51
|
Dong J, Jin L, Bao S, Chen B, Zeng Y, Luo Y, Du X, Sang Q, Wu T, Wang L. Ectopic expression of human TUBB8 leads to increased aneuploidy in mouse oocytes. Cell Discov 2023; 9:105. [PMID: 37875488 PMCID: PMC10598138 DOI: 10.1038/s41421-023-00599-z] [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: 02/07/2023] [Accepted: 08/26/2023] [Indexed: 10/26/2023] Open
Abstract
Aneuploidy seriously compromises female fertility and increases incidence of birth defects. Rates of aneuploidy in human eggs from even young women are significantly higher than those in other mammals. However, intrinsic genetic factors underlying this high incidence of aneuploidy in human eggs remain largely unknown. Here, we found that ectopic expression of human TUBB8 in mouse oocytes increases rates of aneuploidy by causing kinetochore-microtubule (K-MT) attachment defects. Stretched bivalents in mouse oocytes expressing TUBB8 are under less tension, resulting in continuous phosphorylation status of HEC1 by AURKB/C at late metaphase I that impairs the established correct K-MT attachments. This reduced tension in stretched bivalents likely correlates with decreased recruitment of KIF11 on meiotic spindles. We also found that ectopic expression of TUBB8 without its C-terminal tail decreases aneuploidy rates by reducing erroneous K-MT attachments. Importantly, variants in the C-terminal tail of TUBB8 were identified in patients with recurrent miscarriages. Ectopic expression of an identified TUBB8 variant in mouse oocytes also compromises K-MT attachments and increases aneuploidy rates. In conclusion, our study provides novel understanding for physiological mechanisms of aneuploidy in human eggs as well as for pathophysiological mechanisms involved in recurrent miscarriages.
Collapse
Affiliation(s)
- Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shihua Bao
- Department of Reproductive Immunology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai, China
| | - Yang Zeng
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Yuxi Luo
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Xingzhu Du
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| | - Tianyu Wu
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| |
Collapse
|
52
|
Yamane T, Nakayama T, Ekimoto T, Inoue M, Ikezaki K, Sekiguchi H, Kuramochi M, Terao Y, Judai K, Saito M, Ikeguchi M, Sasaki YC. Comparison of the Molecular Motility of Tubulin Dimeric Isoforms: Molecular Dynamics Simulations and Diffracted X-ray Tracking Study. Int J Mol Sci 2023; 24:15423. [PMID: 37895101 PMCID: PMC10607685 DOI: 10.3390/ijms242015423] [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: 07/31/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Tubulin has been recently reported to form a large family consisting of various gene isoforms; however, the differences in the molecular features of tubulin dimers composed of a combination of these isoforms remain unknown. Therefore, we attempted to elucidate the physical differences in the molecular motility of these tubulin dimers using the method of measurable pico-meter-scale molecular motility, diffracted X-ray tracking (DXT) analysis, regarding characteristic tubulin dimers, including neuronal TUBB3 and ubiquitous TUBB5. We first conducted a DXT analysis of neuronal (TUBB3-TUBA1A) and ubiquitous (TUBB5-TUBA1B) tubulin dimers and found that the molecular motility around the vertical axis of the neuronal tubulin dimer was lower than that of the ubiquitous tubulin dimer. The results of molecular dynamics (MD) simulation suggest that the difference in motility between the neuronal and ubiquitous tubulin dimers was probably caused by a change in the major contact of Gln245 in the T7 loop of TUBB from Glu11 in TUBA to Val353 in TUBB. The present study is the first report of a novel phenomenon in which the pico-meter-scale molecular motility between neuronal and ubiquitous tubulin dimers is different.
Collapse
Affiliation(s)
- Tsutomu Yamane
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; (T.E.); (M.I.); (M.I.)
- HPC- and AI-Driven Drug Development Platform Division, Riken Center for Computational Science, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Takahiro Nakayama
- Department of Medical Physiology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Japan; (T.N.); (Y.T.)
| | - Toru Ekimoto
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; (T.E.); (M.I.); (M.I.)
| | - Masao Inoue
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; (T.E.); (M.I.); (M.I.)
| | - Keigo Ikezaki
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8568, Japan; (K.I.); (M.K.)
| | - Hiroshi Sekiguchi
- Japan Synchrotron Radiation Research Institute, SPring-8, 1-1-1 Kouto, Sayo 679-5198, Japan;
| | - Masahiro Kuramochi
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8568, Japan; (K.I.); (M.K.)
| | - Yasuo Terao
- Department of Medical Physiology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Japan; (T.N.); (Y.T.)
| | - Ken Judai
- Department of Physics, College of Humanities and Sciences, Nihon University, Sakurajosui 3-25-40, Tokyo 156-8550, Japan;
| | - Minoru Saito
- Department of Biosciences, College of Humanities and Sciences, Nihon University, Tokyo 156-8550, Japan;
| | - Mitsunori Ikeguchi
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; (T.E.); (M.I.); (M.I.)
- HPC- and AI-Driven Drug Development Platform Division, Riken Center for Computational Science, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Yuji C. Sasaki
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8568, Japan; (K.I.); (M.K.)
- Japan Synchrotron Radiation Research Institute, SPring-8, 1-1-1 Kouto, Sayo 679-5198, Japan;
- AIST-UTokyo Advanced Operando-Measurement Technology Open Innovation Laboratory (OPERANDO-OIL), National Institute of Advanced Industrial Science and Technology (AIST), 6-2-3 Kashiwanoha, Chiba 277-0882, Japan
| |
Collapse
|
53
|
McKenna ED, Sarbanes SL, Cummings SW, Roll-Mecak A. The Tubulin Code, from Molecules to Health and Disease. Annu Rev Cell Dev Biol 2023; 39:331-361. [PMID: 37843925 DOI: 10.1146/annurev-cellbio-030123-032748] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Microtubules are essential dynamic polymers composed of α/β-tubulin heterodimers. They support intracellular trafficking, cell division, cellular motility, and other essential cellular processes. In many species, both α-tubulin and β-tubulin are encoded by multiple genes with distinct expression profiles and functionality. Microtubules are further diversified through abundant posttranslational modifications, which are added and removed by a suite of enzymes to form complex, stereotyped cellular arrays. The genetic and chemical diversity of tubulin constitute a tubulin code that regulates intrinsic microtubule properties and is read by cellular effectors, such as molecular motors and microtubule-associated proteins, to provide spatial and temporal specificity to microtubules in cells. In this review, we synthesize the rapidly expanding tubulin code literature and highlight limitations and opportunities for the field. As complex microtubule arrays underlie essential physiological processes, a better understanding of how cells employ the tubulin code has important implications for human disease ranging from cancer to neurological disorders.
Collapse
Affiliation(s)
- Elizabeth D McKenna
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Stephanie L Sarbanes
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Steven W Cummings
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
54
|
Yue Y, Hotta T, Higaki T, Verhey KJ, Ohi R. Microtubule detyrosination by VASH1/SVBP is regulated by the conformational state of tubulin in the lattice. Curr Biol 2023; 33:4111-4123.e7. [PMID: 37716348 PMCID: PMC10592207 DOI: 10.1016/j.cub.2023.07.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 09/18/2023]
Abstract
Tubulin, a heterodimer of α- and β-tubulin, is a GTPase that assembles into microtubule (MT) polymers whose dynamic properties are intimately coupled to nucleotide hydrolysis. In cells, the organization and dynamics of MTs are further tuned by post-translational modifications (PTMs), which control the ability of MT-associated proteins (MAPs) and molecular motors to engage MTs. Detyrosination is a PTM of α-tubulin, wherein its C-terminal tyrosine residue is enzymatically removed by either the vasohibin (VASH) or MT-associated tyrosine carboxypeptidase (MATCAP) peptidases. How these enzymes generate specific patterns of MT detyrosination in cells is not known. Here, we use a novel antibody-based probe to visualize the formation of detyrosinated MTs in real time and employ single-molecule imaging of VASH1 bound to its regulatory partner small-vasohibin binding protein (SVBP) to understand the process of MT detyrosination in vitro and in cells. We demonstrate that the activity, but not binding, of VASH1/SVBP is much greater on mimics of guanosine triphosphate (GTP)-MTs than on guanosine diphosphate (GDP)-MTs. Given emerging data showing that tubulin subunits in GTP-MTs are in expanded conformation relative to tubulin subunits in GDP-MTs, we reasoned that the lattice conformation of MTs is a key factor that gates the activity of VASH1/SVBP. We show that Taxol, a drug known to expand the MT lattice, promotes MT detyrosination and that CAMSAP2 and CAMSAP3 are two MAPs that spatially regulate detyrosination in cells. Collectively, our work shows that VASH1/SVBP detyrosination is regulated by the conformational state of tubulin in the MT lattice and that this is spatially determined in cells by the activity of MAPs.
Collapse
Affiliation(s)
- Yang Yue
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Takashi Hotta
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Takumi Higaki
- Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; International Research Organization in Advanced Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| | - Kristen J Verhey
- Department of Cell and Developmental Biology, 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
|
55
|
Du C, Cai N, Dong J, Xu C, Wang Q, Zhang Z, Li J, Huang C, Ma T. Uncovering the role of cytoskeleton proteins in the formation of neutrophil extracellular traps. Int Immunopharmacol 2023; 123:110607. [PMID: 37506501 DOI: 10.1016/j.intimp.2023.110607] [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: 05/03/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Neutrophils are a type of lymphocyte involved in innate immune defense. In response to specific stimuli, these phagocytic cells undergo a unique form of cell death, NETosis, during which they release neutrophil extracellular traps (NETs) composed of modified chromatin structures decorated with cytoplasmic and granular proteins. Multiple proteins and pathways have been implicated in the formation of NETs. The cytoskeleton, an interconnected network of filamentous polymers and regulatory proteins, plays a crucial role in resisting deformation, transporting intracellular cargo, and changing shape during movement of eukaryotic cells. It may also have evolved to defend eukaryotic organisms against infection. Recent research focuses on understanding the mechanisms underlying NETs formation and how cytoskeletal networks contribute to this process, by identifying enzymes that trigger NETosis or interact with NETs and influence cellular behavior through cytoskeletal dynamics. An enhanced understanding of the complex relationship between the cytoskeleton and NET formation will provide a framework for future research and the development of targeted therapeutic strategies, and supports the notion that the long-lived cytoskeleton structures may have a lasting impact on this area of research.
Collapse
Affiliation(s)
- Changlin Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Na Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jiahui Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Chuanting Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qi Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhenming Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Taotao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
56
|
Pernar Kovač M, Tadić V, Kralj J, Duran GE, Stefanelli A, Stupin Polančec D, Dabelić S, Bačić N, Tomicic MT, Heffeter P, Sikic BI, Brozovic A. Carboplatin-induced upregulation of pan β-tubulin and class III β-tubulin is implicated in acquired resistance and cross-resistance of ovarian cancer. Cell Mol Life Sci 2023; 80:294. [PMID: 37718345 PMCID: PMC11071939 DOI: 10.1007/s00018-023-04943-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
Resistance to platinum- and taxane-based chemotherapy represents a major obstacle to long-term survival in ovarian cancer (OC) patients. Here, we studied the interplay between acquired carboplatin (CBP) resistance using two OC cell models, MES-OV CBP and SK-OV-3 CBP, and non-P-glycoprotein-mediated cross-resistance to paclitaxel (TAX) observed only in MES-OV CBP cells. Decreased platination, mesenchymal-like phenotype, and increased expression of α- and γ-tubulin were observed in both drug-resistant variants compared with parental cells. Both variants revealed increased protein expression of class III β-tubulin (TUBB3) but differences in TUBB3 branching and nuclear morphology. Transient silencing of TUBB3 sensitized MES-OV CBP cells to TAX, and surprisingly also to CBP. This phenomenon was not observed in the SK-OV-3 CBP variant, probably due to the compensation by other β-tubulin isotypes. Reduced TUBB3 levels in MES-OV CBP cells affected DNA repair protein trafficking and increased whole-cell platination level. Furthermore, TUBB3 depletion augmented therapeutic efficiency in additional OC cells, showing vice versa drug-resistant pattern, lacking β-tubulin isotype compensation visible at the level of total β-tubulin (TUBB) in vitro and ex vivo. In summary, the level of TUBB in OC should be considered together with TUBB3 in therapy response prediction.
