1
|
Cavaco J, Carvalhal S. Non-canonical roles of mitotic proteins in cortical neurons. Trends Neurosci 2025:S0166-2236(25)00105-5. [PMID: 40517024 DOI: 10.1016/j.tins.2025.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 05/02/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025]
Abstract
Mitotic proteins are traditionally studied for their role in chromosome segregation during cell division. However, research increasingly highlights the important non-canonical roles of mitotic proteins beyond mitosis, particularly in the mammalian cerebral cortex. Alterations in the expression levels or mutations of mitotic proteins are increasingly linked to brain disorders such as primary microcephaly and Alzheimer's disease. A central, unresolved question remains: how do mitotic proteins contribute to neuronal pathogenesis? Here, we review emerging literature on the non-canonical roles of mitotic proteins in mature neurons. Additionally, we discuss how these contribute to the complex mechanisms underlying neurodevelopmental and neurodegenerative disorders. We also discuss their potential for identifying therapeutic strategies and as biomarkers in brain pathologies.
Collapse
Affiliation(s)
- Joana Cavaco
- Algarve Biomedical Centre - Research Institute (ABC-Ri), Universidade do Algarve, Campus de Gambelas, Faro 8005-139, Portugal; Algarve Biomedical Centre (ABC), Universidade do Algarve, Campus de Gambelas, Faro 8005-139, Portugal
| | - Sara Carvalhal
- Algarve Biomedical Centre - Research Institute (ABC-Ri), Universidade do Algarve, Campus de Gambelas, Faro 8005-139, Portugal; Algarve Biomedical Centre (ABC), Universidade do Algarve, Campus de Gambelas, Faro 8005-139, Portugal.
| |
Collapse
|
2
|
Watson D, Mentch F, Billings J, Ostberg K, March ME, Kalish JM, Li D, Cannon I, Guay-Woodford LM, Hartung E, Strong A. Elucidating the Molecular Landscape of Cystic Kidney Disease: Old Friends, New Friends and Some Surprises. Am J Med Genet A 2025; 197:e64011. [PMID: 39888183 DOI: 10.1002/ajmg.a.64011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Cystic kidney diseases (CyKD) are a diverse group of disorders affecting more than 1 in 1000 individuals. Over 120 genes are implicated, primarily encoding components of the primary cilium, transcription factors, and morphogens. Prognosis varies greatly by molecular diagnosis. Causal variants are not identified in 10%-60% of individuals due to our limited understanding of CyKD. To elucidate the molecular landscape of CyKD, we queried the CAG Biobank using the ICD10 codes N28.1, Q61.1, Q61.11, Q61.19, Q61.2, Q61.3, and Q61.8 to identify individuals with CyKD. One hundred eight individuals met clinical criteria for CyKD and underwent proband-only exome sequencing. Causal variants were identified in 86/108 (80%) individuals. The most common molecular diagnoses were PKD1-related autosomal dominant polycystic kidney disease (32/108; 30%) and autosomal recessive polycystic kidney disease (21/108; 19%). Other common molecular diagnoses were ciliopathy syndromes (7/108; 6.5%) and Tuberous Sclerosis (6/108; 5.6%). Seven individuals had variants in genes not previously associated with CyKD (7/108; 6.5%). Candidate genes were identified in five individuals (5/108; 4.5%). Discordance between molecular and clinical diagnosis was present in two individuals. We demonstrate a high molecular diagnosis rate in individuals with CyKD that can result in diagnostic reclassification, supporting a role for genetic testing in CyKD.
