1
|
Liu F, Medyukhina A, Olesen KM, Shirinifard A, Jin H, Li L, Mapelli M, Khairy K, Han YG. Canonical Hedgehog Signaling Controls Astral Microtubules and Mitotic Spindle Orientation in Neural Progenitors and iPSCs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639780. [PMID: 40060449 PMCID: PMC11888381 DOI: 10.1101/2025.02.23.639780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Mitotic spindle orientation is crucial for cell fate determination and tissue organization. Although the intracellular machinery governing spindle orientation is well characterized, whether and how secreted factors, such as morphogens, regulate this process remains poorly understood. This study investigated the role of Hedgehog (HH) signaling in modulating mitotic spindle orientation in neural progenitor cells and in induced pluripotent stem cells (iPSCs). Time-lapse microscopy of cerebral organoids and iPSCs revealed that HH signaling increases the angle of the mitotic spindle relative to the apical surface, prolongs mitosis, and enhances spindle rotation. Mechanistically, HH signaling reduces both the number and the length of astral microtubules, key regulators of spindle orientation. This reduction correlates with increased spindle angle in iPSCs. Furthermore, we show that canonical HH signaling, involving GLI-dependent transcriptional regulation, contributes to these effects. RNA sequencing and gene set enrichment analysis (GSEA) revealed that HH signaling upregulates genes associated with microtubule depolymerization, suggesting a transcriptional mechanism by which HH signaling influences astral microtubule dynamics and, consequently, mitotic spindle orientation. These findings highlight a novel link between a morphogen, transcriptional regulation, and the control of mitotic spindle orientation, with implications for development and tissue homeostasis.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Anna Medyukhina
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Kris M Olesen
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Lei Li
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Marina Mapelli
- European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Khaled Khairy
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Young-Goo Han
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
2
|
Majumdar S, Liu ST. Spatiotemporal regulation of MELK during mitosis. Front Cell Dev Biol 2024; 12:1406940. [PMID: 39355119 PMCID: PMC11443572 DOI: 10.3389/fcell.2024.1406940] [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: 03/25/2024] [Accepted: 08/30/2024] [Indexed: 10/03/2024] Open
Abstract
Maternal Embryonic Leucine Zipper Kinase (MELK) has been studied intensively in recent years due to its overexpression in multiple cancers. However, the cell biology of MELK remains less characterized despite its well-documented association with mitosis. Here we report a distinctive pattern of human MELK that translocates from the cytoplasm to cell cortex within 3 min of anaphase onset. The cortex association lasts about 30 min till telophase. The spatiotemporal specific localization of MELK depends on the interaction between its Threonine-Proline (TP) rich domain and kinase associated 1 (KA1) domain, which is regulated by CDK1 kinase and PP4 protein phosphatase. KA1 domains are known to regulate kinase activities through various intramolecular interactions. Our results revealed a new role for KA1 domain to control subcellular localization of a protein kinase.
Collapse
Affiliation(s)
| | - Song-Tao Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
3
|
Peng J, Liu H, Liu Y, Liu J, Zhao Q, Liu W, Niu H, Xue H, Sun J, Wu J. HDAC6 mediates tumorigenesis during mitosis and the development of targeted deactivating agents. Bioorg Chem 2024; 153:107818. [PMID: 39288633 DOI: 10.1016/j.bioorg.2024.107818] [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: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/19/2024]
Abstract
Epigenetics, particularly deacetylation, plays a critical role in tumorigenesis as many carcinogens are under tight control by post-translational modification. HDAC6, an important and special histone deacetylase (HDAC) family member, has been indicated to increase carcinogenesis through various functions. Recent studies demonstrated the effects of HDAC6 inhibitors in mitotic arrest, however, detailed mechanisms still remain unknown. Herein, we review and summarize HDAC6-associated proteins that have been implicated in important roles in mitosis. We also discuss the development of medicinal agents targeting HDAC6.
Collapse
Affiliation(s)
- Jie Peng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Hongyan Liu
- The People's Hospital of Zhaoyuan City, No. 168 Yingbin Road, Zhaoyuan 265400, Shandong Province, PR China
| | - Yujing Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingqian Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Qianlong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Wenjia Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoqian Niu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoyu Xue
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jie Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingde Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
4
|
Rashid N, Juneja P, Rathi A, Sultan I, Rehman SU. Identification of Alternatively Spliced Novel Isoforms of Human HSPB8 Gene. Protein J 2024; 43:782-792. [PMID: 38980537 DOI: 10.1007/s10930-024-10215-y] [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] [Accepted: 06/10/2024] [Indexed: 07/10/2024]
Abstract
HSPB8 is a heat shock protein belonging to a family of ATP-independent stress proteins called HSPB which are present far and wide in the cells of various organisms. They are committed to protein quality control (PQC) and strive to avert protein aggregation and to procreate a pool of non-native proteins that can be swiftly folded. Their fundamental expression or stress inducibility is regulated by various cis-elements localized in the HSPB regulatory regions. In the current study we have predicted and confirmed two alternatively spliced novel transcripts of HSPB8 gene in liver, brain, and heart. These spliced variants have smaller sizes owing to smaller N terminal regions and showed remarkable changes in their cellular localization. Novel isoform (HSPB8-N1) was predicted to be majorly localized to nuclear region while the reported isoform (HSPB8) and one of the novel isoforms (HSPB8-N2) were predicted to be cytoplasmic in nature. There were many changes observed in the phosphorylation sites of the novel isoforms as well. The newly reported isoforms lack several structural motifs that are essential for various functional endeavors of the HSPB8 protein. In silico analysis of the conceptually translated protein was carried out using various bioinformatics tools to gain an understanding of their properties in order to explore their possible potential in therapeutics.
