamate concentrations in Agtpbp1pcd mutants decreased while monoamine concentrations increased. The salient behavioral phenotypes include cerebellar ataxia, a loss in motor coordination, and cognitive deficits. Similar neuropathogical and behavioral profiles have been described in childhood-onset human subjects with biallelic variants of AGTPBP1, including cerebellar ataxia and hypotonia.
Collapse
Affiliation(s)
- Robert Lalonde
- University of Rouen, Dept Psychology, 76821 Mont-Saint-Aignan, France; Laboratory of Stress, Immunity, Pathogens (EA7300), University of Lorraine Medical School, Vandœuvre-les-Nancy, France.
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, Pathogens (EA7300), University of Lorraine Medical School, Vandœuvre-les-Nancy, France; CHRU Nancy, Vandœuvre-les-Nancy, France
| |
Collapse
|
22
|
Kiyozumi D, Ikawa M. Proteolysis in Reproduction: Lessons From Gene-Modified Organism Studies. Front Endocrinol (Lausanne) 2022; 13:876370. [PMID: 35600599 PMCID: PMC9114714 DOI: 10.3389/fendo.2022.876370] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/28/2022] [Indexed: 12/17/2022] Open
Abstract
The physiological roles of proteolysis are not limited to degrading unnecessary proteins. Proteolysis plays pivotal roles in various biological processes through cleaving peptide bonds to activate and inactivate proteins including enzymes, transcription factors, and receptors. As a wide range of cellular processes is regulated by proteolysis, abnormalities or dysregulation of such proteolytic processes therefore often cause diseases. Recent genetic studies have clarified the inclusion of proteases and protease inhibitors in various reproductive processes such as development of gonads, generation and activation of gametes, and physical interaction between gametes in various species including yeast, animals, and plants. Such studies not only clarify proteolysis-related factors but the biological processes regulated by proteolysis for successful reproduction. Here the physiological roles of proteases and proteolysis in reproduction will be reviewed based on findings using gene-modified organisms.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
| |
Collapse
|
23
|
Baltanás FC, Berciano MT, Santos E, Lafarga M. The Childhood-Onset Neurodegeneration with Cerebellar Atrophy (CONDCA) Disease Caused by AGTPBP1 Gene Mutations: The Purkinje Cell Degeneration Mouse as an Animal Model for the Study of this Human Disease. Biomedicines 2021; 9:biomedicines9091157. [PMID: 34572343 PMCID: PMC8464709 DOI: 10.3390/biomedicines9091157] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 12/20/2022] Open
Abstract
Recent reports have identified rare, biallelic damaging variants of the AGTPBP1 gene that cause a novel and documented human disease known as childhood-onset neurodegeneration with cerebellar atrophy (CONDCA), linking loss of function of the AGTPBP1 protein to human neurodegenerative diseases. CONDCA patients exhibit progressive cognitive decline, ataxia, hypotonia or muscle weakness among other clinical features that may be fatal. Loss of AGTPBP1 in humans recapitulates the neurodegenerative course reported in a well-characterised murine animal model harbouring loss-of-function mutations in the AGTPBP1 gene. In particular, in the Purkinje cell degeneration (pcd) mouse model, mutations in AGTPBP1 lead to early cerebellar ataxia, which correlates with the massive loss of cerebellar Purkinje cells. In addition, neurodegeneration in the olfactory bulb, retina, thalamus and spinal cord were also reported. In addition to neurodegeneration, pcd mice show behavioural deficits such as cognitive decline. Here, we provide an overview of what is currently known about the structure and functional role of AGTPBP1 and discuss the various alterations in AGTPBP1 that cause neurodegeneration in the pcd mutant mouse and humans with CONDCA. The sequence of neuropathological events that occur in pcd mice and the mechanisms governing these neurodegenerative processes are also reported. Finally, we describe the therapeutic strategies that were applied in pcd mice and focus on the potential usefulness of pcd mice as a promising model for the development of new therapeutic strategies for clinical trials in humans, which may offer potential beneficial options for patients with AGTPBP1 mutation-related CONDCA.
Collapse
Affiliation(s)
- Fernando C. Baltanás
- Lab.1, CIC-IBMCC, University of Salamanca-CSIC and CIBERONC, 37007 Salamanca, Spain;
- Correspondence: ; Tel.: +34-923294801
| | - María T. Berciano
- Department of Molecular Biology and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Cantabria-IDIVAL, 39011 Santander, Spain;
| | - Eugenio Santos
- Lab.1, CIC-IBMCC, University of Salamanca-CSIC and CIBERONC, 37007 Salamanca, Spain;
| | - Miguel Lafarga
- Department of Anatomy and Cell Biology and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), University of Cantabria-IDIVAL, 39011 Santander, Spain;
| |
Collapse
|
24
|
Bodakuntla S, Yuan X, Genova M, Gadadhar S, Leboucher S, Birling M, Klein D, Martini R, Janke C, Magiera MM. Distinct roles of α- and β-tubulin polyglutamylation in controlling axonal transport and in neurodegeneration. EMBO J 2021; 40:e108498. [PMID: 34309047 PMCID: PMC8408597 DOI: 10.15252/embj.2021108498] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
Tubulin polyglutamylation is a post-translational modification of the microtubule cytoskeleton, which is generated by a variety of enzymes with different specificities. The "tubulin code" hypothesis predicts that modifications generated by specific enzymes selectively control microtubule functions. Our recent finding that excessive accumulation of polyglutamylation in neurons causes their degeneration and perturbs axonal transport provides an opportunity for testing this hypothesis. By developing novel mouse models and a new glutamylation-specific antibody, we demonstrate here that the glutamylases TTLL1 and TTLL7 generate unique and distinct glutamylation patterns on neuronal microtubules. We find that under physiological conditions, TTLL1 polyglutamylates α-tubulin, while TTLL7 modifies β-tubulin. TTLL1, but not TTLL7, catalyses the excessive hyperglutamylation found in mice lacking the deglutamylase CCP1. Consequently, deletion of TTLL1, but not of TTLL7, prevents degeneration of Purkinje cells and of myelinated axons in peripheral nerves in these mice. Moreover, loss of TTLL1 leads to increased mitochondria motility in neurons, while loss of TTLL7 has no such effect. By revealing how specific patterns of tubulin glutamylation, generated by distinct enzymes, translate into specific physiological and pathological readouts, we demonstrate the relevance of the tubulin code for homeostasis.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
- Present address:
National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Xidi Yuan
- Department of NeurologyDevelopmental NeurobiologyUniversity Hospital WürzburgWürzburgGermany
| | - Mariya Genova
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Sudarshan Gadadhar
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Sophie Leboucher
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Marie‐Christine Birling
- CELPHEDIA, PHENOMINInstitut Clinique de la Souris (ICS), CNRS, INSERMUniversity of StrasbourgIllkirchFrance
| | - Dennis Klein
- Department of NeurologyDevelopmental NeurobiologyUniversity Hospital WürzburgWürzburgGermany
| | - Rudolf Martini
- Department of NeurologyDevelopmental NeurobiologyUniversity Hospital WürzburgWürzburgGermany
| | - Carsten Janke
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Maria M Magiera
- Institut CurieUniversité PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| |
Collapse
|
25
|
Türay S, Eröz R, Başak AN. A novel pathogenic variant in the 3' end of the AGTPBP1 gene gives rise to neurodegeneration without cerebellar atrophy: an expansion of the disease phenotype? Neurogenetics 2021; 22:127-132. [PMID: 33909173 DOI: 10.1007/s10048-021-00643-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/20/2021] [Indexed: 11/28/2022]
Abstract
Childhood-onset neurodegeneration with cerebellar atrophy (CONDCA) is a recently described form of the large group of infantile hereditary lower motor neuron diseases (Teoh et al. 2017), resulting from biallelic damaging variants in the AGTPBP1 gene, first described by Shashi et al. in EMBO J 37(23):e100540, 2018. AGTPBP-related neurodegeneration is a severe neurodevelopmental disorder that progresses with global developmental delay and intellectual disability, often accompanied with peripheral nerve damage and lower motor degeneration and a fatal course in the early years of life. The encoded protein is ATP/GTP-Binding Protein1, also known as cytosolic carboxypeptidase 1 (CCP1) or nervous system nuclear protein induced by axotomy (NNA1). Here we report a consanguineous family with four offspring, two of whom are affected. The index patient is a 21-month-old male with global developmental delay and hypotonia. The proband's 17-year-old sister, diagnosed with cerebral palsy, had severe hypotonia accompanied by motor and cognitive retardation. WES analysis revealed a novel homozygous c.3293G > A variant in the AGTPBP1 gene with high pathogenicity scores. Targeted Sanger sequencing confirmed the variant in both affected children and in heterozygous form in the parents. The affected siblings present with hypotonia and motor and cognitive retardation, in line with the studies previously reported. However, in our patients, no signs of cerebellar atrophy in cranial MRI were present, so the acronym CONDCA is not applicable; lower motor neuron findings were also absent. The matching and distinguishing aspects of our patients will add to the present literature and expand our understanding of this rare genetic neurodegenerative disease of early childhood.