Collapse
Affiliation(s)
- Deborah Watson
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Frank Mentch
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan Billings
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kayleigh Ostberg
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michael E March
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jennifer M Kalish
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - India Cannon
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lisa M Guay-Woodford
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Nephrology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Erum Hartung
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Nephrology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Alanna Strong
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Chen L, He C, Ou Z, Zhao C. TNF-α drives bladder cancer metastasis via METTL3-mediated m6A modification to promote CLASP2/IQGAP1-dependent cytoskeleton remodeling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167811. [PMID: 40118293 DOI: 10.1016/j.bbadis.2025.167811] [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: 05/31/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Bladder cancer (BCa) metastasis is a multi-step process triggered by cytoskeleton reorganization. However, the regulation mechanism of cytoskeleton reorganization in BCa remains ambiguous. This study elucidated the influence of tumor necrosis factor-alpha (TNF-α) in cytoskeleton remodeling during BCa metastasis and its possible mechanisms. METHODS Colony formation, scratch, transwell, and the nude mouse model were adopted to evaluate the growth and metastasis. Molecular expression was assessed by immunohistochemical staining, quantitative real-time PCR (qRT-PCR), and Western blotting. The N6-methyladenosine (m6A) level was detected by methylated RNA immunoprecipitation (MeRIP). Protein interaction was validated by Co-immunoprecipitation (Co-IP). Immunofluorescence staining was used to identify rearrangement of actin filament fibers (F-actin) and protein colocalization. RESULTS TNF-α facilitated cytoplasmic linker associated protein 2 (CLASP2) and methyltransferase like 3 (METTL3) expression in a dose (10-50 ng/mL)-dependent manner in BCa. CLASP2 high expression suggested a shorter overall survival of BCa patients. CLASP2 deficiency suppressed BCa cell proliferation, migration, and invasion via disrupting F-actin cytoskeleton. Mechanistically, TNF-α promoted METTL3-mediated m6A modification of CLASP2 to enhance CLASP2 mRNA stability. Moreover, CLASP2 directly interplayed with IQ motif containing GTPase activating protein 1 (IQGAP1) to regulate F-actin cytoskeleton remodeling. In vivo data demonstrated that inhibition of METTL3/CLASP2 axis delayed lung metastasis in nude mice. CONCLUSION TNF-α favors BCa cell metastasis, which involves METTL3-mediated m6A modification of CLASP2 that interacts with IQGAP1, thus leading to F-actin cytoskeleton remodeling. Our findings suggest targeting TNF-α/METTL3/CLASP2/IQGAP1 axis as a potential avenue for promising treatment for BCa.
Collapse
Affiliation(s)
- Lingxiao Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Cheng He
- Department of Urology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Zhenyu Ou
- Department of Urology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Cheng Zhao
- Department of Urology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
4
|
Wang R, Zhao C, Ou Z, Chen L. KHSRP promotes the malignant behavior and cisplatin resistance of bladder cancer cells through the CLASP2/MAPRE1 axis. THE PHARMACOGENOMICS JOURNAL 2025; 25:14. [PMID: 40382315 DOI: 10.1038/s41397-025-00374-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 04/20/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Bladder cancer (BC) is a highly prevalent form of cancer worldwide, and cisplatin (CDDP) resistance poses a major challenge to patients. Cytoplasmic linker-associated protein 2 (CLASP2) is a member of the microtubule plus-end tracking protein family and is involved in the regulation of microtubule dynamics. In this study, we evaluated the influence of CLASP2 on BC progression and cisplatin resistance. Levels of CLASP2, HNRNPA1, NONO, ZRANB2, FUS, KHSRP and QKI in BC tissues and cells were tested by RT-qPCR. Protein levels of CLASP2 and KHSRP were detected by Western blot. Cell viability and IC50 of cisplatin-treated BC cells were measured by CCK-8. Cell proliferation and apoptosis were determined using colony formation assay and flow cytometry, respectively. RNA immunoprecipitation (RIP) and Co-immunoprecipitation (Co-IP) experiments were adopted to verify target genes