Collapse
Affiliation(s)
- Naira Rashid
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Pallavi Juneja
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Akshat Rathi
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Insha Sultan
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Sayeed Ur Rehman
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
5
|
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
|
6
|
Verlhac MH. Exploring the maternal inheritance transmitted by the oocyte to its progeny. C R Biol 2024; 347:45-52. [PMID: 38888193 DOI: 10.5802/crbiol.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Fertility is declining worldwide and many couples are turning towards assisted reproductive technologies (ART) to conceive babies. Organisms that propagate via sexual reproduction often come from the fusion between two gametes, an oocyte and a sperm, whose qualities seem to be decreasing in the human species. Interestingly, while the sperm mostly transmits its haploid genome, the oocyte transmits not only its haploid set of chromosomes but also its huge cytoplasm to its progeny. This is what can be defined as the maternal inheritance composed of chromosomes, organelles, lipids, metabolites, proteins and RNAs. To decipher the decline in oocyte quality, it is essential to explore the nature of the maternal inheritance, and therefore study the last stages of murine oogenesis, namely the end of oocyte growth followed by the two meiotic divisions. These divisions are extremely asymmetric in terms of the size of the daughter cells, allowing to preserve the maternal inheritance accumulated during oocyte growth within these huge cells to support early embryo development. Studies performed in Marie-Hélène Verlhac's lab have allowed to discover the unprecedented impact of original acto-myosin based mechanisms in the constitution as well as the preservation of this maternal inheritance and the consequences when these processes go awry.
Collapse
|
7
|
Miura T, Kado J, Ashisuke K, Masuzawa M, Nakayama F. Sustained activation of the FGF1-MEK-ERK pathway inhibits proliferation, invasion and migration and enhances radiosensitivity in mouse angiosarcoma cells. JOURNAL OF RADIATION RESEARCH 2024; 65:303-314. [PMID: 38637316 PMCID: PMC11115473 DOI: 10.1093/jrr/rrae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/15/2023] [Indexed: 04/20/2024]
Abstract
Angiosarcoma is a rare refractory soft-tissue tumor with a poor prognosis and is treated by radiotherapy. The fibroblast growth factor 1 (FGF1) mutant, with enhanced thermostability due to several substituted amino acids, inhibits angiosarcoma cell metastasis, yet the mechanism of action is unclear. This study aims to clarify the FGF1 mutant mechanism of action using ISOS-1 mouse angiosarcoma cells. The wild-type FGF1 or FGF1 mutant was added to ISOS-1 cells and cultured, evaluating cell numbers over time. The invasive and migratory capacity of ISOS-1 cells was assessed by transwell analysis. ISOS-1 cell radiosensitivity was assessed by colony formation assay after X-ray irradiation. To examine whether mitogen-activated protein kinase (MEK) inhibitor counteracts the FGF1 mutant effects, a combination of MEK inhibitor and FGF1 mutant was added to ISOS-1 cells and cultured. The FGF1 mutant was observed to inhibit ISOS-1 cell proliferation, invasion and migration by sustained FGF1 signaling activation. A MEK inhibitor suppressed the FGF1 mutant-induced inhibition of proliferation, invasion and migration of ISOS-1 cells. Furthermore, the FGF1 mutant enhanced radiosensitivity of ISOS-1 cells, but MEK inhibition suppressed the increased radiosensitivity. In addition, we found that the FGF1 mutant strongly inhibits actin polymerization, suggesting that actin cytoskeletal dynamics are closely related to ISOS-1 cell radiosensitivity. Overall, this study demonstrated that in ISOS-1 cells, the FGF1 mutant inhibits proliferation, invasion and migration while enhancing radiosensitivity through sustained activation of the MEK-mediated signaling pathway.
Collapse
Affiliation(s)
- Taichi Miura
- Regenerative Therapy Research Group, Department of Radiation Regulatory Science Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Junko Kado
- Regenerative Therapy Research Group, Department of Radiation Regulatory Science Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kazuma Ashisuke
- Radiation Effect Research Group, Department of Accelerator and Medical Physics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Mikio Masuzawa
- Department of Dermatology, Iwase General Hospital, 20 Kitamachi, Sukagawa-shi, Fukushima 962-8503, Japan
| | - Fumiaki Nakayama
- Regenerative Therapy Research Group, Department of Radiation Regulatory Science Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
8
|
Wang D, Wang Y, Di X, Wang F, Wanninayaka A, Carnell M, Hardeman EC, Jin D, Gunning PW. Cortical tension drug screen links mitotic spindle integrity to Rho pathway. Curr Biol 2023; 33:4458-4469.e4. [PMID: 37875071 DOI: 10.1016/j.cub.2023.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 07/24/2023] [Accepted: 09/11/2023] [Indexed: 10/26/2023]
Abstract
Mechanical force generation plays an essential role in many cellular functions, including mitosis. Actomyosin contractile forces mediate changes in cell shape in mitosis and are implicated in mitotic spindle integrity via cortical tension. An unbiased screen of 150 small molecules that impact actin organization and 32 anti-mitotic drugs identified two molecular targets, Rho kinase (ROCK) and tropomyosin 3.1/2 (Tpm3.1/2), whose inhibition has the greatest impact on mitotic cortical tension. The converse was found for compounds that depolymerize microtubules. Tpm3.1/2 forms a co-polymer with mitotic cortical actin filaments, and its inhibition prevents rescue of multipolar spindles induced by anti-microtubule chemotherapeutics. We examined the role of mitotic cortical tension in this rescue mechanism. Inhibition of ROCK and Tpm3.1/2 and knockdown (KD) of cortical nonmuscle myosin 2A (NM2A), all of which reduce cortical tension, inhibited rescue of multipolar mitotic spindles, further implicating cortical tension in the rescue mechanism. GEF-H1 released from microtubules by depolymerization increased cortical tension through the RhoA pathway, and its KD also inhibited rescue of multipolar mitotic spindles. We conclude that microtubule depolymerization by anti-cancer drugs induces cortical-tension-based rescue to ensure integrity of the mitotic bipolar spindle mediated via the RhoA pathway. Central to this mechanism is the dependence of NM2A on Tpm3.1/2 to produce the functional engagement of actin filaments responsible for cortical tension.