Collapse
Affiliation(s)
- Sevim Türay
- Department of Pediatric Neurology, Düzce University Medical Faculty, Duzce, 81820, Turkey.
| | - Recep Eröz
- Department of Medical Genetics, Düzce University Medical Faculty, Duzce, Turkey
| | - A Nazlı Başak
- Suna and İnan Kıraç Foundation, Neurodegeneration Research Laboratory, NDAL-KUTTAM, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
26
|
CCP1, a Tubulin Deglutamylase, Increases Survival of Rodent Spinal Cord Neurons following Glutamate-Induced Excitotoxicity. eNeuro 2021; 8:ENEURO.0431-20.2021. [PMID: 33688040 PMCID: PMC8021396 DOI: 10.1523/eneuro.0431-20.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/25/2021] [Accepted: 02/12/2021] [Indexed: 01/21/2023] Open
Abstract
Microtubules (MTs) are cytoskeletal elements that provide structural support and act as roadways for intracellular transport in cells. MTs are also needed for neurons to extend and maintain long axons and dendrites that establish connectivity to transmit information through the nervous system. Therefore, in neurons, the ability to independently regulate cytoskeletal stability and MT-based transport in different cellular compartments is essential. Posttranslational modification of MTs is one mechanism by which neurons regulate the cytoskeleton. The carboxypeptidase CCP1 negatively regulates posttranslational polyglutamylation of MTs. In mammals, loss of CCP1, and the resulting hyperglutamylation of MTs, causes neurodegeneration. It has also long been known that CCP1 expression is activated by neuronal injury; however, whether CCP1 plays a neuroprotective role after injury is unknown. Using shRNA-mediated knock-down of CCP1 in embryonic rat spinal cord cultures, we demonstrate that CCP1 protects spinal cord neurons from excitotoxic death. Unexpectedly, excitotoxic injury reduced CCP1 expression in our system. We previously demonstrated that the CCP1 homolog in Caenorhabditis elegans is important for maintenance of neuronal cilia. Although cilia enhance neuronal survival in some contexts, it is not yet clear whether CCP1 maintains cilia in mammalian spinal cord neurons. We found that knock-down of CCP1 did not result in loss or shortening of cilia in cultured spinal cord neurons, suggesting that its effect on survival of excitotoxicity is independent of cilia. Our results support the idea that enzyme regulators of MT polyglutamylation might be therapeutically targeted to prevent excitotoxic death after spinal cord injuries.
Collapse
|
27
|
Yang WT, Hong SR, He K, Ling K, Shaiv K, Hu J, Lin YC. The Emerging Roles of Axonemal Glutamylation in Regulation of Cilia Architecture and Functions. Front Cell Dev Biol 2021; 9:622302. [PMID: 33748109 PMCID: PMC7970040 DOI: 10.3389/fcell.2021.622302] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cilia, which either generate coordinated motion or sense environmental cues and transmit corresponding signals to the cell body, are highly conserved hair-like structures that protrude from the cell surface among diverse species. Disruption of ciliary functions leads to numerous human disorders, collectively referred to as ciliopathies. Cilia are mechanically supported by axonemes, which are composed of microtubule doublets. It has been recognized for several decades that tubulins in axonemes undergo glutamylation, a post-translational polymodification, that conjugates glutamic acid chains onto the C-terminal tail of tubulins. However, the physiological roles of axonemal glutamylation were not uncovered until recently. This review will focus on how cells modulate glutamylation on ciliary axonemes and how axonemal glutamylation regulates cilia architecture and functions, as well as its physiological importance in human health. We will also discuss the conventional and emerging new strategies used to manipulate glutamylation in cilia.
Collapse
Affiliation(s)
- Wen-Ting Yang
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - Shi-Rong Hong
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - Kai He
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Kritika Shaiv
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - JingHua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
- Department of Medical Science, National Tsing Hua University, HsinChu City, Taiwan
| |
Collapse
|
28
|
Bodakuntla S, Janke C, Magiera MM. Tubulin polyglutamylation, a regulator of microtubule functions, can cause neurodegeneration. Neurosci Lett 2021; 746:135656. [PMID: 33482309 DOI: 10.1016/j.neulet.2021.135656] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases lead to a progressive demise of neuronal functions that ultimately results in neuronal death. Besides a large variety of molecular pathways that have been linked to the degeneration of neurons, dysfunctions of the microtubule cytoskeleton are common features of many human neurodegenerative disorders. Yet, it is unclear whether microtubule dysfunctions are causative, or mere bystanders in the disease progression. A so-far little explored regulatory mechanism of the microtubule cytoskeleton, the posttranslational modifications of tubulin, emerge as candidate mechanisms involved in neuronal dysfunction, and thus, degeneration. Here we review the role of tubulin polyglutamylation, a prominent modification of neuronal microtubules. We discuss the current understanding of how polyglutamylation controls microtubule functions in healthy neurons, and how deregulation of this modification leads to neurodegeneration in mice and humans.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| |
Collapse
|
29
|
Alteration of Neural Stem Cell Functions in Ataxia and Male Sterility Mice: A Possible Role of β-Tubulin Glutamylation in Neurodegeneration. Cells 2021; 10:cells10010155. [PMID: 33466875 PMCID: PMC7830091 DOI: 10.3390/cells10010155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/20/2022] Open
Abstract
Ataxia and Male Sterility (AMS) is a mutant mouse strain that contains a missense mutation in the coding region of Nna1, a gene that encodes a deglutamylase. AMS mice exhibit early cerebellar Purkinje cell degeneration and an ataxic phenotype in an autosomal recessive manner. To understand the underlying mechanism, we generated neuronal stem cell (NSC) lines from wild-type (NMW7), Nna1 mutation heterozygous (NME), and Nna1 mutation homozygous (NMO1) mouse brains. The NNA1 levels were decreased, and the glutamylated tubulin levels were increased in NMO1 cultures as well as in the cerebellum of AMS mice at both 15 and 30 days of age. However, total β-tubulin protein levels were not altered in the AMS cerebellum. In NMO1 neurosphere cultures, β-tubulin protein levels were increased without changes at the transcriptional level. NMO1 grew faster than other NSC lines, and some of the neurospheres were attached to the plate after 3 days. Immunostaining revealed that SOX2 and nestin levels were decreased in NMO1 neurospheres and that the neuronal differentiation potentials were reduced in NMO1 cells compared to NME or NMW7 cells. These results demonstrate that the AMS mutation decreased the NNA1 levels and increased glutamylation in the cerebellum of AMS mice. The observed changes in glutamylation might alter NSC properties and the neuron maturation process, leading to Purkinje cell death in AMS mice.