of CLASP2. Cellular localization of CLASP2 and MAPRE1 was detected utilizing immunofluorescence staining. The xenograft tumor model was established in BALB/c nude mice. We found that iCLASP2 levels were increased in CDDP-resistant BC tissues and cells. Suppression of CLASP2 impeded BC cell proliferation and alleviated their resistance to CDDP. KHSRP positively influenced the stability of CLASP2 mRNA. There was a protein interaction between CLASP2 and MAPRE1. Silencing KHSRP or MAPRE1 reversed the effect exerted of CLASP2 on BC cells. CLASP2 decreased the sensitivity of BC to CDDP in vivo. Our results imply that CLASP2 contributes to tumorigenesis and cisplatin resistance in BC via targeting MAPRE1, thereby promoting BC progression and providing a new therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Ruizhe Wang
- Department of Urology, Xiangya Hospital Central South University, Changsha City, Hunan Province, China
| | - Cheng Zhao
- Department of Urology, Xiangya Hospital Central South University, Changsha City, Hunan Province, China
| | - Zhenyu Ou
- Department of Urology, Xiangya Hospital Central South University, Changsha City, Hunan Province, China
| | - Lingxiao Chen
- Department of Urology, Xiangya Hospital Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
5
|
Wang Y, Liu X, Liu Z, Hua S, Jiang K. APC orchestrates microtubule dynamics by acting as a positive regulator of KIF2A and a negative regulator of CLASPs. CELL INSIGHT 2025; 4:100210. [PMID: 39640087 PMCID: PMC11617872 DOI: 10.1016/j.cellin.2024.100210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 12/07/2024]
Abstract
Tumor suppressor protein Adenomatous polyposis coli protein (APC) is an EB-binding and microtubule (MT) plus end-tracking protein; however, how exactly APC regulates MT dynamics remains elusive. Here, we show that in LLC-PK1 cells, APC and KIF2A, an MT depolymerase, form a complex clustering at the cell edge and destabilize MTs at the MT plus ends. Further biochemical characterization and mutational analysis reveal key residues for the APC-KIF2A interaction. In addition, APC counteracts the major MT-stabilizer CLASPs at MT plus ends and promotes directional cell migration via modulating cell adhesion force. Reconstitution experiments demonstrate that APC potentiates KIF2A-induced MT catastrophes and antagonizes the stabilizing effect of CLASP2 in vitro. In summary, APC functions as a positive regulator of MT-destabilizer and a negative regulator of MT-stabilizer to orchestrate MT dynamics.
Collapse
Affiliation(s)
- Yong Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Xinping Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Zheng Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Shasha Hua
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Kai Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
6
|
Aquino-Perez C, Safaralizade M, Podhajecky R, Wang H, Lansky Z, Grosse R, Macurek L. FAM110A promotes mitotic spindle formation by linking microtubules with actin cytoskeleton. Proc Natl Acad Sci U S A 2024; 121:e2321647121. [PMID: 38995965 PMCID: PMC11260166 DOI: 10.1073/pnas.2321647121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/06/2024] [Indexed: 07/14/2024] Open
Abstract
Precise segregation of chromosomes during mitosis requires assembly of a bipolar mitotic spindle followed by correct attachment of microtubules to the kinetochores. This highly spatiotemporally organized process is controlled by various mitotic kinases and molecular motors. We have recently shown that Casein Kinase 1 (CK1) promotes timely progression through mitosis by phosphorylating FAM110A leading to its enrichment at spindle poles. However, the mechanism by which FAM110A exerts its function in mitosis is unknown. Using structure prediction and a set of deletion mutants, we mapped here the interaction of the N- and C-terminal domains of FAM110A with actin and tubulin, respectively. Next, we found that the FAM110A-Δ40-61 mutant deficient in actin binding failed to rescue defects in chromosomal alignment caused by depletion of endogenous FAM110A. Depletion of FAM110A impaired assembly of F-actin in the proximity of spindle poles and was rescued by expression of the wild-type FAM110A, but not the FAM110A-Δ40-61 mutant. Purified FAM110A promoted binding of F-actin to microtubules as well as bundling of actin filaments in vitro. Finally, we found that the inhibition of CK1 impaired spindle actin formation and delayed progression through mitosis. We propose that CK1 and FAM110A promote timely progression through mitosis by mediating the interaction between spindle microtubules and filamentous actin to ensure proper mitotic spindle formation.