Collapse
Affiliation(s)
- Dejiang Wang
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Yao Wang
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Xiangjun Di
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Fan Wang
- School of Electrical and Data Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW 2007, Australia; School of Physics, Beihang University, Beijing 100191, P.R. China
| | - Amanda Wanninayaka
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Michael Carnell
- Katharina Gaus Light Microscope Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Edna C Hardeman
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Dayong Jin
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; UTS-SUStech Joint Research Centre for Biomedical Materials & Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, P.R. China
| | - Peter W Gunning
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
9
|
Quadri R, Rotondo G, Sertic S, Pozzi S, dell’Oca MC, Guerrini L, Muzi-Falconi M. A Haspin-ARHGAP11A axis regulates epithelial morphogenesis through Rho-ROCK dependent modulation of LIMK1-Cofilin. iScience 2023; 26:108011. [PMID: 37841592 PMCID: PMC10570125 DOI: 10.1016/j.isci.2023.108011] [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: 05/02/2023] [Revised: 07/20/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Throughout mitosis, a plethora of processes must be efficiently concerted to ensure cell proliferation and tissue functionality. The mitotic spindle does not only mediate chromosome segregation, but also defines the axis of cellular division, thus determining tissue morphology. Functional spindle orientation relies on precise actin dynamics, shaped in mitosis by the LIMK1-Cofilin axis. The kinase Haspin acts as a guardian of faithful chromosome segregation that ensures amphitelic chromosome attachment and prevents unscheduled cohesin cleavage. Here, we report an unprecedented role for Haspin in the determination of spindle orientation in mitosis. We show that, during mitosis, Haspin regulates Rho-ROCK activity through ARHGAP11A, a poorly characterized GAP, and that ROCK is in turn responsible for the mitotic activation of LIMK1 and stabilization of the actin cytoskeleton, thus supporting a functional spindle orientation. By exploiting 3D cell cultures, we show that this pathway is pivotal for the establishment of a morphologically functional tissue.
Collapse
Affiliation(s)
- Roberto Quadri
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Giuseppe Rotondo
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Sarah Sertic
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Sara Pozzi
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | | | - Luisa Guerrini
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Marco Muzi-Falconi
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| |
Collapse
|
10
|
Yabushita T, Chinen T, Nishiyama A, Asada S, Shimura R, Isobe T, Yamamoto K, Sato N, Enomoto Y, Tanaka Y, Fukuyama T, Satoh H, Kato K, Saitoh K, Ishikawa T, Soga T, Nannya Y, Fukagawa T, Nakanishi M, Kitagawa D, Kitamura T, Goyama S. Mitotic perturbation is a key mechanism of action of decitabine in myeloid tumor treatment. Cell Rep 2023; 42:113098. [PMID: 37714156 DOI: 10.1016/j.celrep.2023.113098] [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: 12/20/2022] [Revised: 06/22/2023] [Accepted: 08/21/2023] [Indexed: 09/17/2023] Open
Abstract
Decitabine (DAC) is clinically used to treat myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Our genome-wide CRISPR-dCas9 activation screen using MDS-derived AML cells indicates that mitotic regulation is critical for DAC resistance. DAC strongly induces abnormal mitosis (abscission failure or tripolar mitosis) in human myeloid tumors at clinical concentrations, especially in those with TP53 mutations or antecedent hematological disorders. This DAC-induced mitotic disruption and apoptosis are significantly attenuated in DNMT1-depleted cells. In contrast, overexpression of Dnmt1, but not the catalytically inactive mutant, enhances DAC-induced mitotic defects in myeloid tumors. We also demonstrate that DAC-induced mitotic disruption is enhanced by pharmacological inhibition of the ATR-CLSPN-CHK1 pathway. These data challenge the current assumption that DAC inhibits leukemogenesis through DNMT1 inhibition and subsequent DNA hypomethylation and highlight the potent activity of DAC to disrupt mitosis through aberrant DNMT1-DNA covalent bonds.
Collapse
Affiliation(s)
- Tomohiro Yabushita
- Division of Cellular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takumi Chinen
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Atsuya Nishiyama
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuhei Asada
- Division of Cellular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; The Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Ruka Shimura
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomoya Isobe
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan; Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keita Yamamoto
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Naru Sato
- Division of Cellular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yutaka Enomoto
- Division of Cellular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yosuke Tanaka
- Division of Cellular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tomofusa Fukuyama
- Division of Cellular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Hematology, International University of Health and Welfare Hospital, Tochigi, Japan
| | - Hitoshi Satoh
- Division of Medical Genome Sciences, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Keiko Kato
- Infinity Lab, INC, Yamagata, Japan; Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Kaori Saitoh
- Infinity Lab, INC, Yamagata, Japan; Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Takamasa Ishikawa
- Infinity Lab, INC, Yamagata, Japan; Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Yasuhito Nannya
- Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Daiju Kitagawa
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Hyogo, Japan
| | - Susumu Goyama
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
11
|
Cao M, Zou X, Li C, Lin Z, Wang N, Zou Z, Ye Y, Seemann J, Levine B, Tang Z, Zhong Q. An actin filament branching surveillance system regulates cell cycle progression, cytokinesis and primary ciliogenesis. Nat Commun 2023; 14:1687. [PMID: 36973243 PMCID: PMC10042869 DOI: 10.1038/s41467-023-37340-z] [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: 04/07/2022] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Dysfunction of cell cycle control and defects of primary ciliogenesis are two features of many cancers. Whether these events are interconnected and the driving mechanism coordinating them remains elusive. Here, we identify an actin filament branching surveillance system that alerts cells of actin branching insufficiency and regulates cell cycle progression, cytokinesis and primary ciliogenesis. We find that Oral-Facial-Digital syndrome 1 functions as a class II Nucleation promoting factor to promote Arp2/3 complex-mediated actin branching. Perturbation of actin branching promotes OFD1 degradation and inactivation via liquid-to-gel transition. Elimination of OFD1 or disruption of OFD1-Arp2/3 interaction drives proliferating, non-transformed cells into quiescence with ciliogenesis by an RB-dependent mechanism, while it leads oncogene-transformed/cancer cells to incomplete cytokinesis and irreversible mitotic catastrophe via actomyosin ring malformation. Inhibition of OFD1 leads to suppression of multiple cancer cell growth in mouse xenograft models. Thus, targeting OFD1-mediated actin filament branching surveillance system provides a direction for cancer therapy.