Collapse
|
30
|
Wang R, Lin L, Zheng Y, Cao P, Yuchi Z, Wu HY. Identification of 2-PMPA as a novel inhibitor of cytosolic carboxypeptidases. Biochem Biophys Res Commun 2020; 533:1393-1399. [PMID: 33092792 DOI: 10.1016/j.bbrc.2020.10.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 11/30/2022]
Abstract
Cytosolic carboxypeptidases (CCPs) comprise a unique subfamily of M14 carboxypeptidases and are erasers of the reversible protein posttranslational modification- polyglutamylation. Potent inhibitors for CCPs may serve as leading compounds targeting imbalanced polyglutamylation. However, no efficient CCP inhibitor has yet been reported. Here, we showed that 2-phosphonomethylpentanedioic acid (2-PMPA), a potent inhibitor of the distant M28 family member glutamate carboxypeptidase II (GCPII), rather than the typical M14 inhibitor 2-benzylsuccinic acid, could efficiently inhibit CCP activities. 2-PMPA inhibited the recombinant Nna1 (a.k.a. CCP1) for hydrolyzing a synthetic peptide in a mixed manner, with Ki and Ki' being 0.11 μM and 0.24 μM respectively. It inhibited Nna1 for deglutamylating tubulin, the best-known polyglutamylated protein, with an IC50 of 0.21 mM. Homology modeling predicted that the R-form of 2-PMPA is more favorable to bind Nna1, unlike that GCPII prefers to S-form. This work for the first time identified a potent inhibitor for CCP family.
Collapse
Affiliation(s)
- Ruixue Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Lianyun Lin
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Yiqiang Zheng
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Peng Cao
- Key Laboratory of Drug Targets and Drug Leads for Degenerative Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhiguang Yuchi
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China.
| | - Hui-Yuan Wu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
31
|
Expression of ATP/GTP Binding Protein 1 Has Prognostic Value for the Clinical Outcomes in Non-Small Cell Lung Carcinoma. J Pers Med 2020; 10:jpm10040263. [PMID: 33276627 PMCID: PMC7761608 DOI: 10.3390/jpm10040263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
ATP/GTP binding protein 1 (AGTPBP1) encodes a crucial protein, cytosolic carboxypeptidase 1 (CCP1), which plays a role in modulating the polyglutamylation of tubulin and has been studied in degenerative diseases. However, the role of AGTPBP1 in malignancy has not been completely studied yet. In this study, we examined the role of AGTPBP1 in cancer progression, its association with patient survival, and related mechanisms in lung cancer, using the A549 cell line and lung cancer gene expression datasets. AGTPBP1 knockdown increased the proliferation, migration, sphere formation, and drug resistance of A549 cells. Lung cancer datasets revealed significantly lower mRNA and protein expression levels of AGTPBP1 in lung cancer tissues, as compared to those in normal tissues. Importantly, AGTPBP1 expression positively correlated with patient survival. Analysis of co-expressed genes revealed that AGTPBP1 expression positively correlated with immune infiltration in lung cancer. Our results conclusively suggested that AGTPBP1 expression was correlated with cancer progression and immune infiltration in lung cancer.
Collapse
|
32
|
Li J, Snyder EY, Tang FHF, Pasqualini R, Arap W, Sidman RL. Nna1 gene deficiency triggers Purkinje neuron death by tubulin hyperglutamylation and ER dysfunction. JCI Insight 2020; 5:136078. [PMID: 33004692 PMCID: PMC7566705 DOI: 10.1172/jci.insight.136078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Posttranslational glutamylation/deglutamylation balance in tubulins influences dendritic maturation and neuronal survival of cerebellar Purkinje neurons (PNs). PNs and some additional neuronal types degenerate in several spontaneous, independently occurring Purkinje cell degeneration (pcd) mice featuring mutant neuronal nuclear protein induced by axotomy (Nna1), a deglutamylase gene. This defective deglutamylase allows glutamylases to form hyperglutamylated tubulins. In pcd, all PNs die during postnatal “adolescence.” Neurons in some additional brain regions also die, mostly later than PNs. We show in laser capture microdissected single PNs, in cerebellar granule cell neuronal clusters, and in dissected hippocampus and substantia nigra that deglutamase mRNA and protein were virtually absent before pcd PNs degenerated, whereas glutaminase mRNA and protein remained normal. Hyperglutamylated microtubules and dimeric tubulins accumulated in pcd PNs and were involved in pcd PN death by glutamylase/deglutamylase imbalance. Importantly, treatment with a microtubule depolymerizer corrected the glutamylation/deglutamylation ratio, increasing PN survival. Further, before onset of neuronal death, pcd PNs displayed prominent basal polylisosomal masses rich in ER. We propose a “seesaw” metamorphic model summarizing mutant Nna1-induced tubulin hyperglutamylation, the pcd’s PN phenotype, and report that the neuronal disorder involved ER stress, unfolded protein response, and protein synthesis inhibition preceding PN death by apoptosis/necroptosis. Purkinje cell degeneration is due to ER stress, unfolded protein response, and protein synthesis inhibition preceding Purkinje neuron death by apoptosis/necroptosis.