Collapse
Affiliation(s)
- Margareta Pernar Kovač
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, 10000, Zagreb, Croatia
| | - Vanja Tadić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, 10000, Zagreb, Croatia
| | - Juran Kralj
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, 10000, Zagreb, Croatia
| | - George E Duran
- Division of Oncology, Stanford University School of Medicine, 269 Campus Dr., 94305, Stanford, CA, USA
| | - Alessia Stefanelli
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | | | - Sanja Dabelić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000, Zagreb, Croatia
| | - Niko Bačić
- Division for Marine and Environmental Research, Ruđer Bošković Institute, Bijenička Str. 54, 10000, Zagreb, Croatia
| | - Maja T Tomicic
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| | - Petra Heffeter
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Branimir I Sikic
- Division of Oncology, Stanford University School of Medicine, 269 Campus Dr., 94305, Stanford, CA, USA
| | - Anamaria Brozovic
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, 10000, Zagreb, Croatia.
| |
Collapse
|
57
|
Cabrera-Rodríguez R, Pérez-Yanes S, Lorenzo-Sánchez I, Trujillo-González R, Estévez-Herrera J, García-Luis J, Valenzuela-Fernández A. HIV Infection: Shaping the Complex, Dynamic, and Interconnected Network of the Cytoskeleton. Int J Mol Sci 2023; 24:13104. [PMID: 37685911 PMCID: PMC10487602 DOI: 10.3390/ijms241713104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
HIV-1 has evolved a plethora of strategies to overcome the cytoskeletal barrier (i.e., actin and intermediate filaments (AFs and IFs) and microtubules (MTs)) to achieve the viral cycle. HIV-1 modifies cytoskeletal organization and dynamics by acting on associated adaptors and molecular motors to productively fuse, enter, and infect cells and then traffic to the cell surface, where virions assemble and are released to spread infection. The HIV-1 envelope (Env) initiates the cycle by binding to and signaling through its main cell surface receptors (CD4/CCR5/CXCR4) to shape the cytoskeleton for fusion pore formation, which permits viral core entry. Then, the HIV-1 capsid is transported to the nucleus associated with cytoskeleton tracks under the control of specific adaptors/molecular motors, as well as HIV-1 accessory proteins. Furthermore, HIV-1 drives the late stages of the viral cycle by regulating cytoskeleton dynamics to assure viral Pr55Gag expression and transport to the cell surface, where it assembles and buds to mature infectious virions. In this review, we therefore analyze how HIV-1 generates a cell-permissive state to infection by regulating the cytoskeleton and associated factors. Likewise, we discuss the relevance of this knowledge to understand HIV-1 infection and pathogenesis in patients and to develop therapeutic strategies to battle HIV-1.
Collapse
Affiliation(s)
- Romina Cabrera-Rodríguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Silvia Pérez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Iria Lorenzo-Sánchez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
- Analysis Department, Faculty of Mathematics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain
| | - Judith Estévez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Jonay García-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| | - Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (R.C.-R.); (S.P.-Y.); (I.L.-S.); (R.T.-G.); (J.E.-H.); (J.G.-L.)
| |
Collapse
|
58
|
Mann JR, McKenna ED, Mawrie D, Papakis V, Alessandrini F, Anderson EN, Mayers R, Ball HE, Kaspi E, Lubinski K, Baron DM, Tellez L, Landers JE, Pandey UB, Kiskinis E. Loss of function of the ALS-associated NEK1 kinase disrupts microtubule homeostasis and nuclear import. SCIENCE ADVANCES 2023; 9:eadi5548. [PMID: 37585529 PMCID: PMC10431718 DOI: 10.1126/sciadv.adi5548] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/17/2023] [Indexed: 08/18/2023]
Abstract
Loss-of-function variants in NIMA-related kinase 1 (NEK1) constitute a major genetic cause of amyotrophic lateral sclerosis (ALS), accounting for 2 to 3% of all cases. However, how NEK1 mutations cause motor neuron (MN) dysfunction is unknown. Using mass spectrometry analyses for NEK1 interactors and NEK1-dependent expression changes, we find functional enrichment for proteins involved in the microtubule cytoskeleton and nucleocytoplasmic transport. We show that α-tubulin and importin-β1, two key proteins involved in these processes, are phosphorylated by NEK1 in vitro. NEK1 is essential for motor control and survival in Drosophila models in vivo, while using several induced pluripotent stem cell (iPSC)-MN models, including NEK1 knockdown, kinase inhibition, and a patient mutation, we find evidence for disruptions in microtubule homeostasis and nuclear import. Notably, stabilizing microtubules with two distinct classes of drugs restored NEK1-dependent deficits in both pathways. The capacity of NEK1 to modulate these processes that are critically involved in ALS pathophysiology renders this kinase a formidable therapeutic candidate.
Collapse
Affiliation(s)
- Jacob R. Mann
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Elizabeth D. McKenna
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Darilang Mawrie
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Vasileios Papakis
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Francesco Alessandrini
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eric N. Anderson
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Ryan Mayers
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hannah E. Ball
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Evan Kaspi
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Katherine Lubinski
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Desiree M. Baron
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Liana Tellez
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John E. Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Udai B. Pandey
- Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
59
|
Fan X, McKenney RJ. Control of motor landing and processivity by the CAP-Gly domain in the KIF13B tail. Nat Commun 2023; 14:4715. [PMID: 37543636 PMCID: PMC10404244 DOI: 10.1038/s41467-023-40425-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 07/27/2023] [Indexed: 08/07/2023] Open
Abstract
Microtubules are major components of the eukaryotic cytoskeleton. Posttranslational modifications (PTMs) of tubulin regulates interactions with microtubule-associated proteins (MAPs). One unique PTM is the cyclical removal and re-addition of the C-terminal tyrosine of α-tubulin and MAPs containing CAP-Gly domains specifically recognize tyrosinated microtubules. KIF13B, a long-distance transport kinesin, contains a conserved CAP-Gly domain, but the role of the CAP-Gly domain in KIF13B's motility along microtubules remains unknown. To address this, we investigate the interaction between KIF13B's CAP-Gly domain, and tyrosinated microtubules. We find that KIF13B's CAP-Gly domain influences the initial motor-microtubule interaction, as well as processive motility along microtubules. The effect of the CAP-Gly domain is enhanced when the motor domain is in the ADP state, suggesting an interplay between the N-terminal motor domain and C-terminal CAP-Gly domain. These results reveal that specialized kinesin tail domains play active roles in the initiation and continuation of motor movement.
Collapse
Affiliation(s)
- Xiangyu Fan
- Department of Molecular and Cellular Biology, University of California - Davis, 145 Briggs Hall, Davis, CA, 95616, USA
| | - Richard J McKenney
- Department of Molecular and Cellular Biology, University of California - Davis, 145 Briggs Hall, Davis, CA, 95616, USA.
| |
Collapse
|
60
|
Puri D, Barry BJ, Engle EC. TUBB3 and KIF21A in neurodevelopment and disease. Front Neurosci 2023; 17:1226181. [PMID: 37600020 PMCID: PMC10436312 DOI: 10.3389/fnins.2023.1226181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Neuronal migration and axon growth and guidance require precise control of microtubule dynamics and microtubule-based cargo transport. TUBB3 encodes the neuronal-specific β-tubulin isotype III, TUBB3, a component of neuronal microtubules expressed throughout the life of central and peripheral neurons. Human pathogenic TUBB3 missense variants result in altered TUBB3 function and cause errors either in the growth and guidance of cranial and, to a lesser extent, central axons, or in cortical neuronal migration and organization, and rarely in both. Moreover, human pathogenic missense variants in KIF21A, which encodes an anterograde kinesin motor protein that interacts directly with microtubules, alter KIF21A function and cause errors in cranial axon growth and guidance that can phenocopy TUBB3 variants. Here, we review reported TUBB3 and KIF21A variants, resulting phenotypes, and corresponding functional studies of both wildtype and mutant proteins. We summarize the evidence that, in vitro and in mouse models, loss-of-function and missense variants can alter microtubule dynamics and microtubule-kinesin interactions. Lastly, we highlight additional studies that might contribute to our understanding of the relationship between specific tubulin isotypes and specific kinesin motor proteins in health and disease.
Collapse
Affiliation(s)
- Dharmendra Puri
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Brenda J. Barry
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Elizabeth C. Engle
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
61
|
Şenkardeş S, Atlıhan İ, Çayır E, Mega Tiber P, Orun O, Nigiz Ş, Özkul C, Gündüz MG, Küçükgüzel ŞG. Synthesis and Evaluation of Novel Metacetamol Derivatives with Hydrazone Moiety as Anticancer and Antimicrobial Agents. Chem Biodivers 2023; 20:e202300766. [PMID: 37417710 DOI: 10.1002/cbdv.202300766] [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: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Abstract
By exploiting the wide biological potential of the hydrazone scaffold, a series of hydrazone derivatives were synthesized, starting from N-(3-hydroxyphenyl)acetamide (metacetamol). The structures of the compounds were determined using IR, 1 H and 13 C-NMR, and mass spectroscopic methods. The obtained molecules (3 a-j) were evaluated for their anticancer potential against MDA-MB-231 and MCF-7 breast cancer cell lines. According to the CCK-8 assay, all tested compounds showed moderate to potent anticancer activity. Among them, N-(3-(2-(2-(4-nitrobenzylidene)hydrazinyl)-2-oxoethoxy)phenyl)acetamide (3 e) was found to be the most effective derivative with an IC50 value of 9.89 μM against MDA-MB-231 cell lines. This compound was further tested for its potential effects on the apoptotic pathway. Molecular docking studies was also carried out for 3 e in the colchicine binding pocket of tubulin. Additionally, compound 3 e also demonstrated effective antifungal activity, particularly against Candida krusei (MIC=8 μg/ml), indicating that nitro group at the 4th position of the phenyl ring was the most preferable substituent for both cytotoxic and antimicrobial activity. Our preliminary findings suggest that compound 3 e could be exploited as a leading structure for further anticancer and antifungal drug development.