Collapse
Affiliation(s)
- Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China.
| | - Xiaoxiao Zou
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Chaoyi Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Zaisheng Lin
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Ni Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Zhongju Zou
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Youqiong Ye
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Joachim Seemann
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zaiming Tang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China.
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China.
| |
Collapse
|
12
|
Bellingham-Johnstun K, Tyree ZL, Martinez-Baird J, Thorn A, Laplante C. Actin–Microtubule Crosstalk Imparts Stiffness to the Contractile Ring in Fission Yeast. Cells 2023; 12:cells12060917. [PMID: 36980258 PMCID: PMC10047812 DOI: 10.3390/cells12060917] [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: 03/01/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Actin–microtubule interactions are critical for cell division, yet how these networks of polymers mutually influence their mechanical properties and functions in live cells remains unknown. In fission yeast, the post-anaphase array (PAA) of microtubules assembles in the plane of the contractile ring, and its assembly relies on the Myp2p-dependent recruitment of Mto1p, a component of equatorial microtubule organizing centers (eMTOCs). The general organization of this array of microtubules and the impact on their physical attachment to the contractile ring remain unclear. We found that Myp2p facilitates the recruitment of Mto1p to the inner face of the contractile ring, where the eMTOCs polymerize microtubules without their direct interaction. The PAA microtubules form a dynamic polygon of Ase1p crosslinked microtubules inside the contractile ring. The specific loss of PAA microtubules affects the mechanical properties of the contractile ring of actin by lowering its stiffness. This change in the mechanical properties of the ring has no measurable impact on cytokinesis or on the anchoring of the ring. Our work proposes that the PAA microtubules exploit the contractile ring for their assembly and function during cell division, while the contractile ring may receive no benefit from these interactions.
Collapse
Affiliation(s)
- Kimberly Bellingham-Johnstun
- Molecular Biomedical Sciences Department, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Quantitative and Computational Developmental Biology Cluster, North Carolina State University, Raleigh, NC 27607, USA
| | - Zoe L. Tyree
- Molecular Biomedical Sciences Department, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Quantitative and Computational Developmental Biology Cluster, North Carolina State University, Raleigh, NC 27607, USA
| | - Jessica Martinez-Baird
- Molecular Biomedical Sciences Department, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Quantitative and Computational Developmental Biology Cluster, North Carolina State University, Raleigh, NC 27607, USA
| | - Annelise Thorn
- Molecular Biomedical Sciences Department, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Quantitative and Computational Developmental Biology Cluster, North Carolina State University, Raleigh, NC 27607, USA
| | - Caroline Laplante
- Molecular Biomedical Sciences Department, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Quantitative and Computational Developmental Biology Cluster, North Carolina State University, Raleigh, NC 27607, USA
- Correspondence:
| |
Collapse
|
13
|
Inhibitory Effects of 3-Methylcholanthrene Exposure on Porcine Oocyte Maturation. Int J Mol Sci 2023; 24:ijms24065567. [PMID: 36982641 PMCID: PMC10058619 DOI: 10.3390/ijms24065567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
3-methylcholanthrene (3-MC) is a highly toxic environmental pollutant that impairs animal health. 3-MC exposure can cause abnormal spermatogenesis and ovarian dysfunction. However, the effects of 3-MC exposure on oocyte maturation and embryo development remain unclear. This study revealed the toxic effects of 3-MC exposure on oocyte maturation and embryo development. 3-MC with different concentrations of 0, 25, 50, and 100 μM was applied for in vitro maturation of porcine oocytes. The results showed that 100 μM 3-MC significantly inhibited cumulus expansion and the first polar body extrusion. The rates of cleavage and blastocyst of embryos derived from 3-MC-exposed oocytes were significantly lower than those in the control group. Additionally, the rates of spindle abnormalities and chromosomal misalignments were higher than those in the control group. Furthermore, 3-MC exposure not only decreased the levels of mitochondria, cortical granules (CGs), and acetylated α-Tubulin, but also increased the levels of reactive oxygen species (ROS), DNA damage, and apoptosis. The expression of cumulus expansion and apoptosis-related genes was abnormal in 3-MC-exposed oocytes. In conclusion, 3-MC exposure disrupted the nuclear and cytoplasmic maturation of porcine oocytes through oxidative stress.
Collapse
|
14
|
Bellingham-Johnstun K, Tyree ZL, Martinez-Baird J, Thorn A, Laplante C. Actin-microtubule crosstalk imparts stiffness to the contractile ring in fission yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530611. [PMID: 36909652 PMCID: PMC10002727 DOI: 10.1101/2023.03.01.530611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Actin-microtubule interactions are critical for cell division yet how these networks of polymers mutually influence their mechanical properties and functions in live cells remains unknown. In fission yeast, the post-anaphase array (PAA) of microtubules assembles in the plane of the contractile ring and its assembly relies on the Myp2p-dependent recruitment of Mto1p, a component of equatorial microtubule organizing centers (eMTOCs). The general organization of this array of microtubule and the impact on their physical attachment to the contractile ring remain unclear. We found that Myp2p facilitates the recruitment of Mto1p to the inner face of the contractile ring where the eMTOCs polymerize microtubules without their direct interaction. The PAA microtubules form a dynamic polygon of Ase1p crosslinked microtubules inside the contractile ring. The specific loss of PAA microtubules affects the mechanical properties of the contractile ring of actin by lowering its stiffness. This change in the mechanical properties of the ring has no measurable impact on cytokinesis or on the anchoring of the ring. Our work proposes that the PAA microtubules exploit the contractile ring for their assembly and function during cell division while the contractile ring may receive no benefit from these interactions.