Collapse
Affiliation(s)
- Jianxue Li
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Evan Y Snyder
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Fenny HF Tang
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey and Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey and Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Richard L Sidman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Power KM, Akella JS, Gu A, Walsh JD, Bellotti S, Morash M, Zhang W, Ramadan YH, Ross N, Golden A, Smith HE, Barr MM, O’Hagan R. Mutation of NEKL-4/NEK10 and TTLL genes suppress neuronal ciliary degeneration caused by loss of CCPP-1 deglutamylase function. PLoS Genet 2020; 16:e1009052. [PMID: 33064774 PMCID: PMC7592914 DOI: 10.1371/journal.pgen.1009052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/28/2020] [Accepted: 08/14/2020] [Indexed: 12/29/2022] Open
Abstract
Ciliary microtubules are subject to post-translational modifications that act as a "Tubulin Code" to regulate motor traffic, binding proteins and stability. In humans, loss of CCP1, a cytosolic carboxypeptidase and tubulin deglutamylating enzyme, causes infantile-onset neurodegeneration. In C. elegans, mutations in ccpp-1, the homolog of CCP1, result in progressive degeneration of neuronal cilia and loss of neuronal function. To identify genes that regulate microtubule glutamylation and ciliary integrity, we performed a forward genetic screen for suppressors of ciliary degeneration in ccpp-1 mutants. We isolated the ttll-5(my38) suppressor, a mutation in a tubulin tyrosine ligase-like glutamylase gene. We show that mutation in the ttll-4, ttll-5, or ttll-11 gene suppressed the hyperglutamylation-induced loss of ciliary dye filling and kinesin-2 mislocalization in ccpp-1 cilia. We also identified the nekl-4(my31) suppressor, an allele affecting the NIMA (Never in Mitosis A)-related kinase NEKL-4/NEK10. In humans, NEK10 mutation causes bronchiectasis, an airway and mucociliary transport disorder caused by defective motile cilia. C. elegans NEKL-4 localizes to the ciliary base but does not localize to cilia, suggesting an indirect role in ciliary processes. This work defines a pathway in which glutamylation, a component of the Tubulin Code, is written by TTLL-4, TTLL-5, and TTLL-11; is erased by CCPP-1; is read by ciliary kinesins; and its downstream effects are modulated by NEKL-4 activity. Identification of regulators of microtubule glutamylation in diverse cellular contexts is important to the development of effective therapies for disorders characterized by changes in microtubule glutamylation. By identifying C. elegans genes important for neuronal and ciliary stability, our work may inform research into the roles of the tubulin code in human ciliopathies and neurodegenerative diseases.
Collapse
Affiliation(s)
- Kade M. Power
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Jyothi S. Akella
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Amanda Gu
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Jonathon D. Walsh
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Sebastian Bellotti
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Margaret Morash
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Winnie Zhang
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Yasmin H. Ramadan
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Nicole Ross
- Biology Department, Montclair State University, Montclair, NJ, United States of America
| | - Andy Golden
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Harold E. Smith
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maureen M. Barr
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States of America
| | - Robert O’Hagan
- Biology Department, Montclair State University, Montclair, NJ, United States of America
| |
Collapse
|
34
|
The emerging role of tubulin posttranslational modifications in cilia and ciliopathies. BIOPHYSICS REPORTS 2020. [DOI: 10.1007/s41048-020-00111-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
35
|
Machado AS, Marques HG, Duarte DF, Darmohray DM, Carey MR. Shared and specific signatures of locomotor ataxia in mutant mice. eLife 2020; 9:55356. [PMID: 32718435 PMCID: PMC7386913 DOI: 10.7554/elife.55356] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/09/2020] [Indexed: 01/30/2023] Open
Abstract
Several spontaneous mouse mutants with deficits in motor coordination and associated cerebellar neuropathology have been described. Intriguingly, both visible gait alterations and neuroanatomical abnormalities throughout the brain differ across mutants. We previously used the LocoMouse system to quantify specific deficits in locomotor coordination in mildly ataxic Purkinje cell degeneration mice (pcd; Machado et al., 2015). Here, we analyze the locomotor behavior of severely ataxic reeler mutants and compare and contrast it with that of pcd. Despite clearly visible gait differences, direct comparison of locomotor kinematics and linear discriminant analysis reveal a surprisingly similar pattern of impairments in multijoint, interlimb, and whole-body coordination in the two mutants. These findings capture both shared and specific signatures of gait ataxia and provide a quantitative foundation for mapping specific locomotor impairments onto distinct neuropathologies in mice.
Collapse
Affiliation(s)
- Ana S Machado
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Hugo G Marques
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Diogo F Duarte
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Dana M Darmohray
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Megan R Carey
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| |
Collapse
|
36
|
Amargant F, Barragan M, Vassena R, Vernos I. Insights of the tubulin code in gametes and embryos: from basic research to potential clinical applications in humans†. Biol Reprod 2020; 100:575-589. [PMID: 30247519 DOI: 10.1093/biolre/ioy203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 07/05/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
Microtubules are intracellular filaments that define in space and in time a large number of essential cellular functions such as cell division, morphology and motility, intracellular transport and flagella and cilia assembly. They are therefore essential for spermatozoon and oocyte maturation and function, and for embryo development. The dynamic and functional properties of the microtubules are in large part defined by various classes of interacting proteins including MAPs (microtubule associated proteins), microtubule-dependent motors, and severing and modifying enzymes. Multiple mechanisms regulate these interactions. One of them is defined by the high diversity of the microtubules themselves generated by the combination of different tubulin isotypes and by several tubulin post-translational modifications (PTMs). This generates a so-called tubulin code that finely regulates the specific set of proteins that associates with a given microtubule thereby defining the properties and functions of the network. Here we provide an in depth review of the current knowledge on the tubulin isotypes and PTMs in spermatozoa, oocytes, and preimplantation embryos in various model systems and in the human species. We focus on functional implications of the tubulin code for cytoskeletal function, particularly in the field of human reproduction and development, with special emphasis on gamete quality and infertility. Finally, we discuss some of the knowledge gaps and propose future research directions.
Collapse
Affiliation(s)
- Farners Amargant
- Clínica EUGIN, Barcelona, Spain.,Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | | | - Isabelle Vernos
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
37
|
Embryonic Cerebellar Graft Morphology Differs in Two Mouse Models of Cerebellar Degeneration. THE CEREBELLUM 2020; 18:855-865. [PMID: 31418135 DOI: 10.1007/s12311-019-01067-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cerebellar diseases causing substantial cell loss often lead to severe functional deficits and restoration of cerebellar function is difficult. Neurotransplantation therapy could become a hopeful method, but there are still many limitations and unknown aspects. Studies in a variety of cerebellar mutant mice reflecting heterogeneity of human cerebellar degenerations show promising results as well as new problems and questions to be answered. The aim of this work was to compare the development of embryonic cerebellar grafts in adult B6CBA Lurcher and B6.BR pcd mutant mice and strain-matched healthy wild type mice. Performance in the rotarod test, graft survival, structure, and volume was examined 2 months after the transplantation or sham-operation. The grafts survived in most of the mice of all types. In both B6CBA and B6.BR wild type mice and in pcd mice, colonization of the host's cerebellum was a common finding, while in Lurcher mice, the grafts showed a low tendency to infiltrate the host's cerebellar tissue. There were no significant differences in graft volume between mutant and wild type mice. Nevertheless, B6CBA mice had smaller grafts than their B6.BR counterparts. The transplantation did not improve the performance in the rotarod test. The study showed marked differences in graft integration into the host's cerebellum in two types of cerebellar mutants, suggesting disease-specific factors influencing graft fate.