Collapse
Affiliation(s)
- Sevil Şenkardeş
- Marmara University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Maltepe, Başıbüyük, 34854, Istanbul, Turkey
| | - İrem Atlıhan
- Marmara University, Institute of Health Sciences, Department of Biophysics, 34865, Istanbul, Turkey
| | - Elif Çayır
- Marmara University, Faculty of Pharmacy, 34854, Istanbul, Turkey
| | - Pınar Mega Tiber
- Marmara University, Faculty of Medicine, Department of Biophysics, 34854, Istanbul, Turkey
| | - Oya Orun
- Marmara University, Faculty of Medicine, Department of Biophysics, 34854, Istanbul, Turkey
| | - Şeyma Nigiz
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Sıhhiye, 06100, Ankara, Turkey
| | - Ceren Özkul
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Sıhhiye, 06100, Ankara, Turkey
| | - Miyase Gözde Gündüz
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Sıhhiye, 06100, Ankara, Turkey
| | - Ş Güniz Küçükgüzel
- Fenerbahçe University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ataşehir, 34758, Istanbul, Turkey
| |
Collapse
|
62
|
Lasser M, Sun N, Xu Y, Wang S, Drake S, Law K, Gonzalez S, Wang B, Drury V, Castillo O, Zaltsman Y, Dea J, Bader E, McCluskey KE, State MW, Willsey AJ, Willsey HR. Pleiotropy of autism-associated chromatin regulators. Development 2023; 150:dev201515. [PMID: 37366052 PMCID: PMC10399978 DOI: 10.1242/dev.201515] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/19/2023] [Indexed: 06/28/2023]
Abstract
Gene ontology analyses of high-confidence autism spectrum disorder (ASD) risk genes highlight chromatin regulation and synaptic function as major contributors to pathobiology. Our recent functional work in vivo has additionally implicated tubulin biology and cellular proliferation. As many chromatin regulators, including the ASD risk genes ADNP and CHD3, are known to directly regulate both tubulins and histones, we studied the five chromatin regulators most strongly associated with ASD (ADNP, CHD8, CHD2, POGZ and KMT5B) specifically with respect to tubulin biology. We observe that all five localize to microtubules of the mitotic spindle in vitro in human cells and in vivo in Xenopus. Investigation of CHD2 provides evidence that mutations present in individuals with ASD cause a range of microtubule-related phenotypes, including disrupted localization of the protein at mitotic spindles, cell cycle stalling, DNA damage and cell death. Lastly, we observe that ASD genetic risk is significantly enriched among tubulin-associated proteins, suggesting broader relevance. Together, these results provide additional evidence that the role of tubulin biology and cellular proliferation in ASD warrants further investigation and highlight the pitfalls of relying solely on annotated gene functions in the search for pathological mechanisms.
Collapse
Affiliation(s)
- Micaela Lasser
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nawei Sun
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yuxiao Xu
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sheng Wang
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sam Drake
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Karen Law
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Silvano Gonzalez
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Belinda Wang
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Langley Porter Psychiatric Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vanessa Drury
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Octavio Castillo
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yefim Zaltsman
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeanselle Dea
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ethel Bader
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kate E. McCluskey
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew W. State
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Langley Porter Psychiatric Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - A. Jeremy Willsey
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
63
|
Fu G, Yan S, Khoo CJ, Chao VC, Liu Z, Mukhi M, Hervas R, Li XD, Ti SC. Integrated regulation of tubulin tyrosination and microtubule stability by human α-tubulin isotypes. Cell Rep 2023; 42:112653. [PMID: 37379209 DOI: 10.1016/j.celrep.2023.112653] [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: 07/28/2022] [Revised: 05/03/2023] [Accepted: 05/31/2023] [Indexed: 06/30/2023] Open
Abstract
Tubulin isotypes are critical for the functions of cellular microtubules, which exhibit different stability and harbor various post-translational modifications. However, how tubulin isotypes determine the activities of regulators for microtubule stability and modifications remains unknown. Here, we show that human α4A-tubulin, a conserved genetically detyrosinated α-tubulin isotype, is a poor substrate for enzymatic tyrosination. To examine the stability of microtubules reconstituted with defined tubulin compositions, we develop a strategy to site-specifically label recombinant human tubulin for single-molecule TIRF microscopy-based in vitro assays. The incorporation of α4A-tubulin into the microtubule lattice stabilizes the polymers from passive and MCAK-stimulated depolymerization. Further characterization reveals that the compositions of α-tubulin isotypes and tyrosination/detyrosination states allow graded control for the microtubule binding and the depolymerization activities of MCAK. Together, our results uncover the tubulin isotype-dependent enzyme activity for an integrated regulation of α-tubulin tyrosination/detyrosination states and microtubule stability, two well-correlated features of cellular microtubules.
Collapse
Affiliation(s)
- Guoling Fu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Shan Yan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Chen Jing Khoo
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Victor C Chao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Zheng Liu
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Mayur Mukhi
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Rubén Hervas
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Xiang David Li
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Shih-Chieh Ti
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
64
|
Zocchi R, Bellacchio E, Piccione M, Scardigli R, D’Oria V, Petrini S, Baranano K, Bertini E, Sferra A. Novel loss of function mutation in TUBA1A gene compromises tubulin stability and proteostasis causing spastic paraplegia and ataxia. Front Cell Neurosci 2023; 17:1162363. [PMID: 37435044 PMCID: PMC10332271 DOI: 10.3389/fncel.2023.1162363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
Microtubules are dynamic cytoskeletal structures involved in several cellular functions, such as intracellular trafficking, cell division and motility. More than other cell types, neurons rely on the proper functioning of microtubules to conduct their activities and achieve complex morphologies. Pathogenic variants in genes encoding for α and β-tubulins, the structural subunits of microtubules, give rise to a wide class of neurological disorders collectively known as "tubulinopathies" and mainly involving a wide and overlapping range of brain malformations resulting from defective neuronal proliferation, migration, differentiation and axon guidance. Although tubulin mutations have been classically linked to neurodevelopmental defects, growing evidence demonstrates that perturbations of tubulin functions and activities may also drive neurodegeneration. In this study, we causally link the previously unreported missense mutation p.I384N in TUBA1A, one of the neuron-specific α-tubulin isotype I, to a neurodegenerative disorder characterized by progressive spastic paraplegia and ataxia. We demonstrate that, in contrast to the p.R402H substitution, which is one of the most recurrent TUBA1A pathogenic variants associated to lissencephaly, the present mutation impairs TUBA1A stability, reducing the abundance of TUBA1A available in the cell and preventing its incorporation into microtubules. We also show that the isoleucine at position 384 is an amino acid residue, which is critical for α-tubulin stability, since the introduction of the p.I384N substitution in three different tubulin paralogs reduces their protein level and assembly into microtubules, increasing their propensity to aggregation. Moreover, we demonstrate that the inhibition of the proteasome degradative systems increases the protein levels of TUBA1A mutant, promoting the formation of tubulin aggregates that, as their size increases, coalesce into inclusions that precipitate within the insoluble cellular fraction. Overall, our data describe a novel pathogenic effect of p.I384N mutation that differs from the previously described substitutions in TUBA1A, and expand both phenotypic and mutational spectrum related to this gene.
Collapse
Affiliation(s)
- Riccardo Zocchi
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Emanuele Bellacchio
- Molecular Genetics and Functional Genomics Research Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Michela Piccione
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
| | - Raffaella Scardigli
- Consiglio Nazionale delle Ricerche (CNR), Institute of Translational Pharmacology (IFT), Rome, Italy
- European Brain Research Institute (EBRI) “Rita Levi-Montalcini,” Rome, Italy
| | - Valentina D’Oria
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
| | - Stefania Petrini
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
| | - Kristin Baranano
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Enrico Bertini
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Antonella Sferra
- Unit of Neuromuscular Disorders, Translational Pediatrics and Clinical Genetics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
65
|
Shen Y, Ori-McKenney KM. Macromolecular Crowding Tailors the Microtubule Cytoskeleton Through Tubulin Modifications and Microtubule-Associated Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.14.544846. [PMID: 37398431 PMCID: PMC10312695 DOI: 10.1101/2023.06.14.544846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Cells remodel their cytoskeletal networks to adapt to their environment. Here, we analyze the mechanisms utilized by the cell to tailor its microtubule landscape in response to changes in osmolarity that alter macromolecular crowding. By integrating live cell imaging, ex vivo enzymatic assays, and in vitro reconstitution, we probe the impact of acute perturbations in cytoplasmic density on microtubule-associated proteins (MAPs) and tubulin posttranslational modifications (PTMs), unraveling the molecular underpinnings of cellular adaptation via the microtubule cytoskeleton. We find that cells respond to fluctuations in cytoplasmic density by modulating microtubule acetylation, detyrosination, or MAP7 association, without differentially affecting polyglutamylation, tyrosination, or MAP4 association. These MAP-PTM combinations alter intracellular cargo transport, enabling the cell to respond to osmotic challenges. We further dissect the molecular mechanisms governing tubulin PTM specification, and find that MAP7 promotes acetylation by biasing the conformation of the microtubule lattice, and directly inhibits detyrosination. Acetylation and detyrosination can therefore be decoupled and utilized for distinct cellular purposes. Our data reveal that the MAP code dictates the tubulin code, resulting in remodeling of the microtubule cytoskeleton and alteration of intracellular transport as an integrated mechanism of cellular adaptation.
Collapse
Affiliation(s)
- Yusheng Shen
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Kassandra M Ori-McKenney
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
66
|
Zhou P, Yang G, Xie W. Organization of cortical microtubules in differentiated cells. J Cell Physiol 2023; 238:1141-1147. [PMID: 36960617 DOI: 10.1002/jcp.31011] [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: 01/25/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023]
Abstract
The microtubule cytoskeleton plays a critical role in a variety of cellular activities, and its structures and functions have been extensively studied. However, little is known about cell differentiation-related microtubule remodeling, its regulatory mechanisms, and its physiological functions. Recent studies have shown that microtubule-binding proteins as well as cell junctions, such as desmosomes and adherens junctions, are involved in the remodeling of microtubules in response to cell differentiation. In addition, the microtubule-organizing activity and structural integrity of centrosomes undergo dramatic changes during cell differentiation to promote microtubule remodeling. Here we summarize recent advances revealing the dynamic changes in microtubule organization and functions during cell differentiation. We also highlight the molecular mechanisms underlying microtubule modeling in differentiated cells, focusing on the key roles played by microtubule-binding proteins, cell junctions, and centrosomes.
Collapse
Affiliation(s)
- Ping Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Guiwen Yang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Wei Xie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
67
|
Chen J, Roll-Mecak A. Glutamylation is a negative regulator of microtubule growth. Mol Biol Cell 2023; 34:ar70. [PMID: 37074962 PMCID: PMC10295482 DOI: 10.1091/mbc.e23-01-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/20/2023] Open
Abstract
Microtubules are noncovalent polymers built from αβ-tubulin dimers. The disordered C-terminal tubulin tails are functionalized with multiple glutamate chains of variable lengths added and removed by tubulin tyrosine ligases (TTLLs) and carboxypeptidases (CCPs). Glutamylation is abundant on stable microtubule arrays such as in axonemes and axons, and its dysregulation leads to human pathologies. Despite this, the effects of glutamylation on intrinsic microtubule dynamics are unclear. Here we generate tubulin with short and long glutamate chains and show that glutamylation slows the rate of microtubule growth and increases catastrophes as a function of glutamylation levels. This implies that the higher stability of glutamylated microtubules in cells is due to effectors. Interestingly, EB1 is minimally affected by glutamylation and thus can report on the growth rates of both unmodified and glutamylated microtubules. Finally, we show that glutamate removal by CCP1 and 5 is synergistic and occurs preferentially on soluble tubulin, unlike TTLL enzymes that prefer microtubules. This substrate preference establishes an asymmetry whereby once the microtubule depolymerizes, the released tubulin is reset to a less-modified state, while polymerized tubulin accumulates the glutamylation mark. Our work shows that a modification on the disordered tubulin tails can directly affect microtubule dynamics and furthers our understanding of the mechanistic underpinnings of the tubulin code.