Collapse
|
15
|
Remsburg CM, Konrad KD, Song JL. RNA localization to the mitotic spindle is essential for early development and is regulated by kinesin-1 and dynein. J Cell Sci 2023; 136:jcs260528. [PMID: 36751992 PMCID: PMC10038151 DOI: 10.1242/jcs.260528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023] Open
Abstract
Mitosis is a fundamental and highly regulated process that acts to faithfully segregate chromosomes into two identical daughter cells. Localization of gene transcripts involved in mitosis to the mitotic spindle might be an evolutionarily conserved mechanism to ensure that mitosis occurs in a timely manner. We identified many RNA transcripts that encode proteins involved in mitosis localized at the mitotic spindles in dividing sea urchin embryos and mammalian cells. Disruption of microtubule polymerization, kinesin-1 or dynein results in lack of spindle localization of these transcripts in the sea urchin embryo. Furthermore, results indicate that the cytoplasmic polyadenylation element (CPE) within the 3'UTR of the Aurora B transcript, a recognition sequence for CPEB, is essential for RNA localization to the mitotic spindle in the sea urchin embryo. Blocking this sequence results in arrested development during early cleavage stages, suggesting that RNA localization to the mitotic spindle might be a regulatory mechanism of cell division that is important for early development.
Collapse
Affiliation(s)
- Carolyn M. Remsburg
- University of Delaware, Department of Biological Sciences, Newark, DE 19716, USA
| | - Kalin D. Konrad
- University of Delaware, Department of Biological Sciences, Newark, DE 19716, USA
| | - Jia L. Song
- University of Delaware, Department of Biological Sciences, Newark, DE 19716, USA
| |
Collapse
|
16
|
Daeden A, Mietke A, Derivery E, Seum C, Jülicher F, Gonzalez-Gaitan M. Polarized branched Actin modulates cortical mechanics to produce unequal-size daughters during asymmetric division. Nat Cell Biol 2023; 25:235-245. [PMID: 36747081 PMCID: PMC9928585 DOI: 10.1038/s41556-022-01058-9] [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: 08/03/2021] [Accepted: 11/22/2022] [Indexed: 02/08/2023]
Abstract
The control of cell shape during cytokinesis requires a precise regulation of mechanical properties of the cell cortex. Only few studies have addressed the mechanisms underlying the robust production of unequal-sized daughters during asymmetric cell division. Here we report that unequal daughter-cell sizes resulting from asymmetric sensory organ precursor divisions in Drosophila are controlled by the relative amount of cortical branched Actin between the two cell poles. We demonstrate this by mistargeting the machinery for branched Actin dynamics using nanobodies and optogenetics. We can thereby engineer the cell shape with temporal precision and thus the daughter-cell size at different stages of cytokinesis. Most strikingly, inverting cortical Actin asymmetry causes an inversion of daughter-cell sizes. Our findings uncover the physical mechanism by which the sensory organ precursor mother cell controls relative daughter-cell size: polarized cortical Actin modulates the cortical bending rigidity to set the cell surface curvature, stabilize the division and ultimately lead to unequal daughter-cell size.
Collapse
Affiliation(s)
- Alicia Daeden
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Alexander Mietke
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
| | - Emmanuel Derivery
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Carole Seum
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Marcos Gonzalez-Gaitan
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
17
|
Chen J, Du X, Xu X, Zhang S, Yao L, He X, Wang Y. Comparative Proteomic Analysis Provides New Insights into the Molecular Basis of Thermal-Induced Parthenogenesis in Silkworm ( Bombyx mori). INSECTS 2023; 14:insects14020134. [PMID: 36835703 PMCID: PMC9962255 DOI: 10.3390/insects14020134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 05/27/2023]
Abstract
Artificial parthenogenetic induction via thermal stimuli in silkworm is an important technique that has been used in sericultural production. However, the molecular mechanism underlying it remains largely unknown. We have created a fully parthenogenetic line (PL) with more than 85% occurrence and 80% hatching rate via hot water treatment and genetic selection, while the parent amphigenetic line (AL) has less than 30% pigmentation rate and less than 1% hatching rate when undergoing the same treatment. Here, isobaric tags for relative and absolute quantitation (iTRAQ)-based analysis were used to investigate the key proteins and pathways associated with silkworm parthenogenesis. We uncovered the unique proteomic features of unfertilized eggs in PL. In total, 274 increased abundance proteins and 211 decreased abundance proteins were identified relative to AL before thermal induction. Function analysis displayed an increased level of translation and metabolism in PL. After thermal induction, 97 increased abundance proteins and 187 decreased abundance proteins were identified. An increase in stress response-related proteins and decrease in energy metabolism suggested that PL has a more effective response to buffer the thermal stress than AL. Cell cycle-related proteins, including histones, and spindle-related proteins were decreased in PL, indicating an important role of this decrease in the process of ameiotic parthenogenesis.
Collapse
Affiliation(s)
- Jine Chen
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xin Du
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xia Xu
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Lusong Yao
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xiuling He
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yongqiang Wang
- Institute of Sericulture and Tea, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| |
Collapse
|
18
|
Scotto di Carlo F, Russo S, Muyas F, Mangini M, Garribba L, Pazzaglia L, Genesio R, Biamonte F, De Luca AC, Santaguida S, Scotlandi K, Cortés-Ciriano I, Gianfrancesco F. Profilin 1 deficiency drives mitotic defects and reduces genome stability. Commun Biol 2023; 6:9. [PMID: 36599901 PMCID: PMC9813376 DOI: 10.1038/s42003-022-04392-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023] Open
Abstract
Profilin 1-encoded by PFN1-is a small actin-binding protein with a tumour suppressive role in various adenocarcinomas and pagetic osteosarcomas. However, its contribution to tumour development is not fully understood. Using fix and live cell imaging, we report that Profilin 1 inactivation results in multiple mitotic defects, manifested prominently by anaphase bridges, multipolar spindles, misaligned and lagging chromosomes, and cytokinesis failures. Accordingly, next-generation sequencing technologies highlighted that Profilin 1 knock-out cells display extensive copy-number alterations, which are associated with complex genome rearrangements and chromothripsis events in primary pagetic osteosarcomas with Profilin 1 inactivation. Mechanistically, we show that Profilin 1 is recruited to the spindle midzone at anaphase, and its deficiency reduces the supply of actin filaments to the cleavage furrow during cytokinesis. The mitotic defects are also observed in mouse embryonic fibroblasts and mesenchymal cells deriving from a newly generated knock-in mouse model harbouring a Pfn1 loss-of-function mutation. Furthermore, nuclear atypia is also detected in histological sections of mutant femurs. Thus, our results indicate that Profilin 1 has a role in regulating cell division, and its inactivation triggers mitotic defects, one of the major mechanisms through which tumour cells acquire chromosomal instability.