Collapse
Affiliation(s)
- Cecilia Aquino-Perez
- Cancer Cell Biology, Institute of Molecular Genetics, Czech Academy of Sciences, PragueCZ14220, Czech Republic
| | - Mahira Safaralizade
- Institute for Clinical and Experimental Pharmacology and Toxicology I, Medical Faculty, University of Freiburg, Freiburg79104, Germany
| | - Roman Podhajecky
- Institute of Biotechnology, Czech Academy of Sciences, Biocev, VestecCZ25250, Czech Republic
| | - Hong Wang
- Institute for Clinical and Experimental Pharmacology and Toxicology I, Medical Faculty, University of Freiburg, Freiburg79104, Germany
- Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg79104, Germany
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, Biocev, VestecCZ25250, Czech Republic
| | - Robert Grosse
- Institute for Clinical and Experimental Pharmacology and Toxicology I, Medical Faculty, University of Freiburg, Freiburg79104, Germany
- Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg79104, Germany
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics, Czech Academy of Sciences, PragueCZ14220, Czech Republic
| |
Collapse
|
7
|
Conboy JP, Istúriz Petitjean I, van der Net A, Koenderink GH. How cytoskeletal crosstalk makes cells move: Bridging cell-free and cell studies. BIOPHYSICS REVIEWS 2024; 5:021307. [PMID: 38840976 PMCID: PMC11151447 DOI: 10.1063/5.0198119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cell migration is a fundamental process for life and is highly dependent on the dynamical and mechanical properties of the cytoskeleton. Intensive physical and biochemical crosstalk among actin, microtubules, and intermediate filaments ensures their coordination to facilitate and enable migration. In this review, we discuss the different mechanical aspects that govern cell migration and provide, for each mechanical aspect, a novel perspective by juxtaposing two complementary approaches to the biophysical study of cytoskeletal crosstalk: live-cell studies (often referred to as top-down studies) and cell-free studies (often referred to as bottom-up studies). We summarize the main findings from both experimental approaches, and we provide our perspective on bridging the two perspectives to address the open questions of how cytoskeletal crosstalk governs cell migration and makes cells move.
Collapse
Affiliation(s)
- James P. Conboy
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Irene Istúriz Petitjean
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Anouk van der Net
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Gijsje H. Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| |
Collapse
|
8
|
Kumari R, Ven K, Chastney M, Kokate SB, Peränen J, Aaron J, Kogan K, Almeida-Souza L, Kremneva E, Poincloux R, Chew TL, Gunning PW, Ivaska J, Lappalainen P. Focal adhesions contain three specialized actin nanoscale layers. Nat Commun 2024; 15:2547. [PMID: 38514695 PMCID: PMC10957975 DOI: 10.1038/s41467-024-46868-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
Focal adhesions (FAs) connect inner workings of cell to the extracellular matrix to control cell adhesion, migration and mechanosensing. Previous studies demonstrated that FAs contain three vertical layers, which connect extracellular matrix to the cytoskeleton. By using super-resolution iPALM microscopy, we identify two additional nanoscale layers within FAs, specified by actin filaments bound to tropomyosin isoforms Tpm1.6 and Tpm3.2. The Tpm1.6-actin filaments, beneath the previously identified α-actinin cross-linked actin filaments, appear critical for adhesion maturation and controlled cell motility, whereas the adjacent Tpm3.2-actin filament layer beneath seems to facilitate adhesion disassembly. Mechanistically, Tpm3.2 stabilizes ACF-7/MACF1 and KANK-family proteins at adhesions, and hence targets microtubule plus-ends to FAs to catalyse their disassembly. Tpm3.2 depletion leads to disorganized microtubule network, abnormally stable FAs, and defects in tail retraction during migration. Thus, FAs are composed of distinct actin filament layers, and each may have specific roles in coupling adhesions to the cytoskeleton, or in controlling adhesion dynamics.
Collapse
Affiliation(s)
- Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Katharina Ven
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Megan Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Shrikant B Kokate
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Johan Peränen
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Jesse Aaron
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Leonardo Almeida-Souza
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Elena Kremneva
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland
| | - Renaud Poincloux
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Teng-Leong Chew
- Advanced Imaging Center, HHMI Janelia Research Campus, Ashburn, VA, 20147, USA
| | - Peter W Gunning
- School of Biomedical Sciences, UNSW Sydney, Wallace Wurth Building, Sydney, NSW 2052, Australia
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
- Department of Life Technologies, University of Turku, FI-20520, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Foundation for the Finnish Cancer Institute, Tukholmankatu 8, FI-00014, Helsinki, Finland
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
9
|
Sabo J, Dujava Zdimalova M, Slater PG, Dostal V, Herynek S, Libusova L, Lowery LA, Braun M, Lansky Z. CKAP5 enables formation of persistent actin bundles templated by dynamically instable microtubules. Curr Biol 2024; 34:260-272.e7. [PMID: 38086388 PMCID: PMC10841699 DOI: 10.1016/j.cub.2023.11.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/06/2023] [Accepted: 11/14/2023] [Indexed: 01/25/2024]
Abstract
Cytoskeletal rearrangements and crosstalk between microtubules and actin filaments are vital for living organisms. Recently, an abundantly present microtubule polymerase, CKAP5 (XMAP215 homolog), has been reported to play a role in mediating crosstalk between microtubules and actin filaments in the neuronal growth cones. However, the molecular mechanism of this process is unknown. Here, we demonstrate, in a reconstituted system, that CKAP5 enables the formation of persistent actin bundles templated by dynamically instable microtubules. We explain the templating by the difference in CKAP5 binding to microtubules and actin filaments. Binding to the microtubule lattice with higher affinity, CKAP5 enables the formation of actin bundles exclusively on the microtubule lattice, at CKAP5 concentrations insufficient to support any actin bundling in the absence of microtubules. Strikingly, when the microtubules depolymerize, actin bundles prevail at the positions predetermined by the microtubules. We propose that the local abundance of available CKAP5-binding sites in actin bundles allows the retention of CKAP5, resulting in persisting actin bundles. In line with our observations, we found that reducing CKAP5 levels in vivo results in a decrease in actin-microtubule co-localization in growth cones and specifically decreases actin intensity at microtubule plus ends. This readily suggests a mechanism explaining how exploratory microtubules set the positions of actin bundles, for example, in cytoskeleton-rich neuronal growth cones.