Collapse
Affiliation(s)
- Jiayi Chen
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, and
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, and
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD 20892
| |
Collapse
|
68
|
He J, Zhu Q, Han P, Zhou T, Li J, Wang X, Cheng J. Transcriptomic Networks Reveal the Tissue-Specific Cold Shock Responses in Japanese Flounder ( Paralichthys olivaceus). BIOLOGY 2023; 12:784. [PMID: 37372069 DOI: 10.3390/biology12060784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023]
Abstract
Low temperature is among the important factors affecting the distribution, survival, growth, and physiology of aquatic animals. In this study, coordinated transcriptomic responses to 10 °C acute cold stress were investigated in the gills, hearts, livers, and spleens of Japanese flounder (Paralichthys olivaceus), an important aquaculture species in east Asia. Histological examination suggested different levels of injury among P. olivaceus tissues after cold shock, mainly in the gills and livers. Based on transcriptome and weighted gene coexpression network analysis, 10 tissue-specific cold responsive modules (CRMs) were identified, revealing a cascade of cellular responses to cold stress. Specifically, five upregulated CRMs were enriched with induced differentially expressed genes (DEGs), mainly corresponding to the functions of "extracellular matrix", "cytoskeleton", and "oxidoreductase activity", indicating the induced cellular response to cold shock. The "cell cycle/division" and "DNA complex" functions were enriched in the downregulated CRMs for all four tissues, which comprised inhibited DEGs, suggesting that even with tissue-specific responses, cold shock may induce severely disrupted cellular functions in all tissues, reducing aquaculture productivity. Therefore, our results revealed the tissue-specific regulation of the cellular response to low-temperature stress, which warrants further investigation and provides more comprehensive insights for the conservation and cultivation of P. olivaceus in cold water.
Collapse
Affiliation(s)
- Jiayi He
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
| | - Qing Zhu
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
| | - Ping Han
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
| | - Tianyu Zhou
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
| | - Juyan Li
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
| | - Xubo Wang
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, 169 Qixingnan Road, Ningbo 315832, China
| | - Jie Cheng
- Key Laboratory of Marine Genetics and Breeding (Ocean University of China), Ministry of Education, 5 Yushan Road, Qingdao 266003, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572024, China
- Laboratory for Marine Fisheries Science and Food Production Processes, National Laboratory for Marine Science and Technology (Qingdao), 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
69
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
70
|
Jansen KI, Iwanski MK, Burute M, Kapitein LC. A live-cell marker to visualize the dynamics of stable microtubules throughout the cell cycle. J Cell Biol 2023; 222:e202106105. [PMID: 36880745 PMCID: PMC9998657 DOI: 10.1083/jcb.202106105] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 01/08/2022] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
The microtubule (MT) cytoskeleton underlies processes such as intracellular transport and cell division. Immunolabeling for posttranslational modifications of tubulin has revealed the presence of different MT subsets, which are believed to differ in stability and function. Whereas dynamic MTs can readily be studied using live-cell plus-end markers, the dynamics of stable MTs have remained obscure due to a lack of tools to directly visualize these MTs in living cells. Here, we present StableMARK (Stable Microtubule-Associated Rigor-Kinesin), a live-cell marker to visualize stable MTs with high spatiotemporal resolution. We demonstrate that a rigor mutant of Kinesin-1 selectively binds to stable MTs without affecting MT organization and organelle transport. These MTs are long-lived, undergo continuous remodeling, and often do not depolymerize upon laser-based severing. Using this marker, we could visualize the spatiotemporal regulation of MT stability before, during, and after cell division. Thus, this live-cell marker enables the exploration of different MT subsets and how they contribute to cellular organization and transport.
Collapse
Affiliation(s)
- Klara I. Jansen
- Department of Biology, Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Malina K. Iwanski
- Department of Biology, Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Mithila Burute
- Department of Biology, Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Lukas C. Kapitein
- Department of Biology, Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
71
|
Fermino do Rosário C, Wadsworth P. Measurement of Microtubule Stability in Mammalian Cells. Curr Protoc 2023; 3:e793. [PMID: 37235484 DOI: 10.1002/cpz1.793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The microtubule cytoskeleton is essential for various biological processes such as the intracellular distribution of molecules and organelles, cell morphogenesis, chromosome segregation, and specification of the location of contractile ring formation. Distinct cell types contain microtubules with different extents of stability. For example, microtubules in neurons are highly stabilized to support organelle (or vesicular) transport over large distances, and microtubules in motile cells are more dynamic. In some cases, such as the mitotic spindle, both dynamic and stable microtubules coexist. Alteration of microtubule stability is connected to disease states, making understanding microtubule stability an important area of research. Methods to measure microtubule stability in mammalian cells are described here. Together, these approaches allow microtubule stability to be measured qualitatively or semiquantitatively following staining for post-translational modifications of tubulin or treating cells with microtubule destabilizing agents such as nocodazole. Microtubule stability can also be measured quantitatively by performing fluorescence recovery after photobleaching or fluorescence photoactivation of tubulin in live cells. These methods should be helpful for those seeking to understand microtubule dynamics and stabilization. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Fixing and staining cells for tubulin post-translational modifications Basic Protocol 2: Evaluating microtubule stability following treatment with nocodazole in live or fixed cells Basic Protocol 3: Measurement of microtubule dynamic turnover by quantification of fluorescence recovery after photobleaching Basic Protocol 4: Measurement of microtubule dynamic turnover by quantification of dissipation of fluorescence after photoactivation.
Collapse
Affiliation(s)
- Carline Fermino do Rosário
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Patricia Wadsworth
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, Massachusetts
| |
Collapse
|
72
|
Carmona B, Marinho HS, Matos CL, Nolasco S, Soares H. Tubulin Post-Translational Modifications: The Elusive Roles of Acetylation. BIOLOGY 2023; 12:biology12040561. [PMID: 37106761 PMCID: PMC10136095 DOI: 10.3390/biology12040561] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
Microtubules (MTs), dynamic polymers of α/β-tubulin heterodimers found in all eukaryotes, are involved in cytoplasm spatial organization, intracellular transport, cell polarity, migration and division, and in cilia biology. MTs functional diversity depends on the differential expression of distinct tubulin isotypes and is amplified by a vast number of different post-translational modifications (PTMs). The addition/removal of PTMs to α- or β-tubulins is catalyzed by specific enzymes and allows combinatory patterns largely enriching the distinct biochemical and biophysical properties of MTs, creating a code read by distinct proteins, including microtubule-associated proteins (MAPs), which allow cellular responses. This review is focused on tubulin-acetylation, whose cellular roles continue to generate debate. We travel through the experimental data pointing to α-tubulin Lys40 acetylation role as being a MT stabilizer and a typical PTM of long lived MTs, to the most recent data, suggesting that Lys40 acetylation enhances MT flexibility and alters the mechanical properties of MTs, preventing MTs from mechanical aging characterized by structural damage. Additionally, we discuss the regulation of tubulin acetyltransferases/desacetylases and their impacts on cell physiology. Finally, we analyze how changes in MT acetylation levels have been found to be a general response to stress and how they are associated with several human pathologies.
Collapse
Affiliation(s)
- Bruno Carmona
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
| | - H Susana Marinho
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Catarina Lopes Matos
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Sofia Nolasco
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Helena Soares
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Av. D. João II, Lote 4.69.01, 1990-096 Lisboa, Portugal
| |
Collapse
|
73
|
Novotná Floriančičová K, Baltzis A, Smejkal J, Czerneková M, Kaczmarek Ł, Malý J, Notredame C, Vinopal S. Phylogenetic and functional characterization of water bears (Tardigrada) tubulins. Sci Rep 2023; 13:5194. [PMID: 36997657 PMCID: PMC10063605 DOI: 10.1038/s41598-023-31992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/21/2023] [Indexed: 04/01/2023] Open
Abstract
Tardigrades are microscopic ecdysozoans that can withstand extreme environmental conditions. Several tardigrade species undergo reversible morphological transformations and enter into cryptobiosis, which helps them to survive periods of unfavorable environmental conditions. However, the underlying molecular mechanisms of cryptobiosis are mostly unknown. Tubulins are evolutionarily conserved components of the microtubule cytoskeleton that are crucial in many cellular processes. We hypothesize that microtubules are necessary for the morphological changes associated with successful cryptobiosis. The molecular composition of the microtubule cytoskeleton in tardigrades is unknown. Therefore, we analyzed and characterized tardigrade tubulins and identified 79 tardigrade tubulin sequences in eight taxa. We found three α-, seven β-, one γ-, and one ε-tubulin isoform. To verify in silico identified tardigrade tubulins, we also isolated and sequenced nine out of ten predicted Hypsibius exemplaris tubulins. All tardigrade tubulins were localized as expected when overexpressed in mammalian cultured cells: to the microtubules or to the centrosomes. The presence of a functional ε-tubulin, clearly localized to centrioles, is attractive from a phylogenetic point of view. Although the phylogenetically close Nematoda lost their δ- and ε-tubulins, some groups of Arthropoda still possess them. Thus, our data support the current placement of tardigrades into the Panarthropoda clade.
Collapse
Affiliation(s)
- Kamila Novotná Floriančičová
- Department of Biology, Faculty of Science, J. E. Purkyně University (UJEP), Usti Nad Labem, Czech Republic
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic
| | | | - Jiří Smejkal
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic
| | - Michaela Czerneková
- Department of Biology, Faculty of Science, J. E. Purkyně University (UJEP), Usti Nad Labem, Czech Republic
| | - Łukasz Kaczmarek
- Department of Animal Taxonomy and Ecology, Adam Mickiewicz University in Poznań, Poznań, Poland
| | - Jan Malý
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic
| | - Cedric Notredame
- Centre for Genomic Regulation, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Stanislav Vinopal
- Department of Biology, Faculty of Science, J. E. Purkyně University (UJEP), Usti Nad Labem, Czech Republic.
- Centre for Nanotechnology and Biotechnology, Faculty of Science, UJEP, Usti Nad Labem, Czech Republic.
| |
Collapse
|
74
|
Iwanski MK, Kapitein LC. Cellular cartography: Towards an atlas of the neuronal microtubule cytoskeleton. Front Cell Dev Biol 2023; 11:1052245. [PMID: 37035244 PMCID: PMC10073685 DOI: 10.3389/fcell.2023.1052245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Microtubules, one of the major components of the cytoskeleton, play a crucial role during many aspects of neuronal development and function, such as neuronal polarization and axon outgrowth. Consequently, the microtubule cytoskeleton has been implicated in many neurodevelopmental and neurodegenerative disorders. The polar nature of microtubules is quintessential for their function, allowing them to serve as tracks for long-distance, directed intracellular transport by kinesin and dynein motors. Most of these motors move exclusively towards either the plus- or minus-end of a microtubule and some have been shown to have a preference for either dynamic or stable microtubules, those bearing a particular post-translational modification or those decorated by a specific microtubule-associated protein. Thus, it becomes important to consider the interplay of these features and their combinatorial effects on transport, as well as how different types of microtubules are organized in the cell. Here, we discuss microtubule subsets in terms of tubulin isotypes, tubulin post-translational modifications, microtubule-associated proteins, microtubule stability or dynamicity, and microtubule orientation. We highlight techniques used to study these features of the microtubule cytoskeleton and, using the information from these studies, try to define the composition, role, and organization of some of these subsets in neurons.
Collapse
Affiliation(s)
| | - Lukas C. Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
75
|
Akera T. Tubulin post-translational modifications in meiosis. Semin Cell Dev Biol 2023; 137:38-45. [PMID: 34836784 PMCID: PMC9124733 DOI: 10.1016/j.semcdb.2021.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/22/2021] [Accepted: 11/14/2021] [Indexed: 11/18/2022]
Abstract
Haploid gametes are produced from diploid parents through meiosis, a process inherent to all sexually reproducing eukaryotes. Faithful chromosome segregation in meiosis is essential for reproductive success, although it is less clear how the meiotic spindle achieves this compared to the mitotic spindle. It is becoming increasingly clear that tubulin post-translational modifications (PTMs) play critical roles in regulating microtubule functions in many biological processes, and meiosis is no exception. Here, I review recent advances in the understanding of tubulin PTMs in meiotic spindles, especially focusing on their roles in spindle integrity, oocyte aging, and non-Mendelian transmission.