Collapse
Affiliation(s)
- Federica Scotto di Carlo
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “Adriano Buzzati-Traverso” (IGB), National Research Council of Italy (CNR), Naples, Italy
| | - Sharon Russo
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “Adriano Buzzati-Traverso” (IGB), National Research Council of Italy (CNR), Naples, Italy ,grid.9841.40000 0001 2200 8888Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Francesc Muyas
- grid.52788.300000 0004 0427 7672European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Maria Mangini
- grid.429047.c0000 0004 6477 0469Institute for Experimental Endocrinology and Oncology, “G. Salvatore” (IEOS), National Research Council of Italy (CNR), Naples, Italy
| | - Lorenza Garribba
- grid.15667.330000 0004 1757 0843Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Laura Pazzaglia
- grid.419038.70000 0001 2154 6641IRCCS Istituto Ortopedico Rizzoli, Laboratory of Experimental Oncology, Bologna, Italy
| | - Rita Genesio
- grid.4691.a0000 0001 0790 385XDepartment of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Flavia Biamonte
- grid.411489.10000 0001 2168 2547Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy ,grid.411489.10000 0001 2168 2547Center of Interdepartmental Services (CIS), Magna Graecia University, Catanzaro, Italy
| | - Anna Chiara De Luca
- grid.429047.c0000 0004 6477 0469Institute for Experimental Endocrinology and Oncology, “G. Salvatore” (IEOS), National Research Council of Italy (CNR), Naples, Italy
| | - Stefano Santaguida
- grid.15667.330000 0004 1757 0843Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy ,grid.4708.b0000 0004 1757 2822Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Katia Scotlandi
- grid.419038.70000 0001 2154 6641IRCCS Istituto Ortopedico Rizzoli, Laboratory of Experimental Oncology, Bologna, Italy
| | - Isidro Cortés-Ciriano
- grid.52788.300000 0004 0427 7672European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Fernando Gianfrancesco
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “Adriano Buzzati-Traverso” (IGB), National Research Council of Italy (CNR), Naples, Italy
| |
Collapse
|
19
|
WDR73 Depletion Destabilizes PIP4K2C Activity and Impairs Focal Adhesion Formation in Galloway–Mowat Syndrome. BIOLOGY 2022; 11:biology11101397. [PMID: 36290302 PMCID: PMC9598763 DOI: 10.3390/biology11101397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Galloway–Mowat syndrome is a rare genetic disease, classically characterized by a combination of various neurological symptoms and nephrotic syndrome. WDR73 is the pathogenic gene responsible for Galloway–Mowat syndrome. However, the pathological and molecular mechanisms of Galloway–Mowat syndrome, especially nephrotic syndrome caused by WDR73 deficiency, remains unknown. In this study, we knocked out the WDR73 in human embryonic kidney 293 cells to observe the morphological characteristics of the cells and elucidate the functions of WDR73. Additionally, we used a combination of proteomics, transcriptomics, and biochemical assays to identify the regulated targets of WDR73. We aimed to discover directly interacting molecules and the regulatory pathway of WDR73 and to illustrate the molecular mechanism between the WDR73 pathway and nephrotic disease in Galloway–Mowat syndrome. From the molecular mechanism we found in vitro, we draw a hypothesis that the damage to focal adhesion of podocytes caused by WDR73 defect is the key issue of kidney dysfunction. Finally, we verified the hypothesis in a podocyte-specific conditional knockout Wdr73 mouse model. Abstract (1) Background: Galloway–Mowat syndrome (GAMOS) is a rare genetic disease, classically characterized by a combination of various neurological symptoms and nephrotic syndrome. WDR73 is the pathogenic gene responsible for GAMOS1. However, the pathological and molecular mechanisms of GAMOS1, especially nephrotic syndrome caused by WDR73 deficiency, remain unknown. (2) Methods and Results: In this study, we first observed remarkable cellular morphological changes including impaired cell adhesion, decreased pseudopodia, and G2/M phase arrest in WDR73 knockout (KO) HEK 293 cells. The differentially expressed genes in WDR73 KO cells were enriched in the focal adhesion (FA) pathway. Additionally, PIP4K2C, a phospholipid kinase also involved in the FA pathway, was subsequently validated to interact with WDR73 via protein microarray and GST pulldown. WDR73 regulates PIP4K2C protein stability through the autophagy–lysosomal pathway. The stability of PIP4K2C was significantly disrupted by WDR73 KO, leading to a remarkable reduction in PIP2 and thus weakening the FA formation. In addition, we found that podocyte-specific conditional knockout (Wdr73 CKO) mice showed high levels of albuminuria and podocyte foot process injury in the ADR-induced model. FA formation was impaired in primary podocytes derived from Wdr73 CKO mice. (3) Conclusions: Since FA has been well known for its critical roles in maintaining podocyte structures and function, our study indicated that nephrotic syndrome in GAMOS1 is associated with disruption of FA caused by WDR73 deficiency.
Collapse
|
20
|
Sun F, Zhu G, He P, Wei E, Wang R, Wang Q, Tang X, Zhang Y, Shen Z. Identification, expression and subcellular localization of Orc1 in the microsporidian Nosema bombycis. Gene X 2022; 834:146607. [PMID: 35609797 DOI: 10.1016/j.gene.2022.146607] [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: 04/13/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 11/30/2022] Open
Abstract
As a typical species of microsporidium, Nosema bombycis is the pathogen causing the pébrine disease of silkworm. Rapid proliferation of N. bombycis in host cells requires replication of genetic material. As eukaryotic origin recognition protein, origin recognition complex (ORC) plays an important role in regulating DNA replication, and Orc1 is a key subunit of the origin recognition complex. In this study, we identified the Orc1 in the microsporidian N. bombycis (NbOrc1) for the first time. The NbOrc1 gene contains a complete ORF of 987 bp in length that encodes a 328 amino acid polypeptide. Indirect immunofluorescence results showed that NbOrc1 were colocalized with Nbactin and NbSAS-6 in the nuclei of N. bombycis. Subsequently, we further identified the interaction between the NbOrc1 and Nbactin by CO-IP and Western blot. These results imply that Orc1 may be involved in the proliferation of the microsporidian N. bombycis through interacting with actin.