Collapse
|
38
|
The tubulin code and its role in controlling microtubule properties and functions. Nat Rev Mol Cell Biol 2020; 21:307-326. [PMID: 32107477 DOI: 10.1038/s41580-020-0214-3] [Citation(s) in RCA: 496] [Impact Index Per Article: 99.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Microtubules are core components of the eukaryotic cytoskeleton with essential roles in cell division, shaping, motility and intracellular transport. Despite their functional heterogeneity, microtubules have a highly conserved structure made from almost identical molecular building blocks: the tubulin proteins. Alternative tubulin isotypes and a variety of post-translational modifications control the properties and functions of the microtubule cytoskeleton, a concept known as the 'tubulin code'. Here we review the current understanding of the molecular components of the tubulin code and how they impact microtubule properties and functions. We discuss how tubulin isotypes and post-translational modifications control microtubule behaviour at the molecular level and how this translates into physiological functions at the cellular and organism levels. We then go on to show how fine-tuning of microtubule function by some tubulin modifications can affect homeostasis and how perturbation of this fine-tuning can lead to a range of dysfunctions, many of which are linked to human disease.
Collapse
|
39
|
Lalonde R, Strazielle C. Motor Performances of Spontaneous and Genetically Modified Mutants with Cerebellar Atrophy. THE CEREBELLUM 2019; 18:615-634. [PMID: 30820866 DOI: 10.1007/s12311-019-01017-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chance discovery of spontaneous mutants with atrophy of the cerebellar cortex has unearthed genes involved in optimizing motor coordination. Rotorod, stationary beam, and suspended wire tests are useful in delineating behavioral phenotypes of spontaneous mutants with cerebellar atrophy such as Grid2Lc, Grid2ho, Rorasg, Agtpbp1pcd, Relnrl, and Dab1scm. Likewise, transgenic or null mutants serving as experimental models of spinocerebellar ataxia (SCA) are phenotyped with the same tests. Among experimental models of autosomal dominant SCA, rotorod deficits were reported in SCA1 to 3, SCA5 to 8, SCA14, SCA17, and SCA27 and stationary beam deficits in SCA1 to 3, SCA5, SCA6, SCA13, SCA17, and SCA27. Beam tests are sensitive to experimental therapies of various kinds including molecules affecting glutamate signaling, mesenchymal stem cells, anti-oligomer antibodies, lentiviral vectors carrying genes, interfering RNAs, or neurotrophic factors, and interbreeding with other mutants.
Collapse
Affiliation(s)
- Robert Lalonde
- Department of Psychology, University of Rouen, 76821, Mont-Saint-Aignan Cedex, France.
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, and Pathogens EA7300, and CHRU of Nancy, University of Lorraine, 54500, Vandoeuvre-les-Nancy, France
| |
Collapse
|
40
|
Scott EY, Woolard KD, Finno CJ, Murray JD. Cerebellar Abiotrophy Across Domestic Species. THE CEREBELLUM 2019; 17:372-379. [PMID: 29294214 DOI: 10.1007/s12311-017-0914-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cerebellar abiotrophy (CA) is a neurodegenerative disorder affecting the cerebellum and occurs in multiple species. Although CA is well researched in humans and mice, domestic species such as the dog, cat, sheep, cow, and horse receive little recognition. This may be due to few studies addressing the mechanism of CA in these species. However, valuable information can still be extracted from these cases. A review of the clinicohistologic phenotype of CA in these species and determining the various etiologies of CA may aid in determining conserved and required pathways necessary for proper cerebellar development and function. This review outlines research approaches of studies of CA in domestic species, compared to the approaches used in mice, with the objective of comparing CA in domestic species while identifying areas for further research efforts.
Collapse
Affiliation(s)
- Erica Yuki Scott
- Department of Animal Science, University of California, Davis, Meyer Hall, 1 Shields Ave, Davis, CA, 95616, USA
| | - Kevin Douglas Woolard
- Department of Pathology, Microbiology & Immunology, University of California, Davis, Davis, CA, USA
| | - Carrie J Finno
- Department of Population Health and Reproduction, University of California, Davis, Davis, CA, USA
| | - James D Murray
- Department of Animal Science, University of California, Davis, Meyer Hall, 1 Shields Ave, Davis, CA, 95616, USA.
- Department of Population Health and Reproduction, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
41
|
Baltanás FC, Berciano MT, Tapia O, Narcis JO, Lafarga V, Díaz D, Weruaga E, Santos E, Lafarga M. Nucleolin reorganization and nucleolar stress in Purkinje cells of mutant PCD mice. Neurobiol Dis 2019; 127:312-322. [PMID: 30905767 DOI: 10.1016/j.nbd.2019.03.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/20/2019] [Indexed: 01/12/2023] Open
Abstract
The Purkinje cell (PC) degeneration (pcd) mouse harbors a mutation in Agtpbp1 gene that encodes for the cytosolic carboxypeptidase, CCP1. The mutation causes degeneration and death of PCs during the postnatal life, resulting in clinical and pathological manifestation of cerebellar ataxia. Monogenic biallelic damaging variants in the Agtpbp1 gene cause infantile-onset neurodegeneration and cerebellar atrophy, linking loss of functional CCP1 with human neurodegeneration. Although CCP1 plays a key role in the regulation of tubulin stabilization, its loss of function in PCs leads to a severe nuclear phenotype with heterochromatinization and accumulation of DNA damage. Therefore, the pcd mice provides a useful neuronal model to investigate nuclear mechanisms involved in neurodegeneration, particularly the nucleolar stress. In this study, we demonstrated that the Agtpbp1 gene mutation induces a p53-dependent nucleolar stress response in PCs, which is characterized by nucleolar fragmentation, nucleoplasmic and cytoplasmic mislocalization of nucleolin, and dysfunction of both pre-rRNA processing and mRNA translation. RT-qPCR analysis revealed reduction of mature 18S rRNA, with a parallel increase of its intermediate 18S-5'-ETS precursor, that correlates with a reduced expression of Fbl mRNA, which encodes an essential factor for rRNA processing. Moreover, nucleolar alterations were accompanied by a reduction of PTEN mRNA and protein levels, which appears to be related to the chromosome instability and accumulation of DNA damage in degenerating PCs. Our results highlight the essential contribution of nucleolar stress to PC degeneration and also underscore the nucleoplasmic mislocalization of nucleolin as a potential indicator of neurodegenerative processes.
Collapse
Affiliation(s)
- Fernando C Baltanás
- Lab.1, CIC-IBMCC (Universidad de Salamanca-CSIC) and CIBERONC, Salamanca, Spain
| | - María T Berciano
- Department of Anat and Cell Biology and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", University of Cantabria-IDIVAL, Santander, Spain
| | - Olga Tapia
- Department of Anat and Cell Biology and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", University of Cantabria-IDIVAL, Santander, Spain
| | - Josep Oriol Narcis
- Department of Anat and Cell Biology and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", University of Cantabria-IDIVAL, Santander, Spain
| | - Vanesa Lafarga
- Laboratory of Genomic Instability, "Centro Nacional de Investigaciones Oncológicas" (CNIO), Madrid, Spain
| | - David Díaz
- Laboratory of Neural Plasticity and Neurorepair, Institute for Neuroscience of Castilla y León, Universidad de Salamanca, Salamanca, Spain
| | - Eduardo Weruaga
- Laboratory of Neural Plasticity and Neurorepair, Institute for Neuroscience of Castilla y León, Universidad de Salamanca, Salamanca, Spain
| | - Eugenio Santos
- Lab.1, CIC-IBMCC (Universidad de Salamanca-CSIC) and CIBERONC, Salamanca, Spain
| | - Miguel Lafarga
- Department of Anat and Cell Biology and "Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)", University of Cantabria-IDIVAL, Santander, Spain.