Collapse
Affiliation(s)
- Takashi Akera
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda 20892, MD, USA.
| |
Collapse
|
76
|
Verhey KJ, Ohi R. Causes, costs and consequences of kinesin motors communicating through the microtubule lattice. J Cell Sci 2023; 136:293511. [PMID: 36866642 PMCID: PMC10022682 DOI: 10.1242/jcs.260735] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Microtubules are critical for a variety of important functions in eukaryotic cells. During intracellular trafficking, molecular motor proteins of the kinesin superfamily drive the transport of cellular cargoes by stepping processively along the microtubule surface. Traditionally, the microtubule has been viewed as simply a track for kinesin motility. New work is challenging this classic view by showing that kinesin-1 and kinesin-4 proteins can induce conformational changes in tubulin subunits while they are stepping. These conformational changes appear to propagate along the microtubule such that the kinesins can work allosterically through the lattice to influence other proteins on the same track. Thus, the microtubule is a plastic medium through which motors and other microtubule-associated proteins (MAPs) can communicate. Furthermore, stepping kinesin-1 can damage the microtubule lattice. Damage can be repaired by the incorporation of new tubulin subunits, but too much damage leads to microtubule breakage and disassembly. Thus, the addition and loss of tubulin subunits are not restricted to the ends of the microtubule filament but rather, the lattice itself undergoes continuous repair and remodeling. This work leads to a new understanding of how kinesin motors and their microtubule tracks engage in allosteric interactions that are critical for normal cell physiology.
Collapse
Affiliation(s)
- Kristen J. Verhey
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Author for correspondence ()
| | - Ryoma Ohi
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
77
|
Genova M, Grycova L, Puttrich V, Magiera MM, Lansky Z, Janke C, Braun M. Tubulin polyglutamylation differentially regulates microtubule-interacting proteins. EMBO J 2023; 42:e112101. [PMID: 36636822 PMCID: PMC9975938 DOI: 10.15252/embj.2022112101] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
Tubulin posttranslational modifications have been predicted to control cytoskeletal functions by coordinating the molecular interactions between microtubules and their associating proteins. A prominent tubulin modification in neurons is polyglutamylation, the deregulation of which causes neurodegeneration. Yet, the underlying molecular mechanisms have remained elusive. Here, using in-vitro reconstitution, we determine how polyglutamylation generated by the two predominant neuronal polyglutamylases, TTLL1 and TTLL7, specifically modulates the activities of three major microtubule interactors: the microtubule-associated protein Tau, the microtubule-severing enzyme katanin and the molecular motor kinesin-1. We demonstrate that the unique modification patterns generated by TTLL1 and TTLL7 differentially impact those three effector proteins, thus allowing for their selective regulation. Given that our experiments were performed with brain tubulin from mouse models in which physiological levels and patterns of polyglutamylation were altered by the genetic knockout of the main modifying enzymes, our quantitative measurements provide direct mechanistic insight into how polyglutamylation could selectively control microtubule interactions in neurons.
Collapse
Affiliation(s)
- Mariya Genova
- Institut Curie, Université PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Lenka Grycova
- Institute of BiotechnologyCzech Academy of Sciences, BIOCEVPrague WestCzech Republic
| | - Verena Puttrich
- Institute of BiotechnologyCzech Academy of Sciences, BIOCEVPrague WestCzech Republic
| | - Maria M Magiera
- Institut Curie, Université PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Zdenek Lansky
- Institute of BiotechnologyCzech Academy of Sciences, BIOCEVPrague WestCzech Republic
| | - Carsten Janke
- Institut Curie, Université PSL, CNRS UMR3348OrsayFrance
- Université Paris‐Saclay, CNRS UMR3348OrsayFrance
| | - Marcus Braun
- Institute of BiotechnologyCzech Academy of Sciences, BIOCEVPrague WestCzech Republic
| |
Collapse
|
78
|
Zhang L, Wei X, Wang Z, Liu P, Hou Y, Xu Y, Su H, Koci MD, Yin H, Zhang C. NF-κB activation enhances STING signaling by altering microtubule-mediated STING trafficking. Cell Rep 2023; 42:112185. [PMID: 36857187 DOI: 10.1016/j.celrep.2023.112185] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/12/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
It is widely known that stimulator of interferon genes (STING) can trigger nuclear factor κB (NF-κB) signaling. However, whether and how the NF-κB pathway affects STING signaling remains largely unclear. Here, we report that Toll-like receptor (TLR)-, interleukin-1 receptor (IL-1R)-, tumor necrosis factor receptor (TNFR)-, growth factor receptor (GF-R)-, and protein kinase C (PKC)-mediated NF-κB signaling activation dramatically enhances STING-mediated immune responses. Mechanistically, we find that STING interacts with microtubules, which plays a crucial role in STING intracellular trafficking. We further uncover that activation of the canonical NF-κB pathway induces microtubule depolymerization, which inhibits STING trafficking to lysosomes for degradation. This leads to increased levels of activated STING that persist for a longer period of time. The synergy between NF-κB and STING triggers a cascade-amplified interferon response and robust host antiviral defense. In addition, we observe that several gain-of-function mutations of STING abolish the microtubule-STING interaction and cause abnormal STING trafficking and ligand-independent STING autoactivation. Collectively, our data demonstrate that NF-κB activation enhances STING signaling by regulating microtubule-mediated STING trafficking.
Collapse
Affiliation(s)
- Lulu Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xubiao Wei
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Zhimeng Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Peiyuan Liu
- School of Life Science, Tianjin University, Tianjin, China
| | - Yanfei Hou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yifang Xu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Huili Su
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Matthew D Koci
- Prestage Department of Poultry Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA
| | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| | - Conggang Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
79
|
The interconnection of endoplasmic reticulum and microtubule and its implication in Hereditary Spastic Paraplegia. Comput Struct Biotechnol J 2023; 21:1670-1677. [PMID: 36860342 PMCID: PMC9968982 DOI: 10.1016/j.csbj.2023.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The endoplasmic reticulum (ER) and microtubule (MT) network form extensive contact with each other and their interconnection plays a pivotal role in ER maintenance and distribution as well as MT stability. The ER participates in a variety of biological processes including protein folding and processing, lipid biosynthesis, and Ca2+ storage. MTs specifically regulate cellular architecture, provide routes for transport of molecules or organelles, and mediate signaling events. The ER morphology and dynamics are regulated by a class of ER shaping proteins, which also provide the physical contact structure for linking of ER and MT. In addition to these ER-localized and MT-binding proteins, specific motor proteins and adaptor-linking proteins also mediate bidirectional communication between the two structures. In this review, we summarize the current understanding of the structure and function of ER-MT interconnection. We further highlight the morphologic factors which coordinate the ER-MT network and maintain the normal physiological function of neurons, with their defect causing neurodegenerative diseases such as Hereditary Spastic Paraplegia (HSP). These findings promote our understanding of the pathogenesis of HSP and provide important therapeutic targets for treatment of these diseases.
Collapse
|
80
|
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
|
81
|
Zhen J, Zhou Z, He M, Han HX, Lv EH, Wen PB, Liu X, Wang YT, Cai XC, Tian JQ, Zhang MY, Xiao L, Kang XX. The gut microbial metabolite trimethylamine N-oxide and cardiovascular diseases. Front Endocrinol (Lausanne) 2023; 14:1085041. [PMID: 36824355 PMCID: PMC9941174 DOI: 10.3389/fendo.2023.1085041] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Morbidity and mortality of cardiovascular diseases (CVDs) are exceedingly high worldwide. Researchers have found that the occurrence and development of CVDs are closely related to intestinal microecology. Imbalances in intestinal microecology caused by changes in the composition of the intestinal microbiota will eventually alter intestinal metabolites, thus transforming the host physiological state from healthy mode to pathological mode. Trimethylamine N-oxide (TMAO) is produced from the metabolism of dietary choline and L-carnitine by intestinal microbiota, and many studies have shown that this important product inhibits cholesterol metabolism, induces platelet aggregation and thrombosis, and promotes atherosclerosis. TMAO is directly or indirectly involved in the pathogenesis of CVDs and is an important risk factor affecting the occurrence and even prognosis of CVDs. This review presents the biological and chemical characteristics of TMAO, and the process of TMAO produced by gut microbiota. In particular, the review focuses on summarizing how the increase of gut microbial metabolite TMAO affects CVDs including atherosclerosis, heart failure, hypertension, arrhythmia, coronary artery disease, and other CVD-related diseases. Understanding the mechanism of how increases in TMAO promotes CVDs will potentially facilitate the identification and development of targeted therapy for CVDs.
Collapse
Affiliation(s)
- Jing Zhen
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
- School of Chemical Engineering and Technology, China University of Mining and Technology, Xuzhou, Jiangsu, China
| | - Zhou Zhou
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng He
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hai-Xiang Han
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - En-Hui Lv
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peng-Bo Wen
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Liu
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan-Ting Wang
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xun-Chao Cai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Jia-Qi Tian
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng-Ying Zhang
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei Xiao
- School of Chemical Engineering and Technology, China University of Mining and Technology, Xuzhou, Jiangsu, China
| | - Xing-Xing Kang
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
82
|
Zaniewski TM, Hancock WO. Positive charge in the K-loop of the kinesin-3 motor KIF1A regulates superprocessivity by enhancing microtubule affinity in the one-head-bound state. J Biol Chem 2023; 299:102818. [PMID: 36549649 PMCID: PMC9871336 DOI: 10.1016/j.jbc.2022.102818] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
KIF1A is an essential neuronal transport motor protein in the kinesin-3 family, known for its superprocessive motility. However, structural features underlying this function are unclear. Here, we determined that superprocessivity of KIF1A dimers originates from a unique structural domain, the lysine-rich insertion in loop-12 termed the 'K-loop', which enhances electrostatic interactions between the motor and the microtubule. In 80 mM PIPES buffer, replacing the native KIF1A loop-12 with that of kinesin-1 resulted in a 6-fold decrease in run length, whereas adding additional positive charge to loop-12 enhanced the run length. Interestingly, swapping the KIF1A loop-12 into kinesin-1 did not enhance its run length, consistent with the two motor families using different mechanochemical tuning to achieve persistent transport. To investigate the mechanism by which the KIF1A K-loop enhances processivity, we used microtubule pelleting and single-molecule dwell time assays in ATP and ADP. First, the microtubule affinity was similar in ATP and in ADP, consistent with the motor spending the majority of its cycle in a weakly bound state. Second, the microtubule affinity and single-molecule dwell time in ADP were 6-fold lower in the loop-swap mutant than WT. Thus, the positive charge in loop-12 of KIF1A enhances the run length by stabilizing binding of the motor in its vulnerable one-head-bound state. Finally, through a series of mutants with varying positive charge in the K-loop, we found that KIF1A processivity is linearly dependent on the charge of loop-12, further highlighting how loop-12 contributes to the function of this key motor protein.
Collapse
Affiliation(s)
- Taylor M Zaniewski
- Departments of Chemistry and Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - William O Hancock
- Departments of Chemistry and Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA.
| |
Collapse
|
83
|
Eiró-Quirino L, Lima WFD, Aragão WAB, Bittencourt LO, Mendes PFS, Fernandes RM, Rodrigues CA, Dionízio A, Buzalaf MAR, Monteiro MC, Cirovic A, Cirovic A, Puty B, Lima RR. Exposure to tolerable concentrations of aluminum triggers systemic and local oxidative stress and global proteomic modulation in the spinal cord of rats. CHEMOSPHERE 2023; 313:137296. [PMID: 36410523 DOI: 10.1016/j.chemosphere.2022.137296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
The tolerable aluminum (Al) intake levels for humans are constantly under review by regulatory agencies due to novel pre-clinical evidence on the neurotoxicity of prolonged Al exposure; however, little is known about the effects of Al on the spinal cord. This study aimed to investigate potential adverse effects on both spinal cord and systemic biochemical balance after prolonged exposure to a low dose of Al. Twenty adult rats were distributed in the control (distilled water) and exposed group (8.3 mg of AlCl3/kg/day). After 60 days, both blood and spinal cord samples were collected for oxidative stress and proteomic analyses. In plasma and erythrocytes, glutathione level was not different between groups; however, exposure to AlCl3 significantly decreased glutathione level in the spinal cord. Thiobarbituric acid reactive substances levels in the plasma and spinal cord of animals from the control group were significantly lower than those animals exposed to AlCl3. Exposure to AlCl3 significantly modulated the expression of proteins associated with the cell cycle, stimulus-response, cytoskeleton, nervous system regulation, protein activity, and synaptic signaling. Therefore, prolonged exposure to a low dose of Al triggered oxidative stress and proteomic changes that may affect spinal cord homeostasis.