Collapse
Affiliation(s)
- Fuzhen Sun
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China
| | - Guanyu Zhu
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China
| | - Ping He
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China
| | - Erjun Wei
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China
| | - Runpeng Wang
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China
| | - Qiang Wang
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China; Sericulture Research Institute of Chinese Academy of Agricultural Sciences, Zhenjiang 212018, Jiangsu Province, China
| | - Xudong Tang
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China; Sericulture Research Institute of Chinese Academy of Agricultural Sciences, Zhenjiang 212018, Jiangsu Province, China
| | - Yiling Zhang
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China; Sericulture Research Institute of Chinese Academy of Agricultural Sciences, Zhenjiang 212018, Jiangsu Province, China
| | - Zhongyuan Shen
- Jiangsu University of Science and Technology, Zhenjiang 212018, Jiangsu Province, China; Sericulture Research Institute of Chinese Academy of Agricultural Sciences, Zhenjiang 212018, Jiangsu Province, China.
| |
Collapse
|
21
|
Pascal A, Gallaud E, Giet R, Benaud C. Annexin A2 and Ahnak control cortical NuMA-dynein localization and mitotic spindle orientation. J Cell Sci 2022; 135:274948. [PMID: 35362526 DOI: 10.1242/jcs.259344] [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: 09/01/2021] [Accepted: 03/25/2022] [Indexed: 11/20/2022] Open
Abstract
Proper mitotic spindle orientation depends on the correct anchorage of astral microtubules to the cortex. It relies on the remodeling of the cell cortex, a process not fully understood. Annexin A2 (Anx2) is a protein known to be involved in cortical domain remodeling. Here, we report that in early mitosis, Anx2 recruits the scaffold protein Ahnak at the cell cortex facing spindle poles, and the distribution of both proteins is controlled by cell adhesion. Depletion of either protein or impaired cortical Ahnak localization result in delayed anaphase onset and unstable spindle anchoring, which leads to altered spindle orientation. We find that Ahnak is present in a complex with dynein-dynactin. Furthermore, Ahnak and Anx2 are required for dynein and NuMA proper cortical localization and dynamics. We propose that the Ahnak/Anx2 complex influences the cortical organization of the astral microtubule anchoring complex, and thereby mitotic spindle positioning in human cells.
Collapse
Affiliation(s)
- Aude Pascal
- University Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, ERL U1305, F-35000 Rennes, France
| | - Emmanuel Gallaud
- University Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, ERL U1305, F-35000 Rennes, France
| | - Regis Giet
- University Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, ERL U1305, F-35000 Rennes, France
| | - Christelle Benaud
- University Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, ERL U1305, F-35000 Rennes, France
| |
Collapse
|
22
|
Donà F, Eli S, Mapelli M. Insights Into Mechanisms of Oriented Division From Studies in 3D Cellular Models. Front Cell Dev Biol 2022; 10:847801. [PMID: 35356279 PMCID: PMC8959941 DOI: 10.3389/fcell.2022.847801] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs, that ensure the correct organ formation and functioning. In these processes, mitotic rates and division orientation are crucial in regulating the velocity and the timing of the forming tissue. Division orientation, specified by mitotic spindle placement with respect to epithelial apico-basal polarity, controls not only the partitioning of cellular components but also the positioning of the daughter cells within the tissue, and hence the contacts that daughter cells retain with the surrounding microenvironment. Daughter cells positioning is important to determine signal sensing and fate, and therefore the final function of the developing organ. In this review, we will discuss recent discoveries regarding the mechanistics of planar divisions in mammalian epithelial cells, summarizing technologies and model systems used to study oriented cell divisions in vitro such as three-dimensional cysts of immortalized cells and intestinal organoids. We also highlight how misorientation is corrected in vivo and in vitro, and how it might contribute to the onset of pathological conditions.
Collapse
Affiliation(s)
- Federico Donà
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Susanna Eli
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | |
Collapse
|
23
|
Ambaru B, Gangadharan GM, Subramanya HS, Gupta CM. Profilin is involved in G1 to S phase progression and mitotic spindle orientation during Leishmania donovani cell division cycle. PLoS One 2022; 17:e0265692. [PMID: 35316283 PMCID: PMC8939790 DOI: 10.1371/journal.pone.0265692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/04/2022] [Indexed: 11/27/2022] Open
Abstract
Profilin is a multi-ligand binding protein, which is a key regulator of actin dynamics and involved in regulating several cellular functions. It is present in all eukaryotes, including trypanosomatids such as Leishmania. However, not much is known about its functions in these organisms. Our earlier studies have shown that Leishmania parasites express a single homologue of profilin (LdPfn) that binds actin, phosphoinositides and poly- L- proline motives, and depletion of its intracellular pool to 50%of normal levels affects the cell growth and intracellular trafficking. Here, we show, employing affinity pull-down and mass spectroscopy, that LdPfn interacted with a large number of proteins, including those involved in mRNA processing and protein translation initiation, such as eIF4A1. Further, we reveal, using mRNA Seq analysis, that depletion of LdPfn in Leishmania cells (LdPfn+/-) resulted in significantly reduced expression of genes which encode proteins involved in cell cycle regulation, mRNA translation initiation, nucleosides and amino acids transport. In addition, we show that in LdPfn+/- cells, cellular levels of eIF4A1 protein were significantly decreased, and during their cell division cycle, G1-to-S phase progression was delayed and orientation of mitotic spindle altered. These changes were, however, reversed to normal by episomal expression of GFP-LdPfn in LdPfn+/- cells. Taken together, our results indicate that profilin is involved in regulation of G1-to-S phase progression and mitotic spindle orientation in Leishmania cell cycle, perhaps through its interaction with elF4A1 protein.