| |
Collapse
|
42
|
Karakaya M, Paketci C, Altmueller J, Thiele H, Hoelker I, Yis U, Wirth B. Biallelic variant in AGTPBP1 causes infantile lower motor neuron degeneration and cerebellar atrophy. Am J Med Genet A 2019; 179:1580-1584. [PMID: 31102495 DOI: 10.1002/ajmg.a.61198] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/03/2019] [Accepted: 05/05/2019] [Indexed: 11/08/2022]
Abstract
Infantile hereditary lower motor neuron disorders beyond 5q-spinal muscular atrophy (5q-SMA) are usually caused by mutations other than deletions or mutations in SMN1. In addition to motor neuron degeneration, further neurologic or multisystemic pathologies in non-5q-SMAs are not seldom. Some of the non-5q-SMA phenotypes, such as pontocerebellar hypoplasia (PCH1), have been classified later as a different disease group due to distinctive primary pathologies. Likewise, a novel phenotype, childhood-onset neurodegeneration with cerebellar atrophy (CONDCA) has been described recently in individuals with lower motor neuron disorder and cerebellar atrophy due to biallelic loss-of-function variants in AGTPBP1 that encodes cytosolic carboxypeptidase 1 (CCP1). Here we present two individuals with CONDCA in whom a biallelic missense AGTPBP1 variant (NM_001330701.1:c.2396G>T, p.Arg799Leu) was identified by whole exome sequencing. Affected individuals in this report correspond to the severe infantile spectrum of the disease and underline the severe pathogenic effect of this missense variant. This report is the second in the literature that delineates the pathogenic effects of biallelic AGTPBP1 variants presenting the recently described CONDCA disease.
Collapse
Affiliation(s)
- Mert Karakaya
- Institute of Human Genetics, Center for Molecular Medicine Cologne (CMMC), Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Cem Paketci
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Janine Altmueller
- Institute of Human Genetics, Center for Molecular Medicine Cologne (CMMC), Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany.,Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Holger Thiele
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Irmgard Hoelker
- Institute of Human Genetics, Center for Molecular Medicine Cologne (CMMC), Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Uluc Yis
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne (CMMC), Institute of Genetics, and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
43
|
Gagnier L, Belancio VP, Mager DL. Mouse germ line mutations due to retrotransposon insertions. Mob DNA 2019; 10:15. [PMID: 31011371 PMCID: PMC6466679 DOI: 10.1186/s13100-019-0157-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/01/2019] [Indexed: 12/24/2022] Open
Abstract
Transposable element (TE) insertions are responsible for a significant fraction of spontaneous germ line mutations reported in inbred mouse strains. This major contribution of TEs to the mutational landscape in mouse contrasts with the situation in human, where their relative contribution as germ line insertional mutagens is much lower. In this focussed review, we provide comprehensive lists of TE-induced mouse mutations, discuss the different TE types involved in these insertional mutations and elaborate on particularly interesting cases. We also discuss differences and similarities between the mutational role of TEs in mice and humans.
Collapse
Affiliation(s)
- Liane Gagnier
- Terry Fox Laboratory, BC Cancer and Department of Medical Genetics, University of British Columbia, V5Z1L3, Vancouver, BC Canada
| | - Victoria P. Belancio
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane Cancer Center, Tulane Center for Aging, New Orleans, LA 70112 USA
| | - Dixie L. Mager
- Terry Fox Laboratory, BC Cancer and Department of Medical Genetics, University of British Columbia, V5Z1L3, Vancouver, BC Canada
| |
Collapse
|
44
|
Biallelic variants in AGTPBP1, involved in tubulin deglutamylation, are associated with cerebellar degeneration and motor neuropathy. Eur J Hum Genet 2019; 27:1419-1426. [PMID: 30976113 DOI: 10.1038/s41431-019-0400-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/16/2019] [Accepted: 03/26/2019] [Indexed: 11/08/2022] Open
Abstract
The ATP/GTP-Binding Protein 1 (AGTPBP1) gene (OMIM *606830) catalyzes deglutamylation of polyglutamylated proteins, and its deficiency manifests by cerebellar ataxia and peripheral neuropathy in mice and lower motor neuron-like disease in sheep. In the mutant mice, cerebellar atrophy due to Purkinje cell degeneration is observed, likely due to increased tubulin polyglutamylation in affected brain areas. We report two unrelated individuals who presented with early onset cerebellar atrophy, developmental arrest with progressive muscle weakness, and feeding and respiratory difficulties, accompanied by severe motor neuronopathy. Whole exome sequencing followed by segregation analysis in the families and cDNA studies revealed deleterious biallelic variants in the AGTPBP1 gene. We conclude that complete loss-of-function of AGTPBP1 in humans, just like in mice and sheep, is associated with cerebellar and motor neuron disease, reminiscent of Pontocerebellar Hypoplasia Type 1 (PCH1).
Collapse
|
45
|
Inberg S, Meledin A, Kravtsov V, Iosilevskii Y, Oren-Suissa M, Podbilewicz B. Lessons from Worm Dendritic Patterning. Annu Rev Neurosci 2019; 42:365-383. [PMID: 30939099 DOI: 10.1146/annurev-neuro-072116-031437] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The structural and functional properties of neurons have intrigued scientists since the pioneering work of Santiago Ramón y Cajal. Since then, emerging cutting-edge technologies, including light and electron microscopy, electrophysiology, biochemistry, optogenetics, and molecular biology, have dramatically increased our understanding of dendritic properties. This advancement was also facilitated by the establishment of different animal model organisms, from flies to mammals. Here we describe the emerging model system of a Caenorhabditis elegans polymodal neuron named PVD, whose dendritic tree follows a stereotypical structure characterized by repeating candelabra-like structural units. In the past decade, progress has been made in understanding PVD's functions, morphogenesis, regeneration, and aging, yet many questions still remain.
Collapse
Affiliation(s)
- Sharon Inberg
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel;
| | - Anna Meledin
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel;
| | - Veronika Kravtsov
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel;
| | - Yael Iosilevskii
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel;
| | - Meital Oren-Suissa
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Benjamin Podbilewicz
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel;
| |
Collapse
|
46
|
Gilmore-Hall S, Kuo J, Ward JM, Zahra R, Morrison RS, Perkins G, La Spada AR. CCP1 promotes mitochondrial fusion and motility to prevent Purkinje cell neuron loss in pcd mice. J Cell Biol 2019; 218:206-219. [PMID: 30337352 PMCID: PMC6314562 DOI: 10.1083/jcb.201709028] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 08/03/2018] [Accepted: 10/01/2018] [Indexed: 12/22/2022] Open
Abstract
A perplexing question in neurodegeneration is why different neurons degenerate. The Purkinje cell degeneration (pcd) mouse displays a dramatic phenotype of degeneration of cerebellar Purkinje cells. Loss of CCP1/Nna1 deglutamylation of tubulin accounts for pcd neurodegeneration, but the mechanism is unknown. In this study, we modulated the dosage of fission and fusion genes in a Drosophila melanogaster loss-of-function model and found that mitochondrial fragmentation and disease phenotypes were rescued by reduced Drp1. We observed mitochondrial fragmentation in CCP1 null cells and in neurons from pcd mice, and we documented reduced mitochondrial fusion in cells lacking CCP1. We examined the effect of tubulin hyperglutamylation on microtubule-mediated mitochondrial motility in pcd neurons and noted markedly reduced retrograde axonal transport. Mitochondrial stress promoted Parkin-dependent turnover of CCP1, and CCP1 and Parkin physically interacted. Our results indicate that CCP1 regulates mitochondrial motility through deglutamylation of tubulin and that loss of CCP1-mediated mitochondrial fusion accounts for the exquisite vulnerability of Purkinje neurons in pcd mice.