Collapse
Affiliation(s)
- Luciana Eiró-Quirino
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Weslley Ferreira de Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Walessa Alana Bragança Aragão
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Paulo Fernando Santos Mendes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Rafael Monteiro Fernandes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Caroline Azulay Rodrigues
- Laboratory of Clinical Immunology and Oxidative Stress, Pharmacy Faculty, Institute of Health Sciences, Federal University of Pará, Belém, Brazil
| | - Aline Dionízio
- Bauru School of Dentistry, Department of Biological Sciences, University of São Paulo, Bauru, Brazil
| | | | - Marta Chagas Monteiro
- Laboratory of Clinical Immunology and Oxidative Stress, Pharmacy Faculty, Institute of Health Sciences, Federal University of Pará, Belém, Brazil
| | - Ana Cirovic
- University of Belgrade, Faculty of Medicine, Institute of Anatomy, Belgrade, Serbia
| | - Aleksandar Cirovic
- University of Belgrade, Faculty of Medicine, Institute of Anatomy, Belgrade, Serbia
| | - Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil.
| |
Collapse
|
84
|
Guan G, Cannon RD, Coates DE, Mei L. Effect of the Rho-Kinase/ROCK Signaling Pathway on Cytoskeleton Components. Genes (Basel) 2023; 14:272. [PMID: 36833199 PMCID: PMC9957420 DOI: 10.3390/genes14020272] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The mechanical properties of cells are important in tissue homeostasis and enable cell growth, division, migration and the epithelial-mesenchymal transition. Mechanical properties are determined to a large extent by the cytoskeleton. The cytoskeleton is a complex and dynamic network composed of microfilaments, intermediate filaments and microtubules. These cellular structures confer both cell shape and mechanical properties. The architecture of the networks formed by the cytoskeleton is regulated by several pathways, a key one being the Rho-kinase/ROCK signaling pathway. This review describes the role of ROCK (Rho-associated coiled-coil forming kinase) and how it mediates effects on the key components of the cytoskeleton that are critical for cell behaviour.
Collapse
Affiliation(s)
- Guangzhao Guan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Diagnostic and Surgical Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Richard D. Cannon
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Dawn E. Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Li Mei
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| |
Collapse
|
85
|
Fang CT, Kuo HH, Amartuvshin O, Hsu HJ, Liu SL, Yao JS, Yih LH. Inhibition of acetyl-CoA carboxylase impaired tubulin palmitoylation and induced spindle abnormalities. Cell Death Dis 2023; 9:4. [PMID: 36617578 PMCID: PMC9826786 DOI: 10.1038/s41420-023-01301-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/20/2022] [Accepted: 01/03/2023] [Indexed: 01/10/2023]
Abstract
Tubulin s-palmitoylation involves the thioesterification of a cysteine residue in tubulin with palmitate. The palmitate moiety is produced by the fatty acid synthesis pathway, which is rate-limited by acetyl-CoA carboxylase (ACC). While it is known that ACC is phosphorylated at serine 79 (pSer79) by AMPK and accumulates at the spindle pole (SP) during mitosis, a functional role for tubulin palmitoylation during mitosis has not been identified. In this study, we found that modulating pSer79-ACC level at the SP using AMPK agonist and inhibitor induced spindle defects. Loss of ACC function induced spindle abnormalities in cell lines and in germ cells of the Drosophila germarium, and palmitic acid (PA) rescued the spindle defects in the cell line treated transiently with the ACC inhibitor, TOFA. Furthermore, inhibition of protein palmitoylating or depalmitoylating enzymes also induced spindle defects. Together, these data suggested that precisely regulated cellular palmitate level and protein palmitoylation may be required for accurate spindle assembly. We then showed that tubulin was largely palmitoylated in interphase cells but less palmitoylated in mitotic cells. TOFA treatment diminished tubulin palmitoylation at doses that disrupt microtubule (MT) instability and cause spindle defects. Moreover, spindle MTs comprised of α-tubulins mutated at the reported palmitoylation site exhibited disrupted dynamic instability. We also found that TOFA enhanced the MT-targeting drug-induced spindle abnormalities and cytotoxicity. Thus, our study reveals that precise regulation of ACC during mitosis impacts tubulin palmitoylation to delicately control MT dynamic instability and spindle assembly, thereby safeguarding nuclear and cell division.
Collapse
Affiliation(s)
- Chieh-Ting Fang
- grid.506933.a0000 0004 0633 7835Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hsiao-Hui Kuo
- grid.506933.a0000 0004 0633 7835Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Oyundari Amartuvshin
- grid.506933.a0000 0004 0633 7835Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan ,grid.28665.3f0000 0001 2287 1366Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan ,grid.260565.20000 0004 0634 0356Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Hwei-Jan Hsu
- grid.506933.a0000 0004 0633 7835Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan ,grid.28665.3f0000 0001 2287 1366Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan ,grid.260565.20000 0004 0634 0356Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Sih-Long Liu
- grid.506933.a0000 0004 0633 7835Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jhong-Syuan Yao
- grid.506933.a0000 0004 0633 7835Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Ling-Huei Yih
- grid.506933.a0000 0004 0633 7835Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
86
|
Cason SE, Fenton AR, Holzbaur ELF. Employing Live-Cell Imaging to Study Motor-Mediated Transport. Methods Mol Biol 2023; 2623:45-59. [PMID: 36602678 DOI: 10.1007/978-1-0716-2958-1_3] [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] [Indexed: 06/17/2023]
Abstract
Microtubule-based transport is a highly regulated process, requiring kinesin and/or dynein motors, a multitude of motor-associated regulatory proteins including activating adaptors and scaffolding proteins, and microtubule tracks that also provide regulatory cues. While in vitro studies are invaluable, fully replicating the physiological conditions under which motility occurs in cells is not yet possible. Here, we describe two methods that can be employed to study motor-based transport and motor regulation in a cellular context. Live-cell imaging of organelle transport in neurons leverages the uniform polarity of microtubules in axons to better understand the factors regulating microtubule-based motility. Peroxisome recruitment assays allow users to examine the net effect of motors and motor-regulatory proteins on organelle distribution. Together, these methods open the door to motility experiments that more fully interrogate the complex cellular environment.
Collapse
Affiliation(s)
- Sydney E Cason
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Adam R Fenton
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
87
|
Satouh Y, Sato K. Reorganization, specialization, and degradation of oocyte maternal components for early development. Reprod Med Biol 2023; 22:e12505. [PMID: 36726596 PMCID: PMC9884333 DOI: 10.1002/rmb2.12505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/30/2023] Open
Abstract
Background Oocyte components are maternally provided, solely determine oocyte quality, and coordinately determine embryo quality with zygotic gene expression. During oocyte maturation, maternal organelles are drastically reorganized and specialized to support oocyte characteristics. A large number of maternal components are actively degraded after fertilization and gradually replaced by zygotic gene products. The molecular basis and the significance of these processes on oocyte/embryo quality are not fully understood. Methods Firstly, recent findings in organelle characteristics of other cells or oocytes from model organisms are introduced for further understanding of oocyte organelle reorganization/specialization. Secondly, recent progress in studies on maternal components degradation and their molecular mechanisms are introduced. Finally, future applications of these advancements for predicting mammalian oocyte/embryo quality are discussed. Main findings The significance of cellular surface protein degradation via endocytosis for embryonic development, and involvement of biogenesis of lipid droplets in embryonic quality, were recently reported using mammalian model organisms. Conclusion Identifying key oocyte component characteristics and understanding their dynamics may lead to new applications in oocyte/embryo quality prediction and improvement. To implement these multidimensional concepts, development of new technical approaches that allow us to address the complexity and efficient studies using model organisms are required.
Collapse
Affiliation(s)
- Yuhkoh Satouh
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular RegulationGunma UniversityMaebashiJapan
| | - Ken Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular RegulationGunma UniversityMaebashiJapan
| |
Collapse
|
88
|
Li T, Chen Q, Zhang Q, Feng T, Zhang J, Lin Y, Yang P, He S, Zhang H. Transcriptomic Analysis on the Effects of Altered Water Temperature Regime on the Fish Ovarian Development of Coreius guichenoti under the Impact of River Damming. BIOLOGY 2022; 11:1829. [PMID: 36552338 PMCID: PMC9775624 DOI: 10.3390/biology11121829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
Field investigation indicated that the reduction in fish spawning was associated with the alteration in water temperatures, even a 2-3 °C monthly difference due to reservoir operations. However, the physiological mechanism that influences the development of fish ovary (DFO) remains unclear. Thus, experiments of Coreius guichenoti were conducted at three different temperatures, optimal temperature (~20 °C, N) for fish spawning, lower (~17 °C, L), and higher (~23 °C, H), to reveal the effects of altered water temperature on the DFO. Comparisons were made between the L and N (LvsN) conditions and H and N (HvsN) conditions. Transcriptomic analysis differentially expressed transcripts (DETs) related to heat stress were observed only in LvsN conditions, indicating that the DFO showed a stronger response to changes in LvsN than in HvsN conditions. Upregulation of DETs of vitellogenin receptors in N temperature showed that normal temperature was conducive to vitellogenin entry into the oocytes. Other temperature-sensitive DETs, including microtubule, kinesin, dynein, and actin, were closely associated with cell division and material transport. LvsN significantly impacted cell division and nutrient accumulation in the yolk, whereas HvsN only influenced cell division. Our results highlight the impact of altered water temperature on the DFO, thereby providing insights for future reservoir operations regarding river damming and climate change and establishing fish conservation measures.
Collapse
Affiliation(s)
- Ting Li
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- College of Water Resource and Hydropower, Sichuan University, Chengdu 610065, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- Changjiang River Scientific Research Institute, Wuhan 430010, China
| | - Qiuwen Chen
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
| | - Qi Zhang
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- College of Water Resource and Hydropower, Sichuan University, Chengdu 610065, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
| | - Tao Feng
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
| | - Jianyun Zhang
- Yangtze Institute for Conservation and Green Development, Nanjing 210029, China
| | - Yuqing Lin
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
| | - Peisi Yang
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
| | - Shufeng He
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
| | - Hui Zhang
- State Key Laboratory of Hydrology-Water Resources and Hydraulic Engineering, Nanjing Hydraulic Research Institute, Nanjing 210029, China
- Center for Eco-Environmental Research, Nanjing Hydraulic Research Institute, Nanjing 210029, China
| |
Collapse
|
89
|
Ramirez-Rios S, Choi SR, Sanyal C, Blum TB, Bosc C, Krichen F, Denarier E, Soleilhac JM, Blot B, Janke C, Stoppin-Mellet V, Magiera MM, Arnal I, Steinmetz MO, Moutin MJ. VASH1-SVBP and VASH2-SVBP generate different detyrosination profiles on microtubules. J Cell Biol 2022; 222:213744. [PMID: 36512346 PMCID: PMC9750192 DOI: 10.1083/jcb.202205096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022] Open
Abstract
The detyrosination/tyrosination cycle of α-tubulin is critical for proper cell functioning. VASH1-SVBP and VASH2-SVBP are ubiquitous enzymes involved in microtubule detyrosination, whose mode of action is little known. Here, we show in reconstituted systems and cells that VASH1-SVBP and VASH2-SVBP drive the global and local detyrosination of microtubules, respectively. We solved the cryo-electron microscopy structure of VASH2-SVBP bound to microtubules, revealing a different microtubule-binding configuration of its central catalytic region compared to VASH1-SVBP. We show that the divergent mode of detyrosination between the two enzymes is correlated with the microtubule-binding properties of their disordered N- and C-terminal regions. Specifically, the N-terminal region is responsible for a significantly longer residence time of VASH2-SVBP on microtubules compared to VASH1-SVBP. We suggest that this VASH region is critical for microtubule detachment and diffusion of VASH-SVBP enzymes on lattices. Our results suggest a mechanism by which VASH1-SVBP and VASH2-SVBP could generate distinct microtubule subpopulations and confined areas of detyrosinated lattices to drive various microtubule-based cellular functions.