Collapse
Affiliation(s)
- Jan Sabo
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, Prague West, Prague 25250, Czech Republic; Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Hlavova 8, Prague 12800, Czech Republic
| | - Michaela Dujava Zdimalova
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, Prague West, Prague 25250, Czech Republic
| | - Paula G Slater
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencias, Universidad San Sebastián, Campus Los Leones, Lota 2465, Providencia, Santiago 7510602, Chile
| | - Vojtech Dostal
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 12800, Czech Republic
| | - Stepan Herynek
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, Prague West, Prague 25250, Czech Republic; Department of Genetics and Microbiology, Faculty of Science, Charles University, Vinicna 7, Prague 12800, Czech Republic
| | - Lenka Libusova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 12800, Czech Republic
| | - Laura A Lowery
- Department of Medicine, Section of Hematology/Oncology, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, Prague West, Prague 25250, Czech Republic.
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Prumyslova 595, Prague West, Prague 25250, Czech Republic.
| |
Collapse
|
10
|
Haque F, Subramanian R. Cytoskeleton crosstalk: Casting stable actin bundles with dynamic microtubule molds. Curr Biol 2024; 34:R72-R74. [PMID: 38262365 DOI: 10.1016/j.cub.2023.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Actin-microtubule crosstalk diversifies cytoskeletal networks. A new study provides insight into how the microtubule polymerase CKAP5 mediates actin-microtubule crosstalk. CKAP5 directs the assembly of stable actin bundles on dynamic microtubules; in turn, the actin bundles align growing microtubules along their length.
Collapse
Affiliation(s)
- Farah Haque
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Lawrence EJ, Chatterjee S, Zanic M. CLASPs stabilize the pre-catastrophe intermediate state between microtubule growth and shrinkage. J Cell Biol 2023; 222:e202107027. [PMID: 37184584 PMCID: PMC10195879 DOI: 10.1083/jcb.202107027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/03/2022] [Accepted: 04/18/2023] [Indexed: 05/16/2023] Open
Abstract
Cytoplasmic linker-associated proteins (CLASPs) regulate microtubules in fundamental cellular processes. CLASPs stabilize dynamic microtubules by suppressing microtubule catastrophe and promoting rescue, the switch-like transitions between growth and shrinkage. How CLASPs specifically modulate microtubule transitions is not understood. Here, we investigate the effects of CLASPs on the pre-catastrophe intermediate state of microtubule dynamics, employing distinct microtubule substrates to mimic the intermediate state. Surprisingly, we find that CLASP1 promotes the depolymerization of stabilized microtubules in the presence of GTP, but not in the absence of nucleotide. This activity is also observed for CLASP2 family members and a minimal TOG2-domain construct. Conversely, we find that CLASP1 stabilizes unstable microtubules upon tubulin dilution in the presence of GTP. Strikingly, our results reveal that CLASP1 drives microtubule substrates with vastly different inherent stabilities into the same slowly depolymerizing state in a nucleotide-dependent manner. We interpret this state as the pre-catastrophe intermediate state. Therefore, we conclude that CLASPs suppress microtubule catastrophe by stabilizing the intermediate state between growth and shrinkage.
Collapse
Affiliation(s)
- Elizabeth J. Lawrence
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Saptarshi Chatterjee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|