Collapse
Affiliation(s)
| | - Jennifer Kuo
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Jacqueline M Ward
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Rabaab Zahra
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | | | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA
| | - Albert R La Spada
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA
- Department of Neurology, Duke University School of Medicine, Durham, NC
- Department of Neurobiology, Duke University School of Medicine, Durham, NC
- Department of Cell Biology, Duke University School of Medicine, Durham, NC
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University School of Medicine, Durham, NC
| |
Collapse
|
47
|
Magiera MM, Bodakuntla S, Žiak J, Lacomme S, Marques Sousa P, Leboucher S, Hausrat TJ, Bosc C, Andrieux A, Kneussel M, Landry M, Calas A, Balastik M, Janke C. Excessive tubulin polyglutamylation causes neurodegeneration and perturbs neuronal transport. EMBO J 2018; 37:e100440. [PMID: 30420556 PMCID: PMC6276888 DOI: 10.15252/embj.2018100440] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
Posttranslational modifications of tubulin are emerging regulators of microtubule functions. We have shown earlier that upregulated polyglutamylation is linked to rapid degeneration of Purkinje cells in mice with a mutation in the deglutamylating enzyme CCP1. How polyglutamylation leads to degeneration, whether it affects multiple neuron types, or which physiological processes it regulates in healthy neurons has remained unknown. Here, we demonstrate that excessive polyglutamylation induces neurodegeneration in a cell-autonomous manner and can occur in many parts of the central nervous system. Degeneration of selected neurons in CCP1-deficient mice can be fully rescued by simultaneous knockout of the counteracting polyglutamylase TTLL1. Excessive polyglutamylation reduces the efficiency of neuronal transport in cultured hippocampal neurons, suggesting that impaired cargo transport plays an important role in the observed degenerative phenotypes. We thus establish polyglutamylation as a cell-autonomous mechanism for neurodegeneration that might be therapeutically accessible through manipulation of the enzymes that control this posttranslational modification.
Collapse
Affiliation(s)
- Maria M Magiera
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Satish Bodakuntla
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Jakub Žiak
- Department of Molecular Neurobiology, Institute of Physiology, Czech Academy of Sciences, Prague 4, Czech Republic
- Faculty of Science, Charles University, Prague 2, Czech Republic
| | - Sabrina Lacomme
- Bordeaux Imaging Center, BIC, UMS 3420, Université Bordeaux, Bordeaux, France
| | - Patricia Marques Sousa
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Sophie Leboucher
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| | - Torben J Hausrat
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christophe Bosc
- Grenoble Institut des Neurosciences, GIN, Université Grenoble Alpes, Grenoble, France
- Inserm U1216, Grenoble, France
| | - Annie Andrieux
- Grenoble Institut des Neurosciences, GIN, Université Grenoble Alpes, Grenoble, France
- Inserm U1216, Grenoble, France
| | - Matthias Kneussel
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Landry
- Interdisciplinary Institute for Neuroscience, CNRS UMR5297, Université Bordeaux, Bordeaux, France
| | - André Calas
- Interdisciplinary Institute for Neuroscience, CNRS UMR5297, Université Bordeaux, Bordeaux, France
| | - Martin Balastik
- Department of Molecular Neurobiology, Institute of Physiology, Czech Academy of Sciences, Prague 4, Czech Republic
| | - Carsten Janke
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Université Paris Sud, Orsay, France
| |
Collapse
|
48
|
Shashi V, Magiera MM, Klein D, Zaki M, Schoch K, Rudnik-Schöneborn S, Norman A, Lopes Abath Neto O, Dusl M, Yuan X, Bartesaghi L, De Marco P, Alfares AA, Marom R, Arold ST, Guzmán-Vega FJ, Pena LD, Smith EC, Steinlin M, Babiker MO, Mohassel P, Foley AR, Donkervoort S, Kaur R, Ghosh PS, Stanley V, Musaev D, Nava C, Mignot C, Keren B, Scala M, Tassano E, Picco P, Doneda P, Fiorillo C, Issa MY, Alassiri A, Alahmad A, Gerard A, Liu P, Yang Y, Ertl-Wagner B, Kranz PG, Wentzensen IM, Stucka R, Stong N, Allen AS, Goldstein DB, Schoser B, Rösler KM, Alfadhel M, Capra V, Chrast R, Strom TM, Kamsteeg EJ, Bönnemann CG, Gleeson JG, Martini R, Janke C, Senderek J. Loss of tubulin deglutamylase CCP1 causes infantile-onset neurodegeneration. EMBO J 2018; 37:e100540. [PMID: 30420557 PMCID: PMC6276871 DOI: 10.15252/embj.2018100540] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 11/09/2022] Open
Abstract
A set of glutamylases and deglutamylases controls levels of tubulin polyglutamylation, a prominent post-translational modification of neuronal microtubules. Defective tubulin polyglutamylation was first linked to neurodegeneration in the Purkinje cell degeneration (pcd) mouse, which lacks deglutamylase CCP1, displays massive cerebellar atrophy, and accumulates abnormally glutamylated tubulin in degenerating neurons. We found biallelic rare and damaging variants in the gene encoding CCP1 in 13 individuals with infantile-onset neurodegeneration and confirmed the absence of functional CCP1 along with dysregulated tubulin polyglutamylation. The human disease mainly affected the cerebellum, spinal motor neurons, and peripheral nerves. We also demonstrate previously unrecognized peripheral nerve and spinal motor neuron degeneration in pcd mice, which thus recapitulated key features of the human disease. Our findings link human neurodegeneration to tubulin polyglutamylation, entailing this post-translational modification as a potential target for drug development for neurodegenerative disorders.