Collapse
Affiliation(s)
- Sacnicte Ramirez-Rios
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Sung Ryul Choi
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Chadni Sanyal
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Thorsten B. Blum
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| | - Christophe Bosc
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Fatma Krichen
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Eric Denarier
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Jean-Marc Soleilhac
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Béatrice Blot
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Carsten Janke
- Institut Curie, Université Paris Sciences et Lettres, Centre National de la Recherche Scientifique UMR3348, Orsay, France,Université Paris-Saclay, Centre National de la Recherche Scientifique UMR3348, Orsay, France
| | - Virginie Stoppin-Mellet
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Maria M. Magiera
- Institut Curie, Université Paris Sciences et Lettres, Centre National de la Recherche Scientifique UMR3348, Orsay, France,Université Paris-Saclay, Centre National de la Recherche Scientifique UMR3348, Orsay, France
| | - Isabelle Arnal
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France
| | - Michel O. Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland,Biozentrum, University of Basel, Basel, Switzerland
| | - Marie-Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Grenoble Institut Neurosciences, Grenoble, France,Correspondence to Marie-Jo Moutin:
| |
Collapse
|
90
|
Ciliate Microtubule Diversities: Insights from the EFBTU3 Tubulin in the Antarctic Ciliate Euplotes focardii. Microorganisms 2022; 10:microorganisms10122415. [PMID: 36557668 PMCID: PMC9784925 DOI: 10.3390/microorganisms10122415] [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: 11/02/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Protozoans of the Phylum Ciliophora (ciliates) assemble many diverse microtubular structures in a single cell throughout the life cycle, a feature that made them useful models to study microtubule complexity and the role of tubulin isotypes. In the Antarctic ciliate Euplotes focardii we identified five β-tubulin isotypes by genome sequencing, named EFBTU1, EFBTU2, EFBTU3, EFBTU4 and EFBTU5. By using polyclonal antibodies directed against EFBTU2/EFBTU1 and EFBTU3, we show that the former isotypes appear to be involved in the formation of all microtubular structures and are particularly abundant in cilia, whereas the latter specifically localizes at the bases of cilia. By RNA interference (RNAi) technology, we silenced the EFBTU3 gene and provided evidence that this isotype has a relevant role in cilia regeneration upon deciliation and in cell division. These results support the long-standing concept that tubulin isotypes possess functional specificity in building diverse microtubular structures.
Collapse
|
91
|
Zhao S, Ji W, Shen Y, Fan Y, Huang H, Huang J, Lai G, Yuan K, Cheng C. Expression of hub genes of endothelial cells in glioblastoma-A prognostic model for GBM patients integrating single-cell RNA sequencing and bulk RNA sequencing. BMC Cancer 2022; 22:1274. [PMID: 36474171 PMCID: PMC9724299 DOI: 10.1186/s12885-022-10305-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND This study aimed to use single-cell RNA-seq (scRNA-seq) to discover marker genes in endothelial cells (ECs) and construct a prognostic model for glioblastoma multiforme (GBM) patients in combination with traditional high-throughput RNA sequencing (bulk RNA-seq). METHODS Bulk RNA-seq data was downloaded from The Cancer Genome Atlas (TCGA) and The China Glioma Genome Atlas (CGGA) databases. 10x scRNA-seq data for GBM were obtained from the Gene Expression Omnibus (GEO) database. The uniform manifold approximation and projection (UMAP) were used for downscaling and cluster identification. Key modules and differentially expressed genes (DEGs) were identified by weighted gene correlation network analysis (WGCNA). A non-negative matrix decomposition (NMF) algorithm was used to identify the different subtypes based on DEGs, and multivariate cox regression analysis to model the prognosis. Finally, differences in mutational landscape, immune cell abundance, immune checkpoint inhibitors (ICIs)-associated genes, immunotherapy effects, and enriched pathways were investigated between different risk groups. RESULTS The analysis of scRNA-seq data from eight samples revealed 13 clusters and four cell types. After applying Fisher's exact test, ECs were identified as the most important cell type. The NMF algorithm identified two clusters with different prognostic and immunological features based on DEGs. We finally built a prognostic model based on the expression levels of four key genes. Higher risk scores were significantly associated with poorer survival outcomes, low mutation rates in IDH genes, and upregulation of immune checkpoints such as PD-L1 and CD276. CONCLUSION We built and validated a 4-gene signature for GBM using 10 scRNA-seq and bulk RNA-seq data in this work.
Collapse
Affiliation(s)
- Songyun Zhao
- grid.460176.20000 0004 1775 8598Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023 Wuxi, Jiangsu China
| | - Wei Ji
- grid.460176.20000 0004 1775 8598Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023 Wuxi, Jiangsu China
| | - Yifan Shen
- grid.460176.20000 0004 1775 8598Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023 Wuxi, Jiangsu China
| | - Yuansheng Fan
- grid.460176.20000 0004 1775 8598Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023 Wuxi, Jiangsu China
| | - Hui Huang
- grid.460176.20000 0004 1775 8598Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023 Wuxi, Jiangsu China
| | - Jin Huang
- grid.460176.20000 0004 1775 8598Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023 Wuxi, Jiangsu China
| | - Guichuan Lai
- grid.203458.80000 0000 8653 0555Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, 400016 Chongqing, China
| | - Kemiao Yuan
- Department of Oncology, Traditional Chinese Medicine Hospital of Wuxi, No.8, West Zhongnan Road, 214071 Wuxi, China
| | - Chao Cheng
- grid.460176.20000 0004 1775 8598Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, 214023 Wuxi, Jiangsu China
| |
Collapse
|
92
|
Miryala CSJ, Holland ED, Dent EW. Contributions of microtubule dynamics and transport to presynaptic and postsynaptic functions. Mol Cell Neurosci 2022; 123:103787. [PMID: 36252720 PMCID: PMC9838116 DOI: 10.1016/j.mcn.2022.103787] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
Microtubules (MT) are elongated, tubular, cytoskeletal structures formed from polymerization of tubulin dimers. They undergo continuous cycles of polymerization and depolymerization, primarily at their plus ends, termed dynamic instability. Although this is an intrinsic property of MTs, there are a myriad of MT-associated proteins that function in regulating MT dynamic instability and other dynamic processes that shape the MT array. Additionally, MTs assemble into long, semi-rigid structures which act as substrates for long-range, motor-driven transport of many different types of cargoes throughout the cell. Both MT dynamics and motor-based transport play important roles in the function of every known type of cell. Within the last fifteen years many groups have shown that MT dynamics and transport play ever-increasing roles in the neuronal function of mature neurons. Not only are neurons highly polarized cells, but they also connect with one another through synapses to form complex networks. Here we will focus on exciting studies that have illuminated how MTs function both pre-synaptically in axonal boutons and post-synaptically in dendritic spines. It is becoming clear that MT dynamics and transport both serve important functions in synaptic plasticity. Thus, it is not surprising that disruption of MTs, either through hyperstabilization or destabilization, has profound consequences for learning and memory. Together, the studies described here suggest that MT dynamics and transport play key roles in synaptic function and when disrupted result in compromised learning and memory.
Collapse
Affiliation(s)
- Chandra S. J. Miryala
- Department of Neuroscience, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705
| | - Elizabeth D. Holland
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705
| | - Erik W. Dent
- Department of Neuroscience, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705,Corresponding Author: Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705,
| |
Collapse
|
93
|
O'Hagan R, Avrutis A, Ramicevic E. Functions of the tubulin code in the C. elegans nervous system. Mol Cell Neurosci 2022; 123:103790. [PMID: 36368428 DOI: 10.1016/j.mcn.2022.103790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Due to their elongated and polarized morphology, neurons rely on the microtubule (MT) cytoskeleton for their shape, as well as for efficient intracellular transport that maintains neuronal function, survival, and connectivity. Although all MTs are constructed from α- and β-tubulins that are highly conserved throughout eukaryotes, different MT networks within neurons exhibit different dynamics and functions. For example, molecular motors must be able to differentially recognize the axonal and dendritic MTs to deliver appropriate cargos to sensory endings and synaptic regions. The Tubulin Code hypothesis proposes that MTs can be specialized in form and function by chemical differences in their composition by inclusion of different α- and β-tubulins into the MT lattice, as well as differences in post-translational enzymatic modifications. The chemical differences encode information that allow MTs to regulate interactions with various microtubule-based molecular motors such as kinesins and dyneins as well as with structural microtubule-associated proteins (MAPs), which can, in turn, modify the function or stability of MTs. Here, we review studies involving C. elegans, a model organism with a relatively simple nervous system that is amenable to genetic analysis, that have contributed to our understanding of how the Tubulin Code can specialize neuronal MT networks to establish differences in neuronal morphology and function. Such studies have revealed molecules and mechanisms that are conserved in vertebrates and have the potential to inform our understanding of neurological diseases involving defects in the cytoskeleton and intracellular transport.
Collapse
Affiliation(s)
- Robert O'Hagan
- formerly at Biology Dept., Montclair State University, Montclair, NJ 07043, United States of America.
| | - Alexandra Avrutis
- formerly at Biology Dept., Montclair State University, Montclair, NJ 07043, United States of America
| | - Ema Ramicevic
- formerly at Biology Dept., Montclair State University, Montclair, NJ 07043, United States of America
| |
Collapse
|
94
|
Kletter T, Biswas A, Reber S. Engineering metaphase spindles: Construction site and building blocks. Curr Opin Cell Biol 2022; 79:102143. [PMID: 36436307 DOI: 10.1016/j.ceb.2022.102143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/08/2022] [Accepted: 10/21/2022] [Indexed: 11/27/2022]
Abstract
In an active, crowded cytoplasm, eukaryotic cells construct metaphase spindles from conserved building blocks to segregate chromosomes. Yet, spindles execute their function in a stunning variety of cell shapes and sizes across orders of magnitude. Thus, the current challenge is to understand how unique mesoscale spindle characteristics emerge from the interaction of molecular collectives. Key components of these collectives are tubulin dimers, which polymerise into microtubules. Despite all conservation, tubulin is a genetically and biochemically complex protein family, and we only begin to uncover how tubulin diversity affects microtubule dynamics and thus spindle assembly. Moreover, it is increasingly appreciated that spindles are dynamically intertwined with the cytoplasm that itself exhibits cell-type specific emergent properties with yet mostly unexplored consequences for spindle construction. Therefore, on our way toward a quantitative picture of spindle function, we need to understand molecular behaviour of the building blocks and connect it to the entire cellular context.
Collapse
Affiliation(s)
- Tobias Kletter
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10115 Berlin, Germany
| | - Abin Biswas
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10115 Berlin, Germany; Max-Planck-Institute for the Science of Light, 91058 Erlangen, Germany
| | - Simone Reber
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10115 Berlin, Germany; University of Applied Sciences Berlin, 13353 Berlin, Germany.
| |
Collapse
|
95
|
Wu YT, Zhang Z, Ji R, Zhang SH, Wang WP, Wu C, Zhang JP, Jiang XP, Zhang H. [Regulatory effects of bio-intensity electric field on microtubule acetylation in human epidermal cell line HaCaT]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2022; 38:1066-1072. [PMID: 36418264 DOI: 10.3760/cma.j.cn501120-20211105-00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Objective: To investigate the regulatory effects of bio-intensity electric field on directional migration and microtubule acetylation in human epidermal cell line HaCaT, aiming to provide molecular theoretical basis for the clinical treatment of wound repair. Methods: The experimental research methods were used. HaCaT cells were collected and divided into simulated electric field group (n=54) placed in the electric field device without electricity for 3 h and electric field treatment group (n=52) treated with 200 mV/mm electric field for 3 h (the same treatment methods below). The cell movement direction was observed in the living cell workstation and the movement velocity, trajectory velocity, and direction of cosθ of cell movement within 3 h of treatment were calculated. HaCaT cells were divided into simulated electric field group and electric field treatment 1 h group, electric field treatment 2 h group, and electric field treatment 3 h group which were treated with 200 mV/mm electric field for corresponding time. HaCaT cells were divided into simulated electric field group and 100 mV/mm electric field group, 200 mV/mm electric field group, and 300 mV/mm electric field group treated with electric field of corresponding intensities for 3 h. The protein expression of acetylated α-tubulin was detected by Western blotting (n=3). HaCaT cells were divided into simulated electric field group and electric field treatment group, and the protein expression of acetylated α-tubulin was detected and located by immunofluorescence method (n=3). Data were statistically analyzed with Kruskal-Wallis H test,Mann-Whitney U test, Bonferroni correction, one-way analysis of variance, least significant difference test, and independent sample t test. Results: Within 3 h of treatment, compared with that in simulated electric field group, the cells in electric field treatment group had obvious tendency to move directionally, the movement velocity and trajectory velocity were increased significantly (with Z values of -8.53 and -2.05, respectively, P<0.05 or P<0.01), and the directionality was significantly enhanced (Z=-8.65, P<0.01). Compared with (0.80±0.14) in simulated electric field group, the protein expressions of acetylated α-tubulin in electric field treatment 1 h group (1.50±0.08) and electric field treatment 2 h group (1.89±0.06) were not changed obviously (P>0.05), while the protein expression of acetylated α-tubulin of cells in electric field treatment 3 h group (3.37±0.36) was increased significantly (Z=-3.06, P<0.05). After treatment for 3 h, the protein expressions of acetylated α-tubulin of cells in 100 mV/mm electric field group, 200 mV/mm electric field group, and 300 mV/mm electric field group were 1.63±0.05, 2.24±0.08, and 2.00±0.13, respectively, which were significantly more than 0.95±0.27 in simulated electric field group (P<0.01). Compared with that in 100 mV/mm electric field group, the protein expressions of acetylated α-tubulin in 200 mV/mm electric field group and 300 mV/mm electric field group were increased significantly (P<0.01); the protein expression of acetylated α-tubulin of cells in 300 mV/mm electric field group was significantly lower than that in 200 mV/mm electric field group (P<0.05). After treatment for 3 h, compared with that in simulated electric field group, the acetylated α-tubulin of cells had enhanced directional distribution and higher protein expression (t=5.78, P<0.01). Conclusions: Bio-intensity electric field can induce the directional migration of HaCaT cells and obviously up-regulate the level of α-ubulin acetylation after treatment at 200 mV/mm bio-intensity electric field for 3 h.
Collapse
Affiliation(s)
- Y T Wu
- Department of Plastic and Burn Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Z Zhang
- Department of Plastic Surgery, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - R Ji
- Department of Plastic and Burn Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - S H Zhang
- Department of Plastic and Burn Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - W P Wang
- Department of Plastic and Burn Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - C Wu
- Department of Plastic Surgery, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - J P Zhang
- Department of Plastic Surgery, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - X P Jiang
- Department of Plastic Surgery, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - Hengshu Zhang
- Department of Plastic and Burn Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
96
|
Szczesna E, Zehr EA, Cummings SW, Szyk A, Mahalingan KK, Li Y, Roll-Mecak A. Combinatorial and antagonistic effects of tubulin glutamylation and glycylation on katanin microtubule severing. Dev Cell 2022; 57:2497-2513.e6. [PMID: 36347241 PMCID: PMC9665884 DOI: 10.1016/j.devcel.2022.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/17/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
Microtubules have spatiotemporally complex posttranslational modification patterns. How cells interpret this tubulin modification code is largely unknown. We show that C. elegans katanin, a microtubule severing AAA ATPase mutated in microcephaly and critical for cell division, axonal elongation, and cilia biogenesis, responds precisely, differentially, and combinatorially to three chemically distinct tubulin modifications-glycylation, glutamylation, and tyrosination-but is insensitive to acetylation. Glutamylation and glycylation are antagonistic rheostats with glycylation protecting microtubules from severing. Katanin exhibits graded and divergent responses to glutamylation on the α- and β-tubulin tails, and these act combinatorially. The katanin hexamer central pore constrains the polyglutamate chain patterns on β-tails recognized productively. Elements distal to the katanin AAA core sense α-tubulin tyrosination, and detyrosination downregulates severing. The multivalent microtubule recognition that enables katanin to read multiple tubulin modification inputs explains in vivo observations and illustrates how effectors can integrate tubulin code signals to produce diverse functional outcomes.
Collapse
Affiliation(s)
- Ewa Szczesna
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Elena A Zehr
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Steven W Cummings
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Agnieszka Szyk
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Kishore K Mahalingan
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Yan Li
- Proteomic Core Facility, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA; Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
97
|
Tang Q, Li X, Wang J. Tubulin deacetylase NDST3 modulates lysosomal acidification: Implications in neurological diseases. Bioessays 2022; 44:e2200110. [PMID: 36135988 PMCID: PMC9829454 DOI: 10.1002/bies.202200110] [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: 06/08/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 01/12/2023]
Abstract
Neurological diseases (NDs), featured by progressive dysfunctions of the nervous system, have become a growing burden for the aging populations. N-Deacetylase and N-sulfotransferase 3 (NDST3) is known to catalyze deacetylation and N-sulfation on disaccharide substrates. Recently, NDST3 is identified as a novel deacetylase for tubulin, and its newly recognized role in modulating microtubule acetylation and lysosomal acidification provides fresh insights into ND therapeutic approaches using NDST3 as a target. Microtubule acetylation and lysosomal acidification have been reported to be critical for activities in neurons, implying that the regulators of these two biological processes, such as the previously known microtubule deacetylases, histone deacetylase 6 (HDAC6) and sirtuin 2 (SIRT2), could play important roles in various NDs. Aberrant NDST3 expression or tubulin acetylation has been observed in an increasing number of NDs, including amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), schizophrenia and bipolar disorder, Alzheimer's disease (AD), and Parkinson's disease (PD), suggesting that NDST3 is a key player in the pathogenesis of NDs and may serve as a target for development of new treatment of NDs.
Collapse
Affiliation(s)
- Qing Tang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiangning Li
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jiou Wang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
98
|
Comprehensive Multiomic Analysis Identified TUBA1C as a Potential Prognostic Biological Marker of Immune-Related Therapy in Pan-Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9493115. [DOI: 10.1155/2022/9493115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/09/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
Abstract
TUBA1C is correlated with an unfavourable prognosis and the infiltration of immune cells in several cancers. However, its function as a significant biomarker for the prognosis of immunotherapy in pan-cancer remains unclear. This study aims at assessing the role of TUBA1C in pan-cancer at multiple levels, including mutations, gene expression, methylation, m6A methylation, and immune cell infiltration levels. Data retrieved from major public databases, such as TCGA, GEO, GTEx, GSCA, CancerSEA, HPA, and RNAactDrugs, revealed that TUBA1C expression was high in 33 cancer types. Survival analysis revealed that TUBA1C was a poor prognostic factor for 12 tumour types, and mutations, CNVs, and methylation affected the prognosis of some cancer types. Furthermore, TUBA1C was found to be related to immune-related genes, immune cell infiltration, and the immune microenvironment. In addition, the sensitivity of 10 anticancer drugs was associated with high TUBA1C expression. Therefore, TUBA1C may serve as a viable prognostic biomarker for immunotherapy of pan-cancer.
Collapse
|
99
|
Embryonic Development and Histological Analysis of Skeletal Muscles of Tenuidactylus caspius (Eichwald, 1831) Lizards (Reptilia: Squamata). J ZOOL SYST EVOL RES 2022. [DOI: 10.1155/2022/3618288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
During embryonic development of the Caspian thin-toed gecko migration, formation of myoblast and myosatellite cells occurs in the cranial-distal direction. Somite formation begins in the body part close to the skull and ends in the tail. The time of separation of somites from the proximal mesoderm depends on the temperature of the air and the substrate. Myoblast cells reach their targets and are connected, and the membranes in the area of their contact are destroyed. Myoblast’s fusion creates myosymplasts. The intermediate stage is observed after the formation of small myosymplasts. After that, the chain shape of myosymplasts are transformed into an intermediate plaque form. At this intermediate stage, the number of a nucleus is small, the shape of the nucleus differs from each other, and the location of the nucleus varies. Afterward, the connection of the intermediate forms with each other and with myoblasts forms a rounded shape, where the initial development of myotubules takes place. A fully formed myotubular and myosatellite cells are surrounded by a basal membrane and shape a muscle fiber. The skeletal muscles of the adult Caspian thin-toed gecko are mainly composed of white fibers. Thus, it allows the gecko to move very fast in a short time. Due to the small number of mitochondria in the myotubes, oxygen gas demand is decreased and the body is prevented from overheating.
Collapse
|
100
|
Hotta T, McAlear TS, Yue Y, Higaki T, Haynes SE, Nesvizhskii AI, Sept D, Verhey KJ, Bechstedt S, Ohi R. EML2-S constitutes a new class of proteins that recognizes and regulates the dynamics of tyrosinated microtubules. Curr Biol 2022; 32:3898-3910.e14. [PMID: 35963242 PMCID: PMC9530018 DOI: 10.1016/j.cub.2022.07.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/13/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023]
Abstract
Tubulin post-translational modifications (PTMs) alter microtubule properties by affecting the binding of microtubule-associated proteins (MAPs). Microtubule detyrosination, which occurs by proteolytic removal of the C-terminal tyrosine from ɑ-tubulin, generates the oldest known tubulin PTM, but we lack comprehensive knowledge of MAPs that are regulated by this PTM. We developed a screening pipeline to identify proteins that discriminate between Y- and ΔY-microtubules and found that echinoderm microtubule-associated protein-like 2 (EML2) preferentially interacts with Y-microtubules. This activity depends on a Y-microtubule interaction motif built from WD40 repeats. We show that EML2 tracks the tips of shortening microtubules, a behavior not previously seen among human MAPs in vivo, and influences dynamics to increase microtubule stability. Our screening pipeline is readily adapted to identify proteins that specifically recognize a wide range of microtubule PTMs.
Collapse
Affiliation(s)
- Takashi Hotta
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Thomas S McAlear
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Yang Yue
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Takumi Higaki
- Faculty of Advanced Science and Technology (FAST), Kumamoto University, Kumamoto, Japan; International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, Kumamoto, Japan
| | - Sarah E Haynes
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - David Sept
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kristen J Verhey
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Susanne Bechstedt
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Ryoma Ohi
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|