Collapse
Affiliation(s)
- Vandana Shashi
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Maria M Magiera
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- CNRS UMR3348, Université Paris Sud, Université Paris-Saclay, Orsay, France
| | - Dennis Klein
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Maha Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Kelly Schoch
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | | | - Andrew Norman
- Department of Clinical Genetics, St. Michael's Hospital, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Osorio Lopes Abath Neto
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Marina Dusl
- Friedrich Baur Institute at the Department of Neurology, Friedrich Baur Institute, University Hospital, LMU Munich, Munich, Germany
| | - Xidi Yuan
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Luca Bartesaghi
- Department of Neuroscience and Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Ahmed A Alfares
- Department of Pediatrics, College of Medicine, Qassim University, Qassim, Saudi Arabia
| | - Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, TX, USA
| | - Stefan T Arold
- Division of Biological and Environmental Sciences and Engineering (BESE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Francisco J Guzmán-Vega
- Division of Biological and Environmental Sciences and Engineering (BESE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Loren Dm Pena
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Edward C Smith
- Division of Neurology, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Maja Steinlin
- Division of Neuropaediatrics, Development and Rehabilitation, University Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | | | - Payam Mohassel
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Rupleen Kaur
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Partha S Ghosh
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Valentina Stanley
- Laboratory for Pediatric Brain Disease, Howard Hughes Medical Institute, University of California, San Diego, CA, USA
| | - Damir Musaev
- Laboratory for Pediatric Brain Disease, Howard Hughes Medical Institute, University of California, San Diego, CA, USA
| | - Caroline Nava
- Department of Genetics, Assistance Publique des Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, Inserm U1127, CNRS, UMR 7225, UPMC Univ Paris 06, Paris, France
| | - Cyril Mignot
- Department of Genetics, Assistance Publique des Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, Inserm U1127, CNRS, UMR 7225, UPMC Univ Paris 06, Paris, France
| | - Boris Keren
- Department of Genetics, Assistance Publique des Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, Inserm U1127, CNRS, UMR 7225, UPMC Univ Paris 06, Paris, France
| | | | | | - Paolo Picco
- IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Paola Doneda
- Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Chiara Fiorillo
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Università degli Studi di Genova, Genova, Italy
| | - Mahmoud Y Issa
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Ali Alassiri
- Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Ahmed Alahmad
- Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Amanda Gerard
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, TX, USA
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Baylor Genetics, Houston, TX, USA
| | - Yaping Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Baylor Genetics, Houston, TX, USA
| | - Birgit Ertl-Wagner
- Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Peter G Kranz
- Division of Neuroradiology, Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | | | - Rolf Stucka
- Friedrich Baur Institute at the Department of Neurology, Friedrich Baur Institute, University Hospital, LMU Munich, Munich, Germany
| | - Nicholas Stong
- Institute of Genomic Medicine, Columbia University, New York, NY, USA
| | - Andrew S Allen
- Center for Statistical Genetics and Genomics, Duke University Medical Center, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - David B Goldstein
- Institute of Genomic Medicine, Columbia University, New York, NY, USA
| | - Benedikt Schoser
- Friedrich Baur Institute at the Department of Neurology, Friedrich Baur Institute, University Hospital, LMU Munich, Munich, Germany
| | - Kai M Rösler
- Neuromuscular Centre, University Department of Neurology, Inselspital, Bern, Switzerland
| | - Majid Alfadhel
- Genetics Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | | | - Roman Chrast
- Department of Neuroscience and Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tim M Strom
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Donders Centre for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, Howard Hughes Medical Institute, University of California, San Diego, CA, USA
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Carsten Janke
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France
- CNRS UMR3348, Université Paris Sud, Université Paris-Saclay, Orsay, France
| | - Jan Senderek
- Friedrich Baur Institute at the Department of Neurology, Friedrich Baur Institute, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
49
|
Zhou L, Hossain MI, Yamazaki M, Abe M, Natsume R, Konno K, Kageyama S, Komatsu M, Watanabe M, Sakimura K, Takebayashi H. Deletion of exons encoding carboxypeptidase domain of Nna1 results in Purkinje cell degeneration (pcd
) phenotype. J Neurochem 2018; 147:557-572. [DOI: 10.1111/jnc.14591] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 08/31/2018] [Accepted: 09/03/2018] [Indexed: 02/02/2023]
Affiliation(s)
- Li Zhou
- Department of Cellular Neurobiology; Brain Research Institute; Niigata University; Niigata Japan
- Division of Neurobiology and Anatomy; Graduate School of Medical and Dental Sciences; Niigata University; Niigata Japan
| | - M. Ibrahim Hossain
- Division of Neurobiology and Anatomy; Graduate School of Medical and Dental Sciences; Niigata University; Niigata Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology; Brain Research Institute; Niigata University; Niigata Japan
| | - Manabu Abe
- Department of Cellular Neurobiology; Brain Research Institute; Niigata University; Niigata Japan
| | - Rie Natsume
- Department of Cellular Neurobiology; Brain Research Institute; Niigata University; Niigata Japan
| | - Kohtaro Konno
- Department of Anatomy; Faculty of Medicine; Hokkaido University; Sapporo Japan
| | - Shun Kageyama
- Department of Biochemistry; Graduate School of Medical and Dental Sciences; Niigata University; Niigata Japan
| | - Masaaki Komatsu
- Department of Biochemistry; Graduate School of Medical and Dental Sciences; Niigata University; Niigata Japan
| | - Masahiko Watanabe
- Department of Anatomy; Faculty of Medicine; Hokkaido University; Sapporo Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology; Brain Research Institute; Niigata University; Niigata Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy; Graduate School of Medical and Dental Sciences; Niigata University; Niigata Japan
| |
Collapse
|
50
|
Ye B, Liu B, Hao L, Zhu X, Yang L, Wang S, Xia P, Du Y, Meng S, Huang G, Qin X, Wang Y, Yan X, Li C, Hao J, Zhu P, He L, Tian Y, Fan Z. Klf4 glutamylation is required for cell reprogramming and early embryonic development in mice. Nat Commun 2018; 9:1261. [PMID: 29593216 PMCID: PMC5871780 DOI: 10.1038/s41467-018-03008-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/10/2018] [Indexed: 12/20/2022] Open
Abstract
Temporal and spatial-specific regulation of pluripotency networks is largely dependent on the precise modifications of core transcription factors. Misregulation of glutamylation is implicated in severe physiological abnormalities. However, how glutamylation regulates cell reprogramming and pluripotency networks remains elusive. Here we show that cytosolic carboxypeptidases 1 (CCP1) or CCP6 deficiency substantially promotes induced pluripotent cell (iPSC) induction and pluripotency of embryonic stem cells (ESCs). Klf4 polyglutamylation at Glu381 by tubulin tyrosine ligase-like 4 (TTLL4) and TTLL1 during cell reprogramming impedes its lysine 48-linked ubiquitination and sustains Klf4 stability. Klf4-E381A knockin mice display impaired blastocyst development and embryonic lethality. Deletion of TTLL4 or TTLL1 abrogates cell reprogramming and early embryogenesis. Thus, Klf4 polyglutamylation plays a critical role in the regulation of cell reprogramming and pluripotency maintenance. Embryonic stem cell pluripotency depends upon precise regulation by a core transcription network. Here the authors show that polyglutamylation mediated stabilization of the transcription factor Klf4 by TTLL1 and TTLL4 promotes reprogramming, pluripotency and preimplantation embryonic development.
Collapse
Affiliation(s)
- Buqing Ye
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Benyu Liu
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lu Hao
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoxiao Zhu
- Laboratory Animal Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liuliu Yang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuo Wang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pengyan Xia
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying Du
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shu Meng
- Laboratory Animal Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guanling Huang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiwen Qin
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanying Wang
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinlong Yan
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chong Li
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Junfeng Hao
- Laboratory Animal Center, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pingping Zhu
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Luyun He
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Tian
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zusen Fan
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|