Collapse
Affiliation(s)
- Bindu Ambaru
- Institute of Bioinformatics and Applied Biotechnology, Bengaluru, Karnataka, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | | | - Chhitar M. Gupta
- Institute of Bioinformatics and Applied Biotechnology, Bengaluru, Karnataka, India
- * E-mail:
| |
Collapse
|
24
|
Mubarok W, Elvitigala KCML, Nakahata M, Kojima M, Sakai S. Modulation of Cell-Cycle Progression by Hydrogen Peroxide-Mediated Cross-Linking and Degradation of Cell-Adhesive Hydrogels. Cells 2022; 11:cells11050881. [PMID: 35269503 PMCID: PMC8909037 DOI: 10.3390/cells11050881] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
The cell cycle is known to be regulated by features such as the mechanical properties of the surrounding environment and interaction of cells with the adhering substrates. Here, we investigated the possibility of regulating cell-cycle progression of the cells on gelatin/hyaluronic acid composite hydrogels obtained through hydrogen peroxide (H2O2)-mediated cross-linking and degradation of the polymers by varying the exposure time to H2O2 contained in the air. The stiffness of the hydrogel varied with the exposure time. Human cervical cancer cells (HeLa) and mouse mammary gland epithelial cells (NMuMG) expressing cell-cycle reporter Fucci2 showed the exposure-time-dependent different cell-cycle progressions on the hydrogels. Although HeLa/Fucci2 cells cultured on the soft hydrogel (Young’s modulus: 0.20 and 0.40 kPa) obtained through 15 min and 120 min of the H2O2 exposure showed a G2/M-phase arrest, NMuMG cells showed a G1-phase arrest. Additionally, the cell-cycle progression of NMuMG cells was not only governed by the hydrogel stiffness, but also by the low-molecular-weight HA resulting from H2O2-mediated degradation. These results indicate that H2O2-mediated cross-linking and degradation of gelatin/hyaluronic acid composite hydrogel could be used to control the cell adhesion and cell-cycle progression.
Collapse
|
25
|
Uittenboogaard A, Neutel CLG, Ket JCF, Njuguna F, Huitema ADR, Kaspers GJL, van de Velde ME. Pharmacogenomics of Vincristine-Induced Peripheral Neuropathy in Children with Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14030612. [PMID: 35158880 PMCID: PMC8833506 DOI: 10.3390/cancers14030612] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Vincristine is a drug that is part of the treatment for many children with cancer. Its main side-effect is vincristine-induced peripheral neuropathy (VIPN), which often presents as tingling, pain, and lack of strength in the hands and feet. It is not yet possible to predict which children will suffer from VIPN. In this review, we report on all genetic variations that are associated with VIPN. We found that variations in genes related to vincristine transport, cell structure, hereditary nerve disease, and genes without a previously known connection to vincristine or VIPN are related to VIPN. Variations in genes involved in vincristine breakdown are not significantly associated with VIPN. In conclusion, genetic variations affect a child’s tendency to develop VIPN. In the future, this information might be used to predict the risk of VIPN and adapt treatment on this. Abstract Vincristine-induced peripheral neuropathy (VIPN) is a debilitating side-effect of vincristine. It remains a challenge to predict which patients will suffer from VIPN. Pharmacogenomics may explain an individuals’ susceptibility to side-effects. In this systematic review and meta-analysis, we describe the influence of pharmacogenomic parameters on the development of VIPN in children with cancer. PubMed, Embase and Web of Science were searched. In total, 1597 records were identified and 21 studies were included. A random-effects meta-analysis was performed for the influence of CYP3A5 expression on the development of VIPN. Single-nucleotide polymorphisms (SNPs) in transporter-, metabolism-, cytoskeleton-, and hereditary neuropathy-associated genes and SNPs in genes previously unrelated to vincristine or neuropathy were associated with VIPN. CYP3A5 expression status was not significantly associated with VIPN. The comparison and interpretation of the results of the included studies was limited due to heterogeneity in the study population, treatment protocol and assessment methods and definitions of VIPN. Independent replication is essential to validate the clinical significance of the reported associations. Future research should aim for prospective VIPN assessment in both a discovery and a replication cohort. Ultimately, the goal would be to screen patients upfront to determine optimal vincristine dosage with regards to efficacy and risk of VIPN.
Collapse
Affiliation(s)
- Aniek Uittenboogaard
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1105 AZ Amsterdam, The Netherlands;
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Correspondence: (A.U.); (G.J.L.K.)
| | - Céline L. G. Neutel
- Department of Neurosurgery, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Johannes C. F. Ket
- Medical Library, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Festus Njuguna
- Department of Pediatric Oncology, Moi University, Eldoret 30107, Kenya;
| | - Alwin D. R. Huitema
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Gertjan J. L. Kaspers
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1105 AZ Amsterdam, The Netherlands;
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Correspondence: (A.U.); (G.J.L.K.)
| | - Mirjam E. van de Velde
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|
26
|
Zhang Z, Liu R, Wang Y, Wang Y, Shuai Y, Ke C, Jin R, Wang X, Luo J. Phosphorylation of MICAL2 by ARG promotes head and neck cancer tumorigenesis by regulating skeletal rearrangement. Oncogene 2022; 41:334-346. [PMID: 34750518 DOI: 10.1038/s41388-021-02101-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/22/2022]
Abstract
The actin cytoskeletal architecture provides the structural underpinnings for crucial cellular behaviors. In cancer cells, changes in the actin cytoskeleton may serve as prerequisites for proliferation, invasion, and metastatic dissemination. However, the underlying mechanisms remain largely unknown. Here, we show that MICAL2, which is increased in head and neck squamous cell carcinoma (HNSCC) and inversely associated with patient survival, promotes HNSCC growth, invasion, and migration. MICAL2 serves as a flavoprotein monooxygenase and directly induces actin filament depolymerization by specifically oxidizing the methionine 44 and 47 residues of F-actin. The kinase ARG interacts with MICAL2 and augments MICAL2-mediated actin disassembly. Direct phosphorylation assay and mass spectrometry confirmed that ARG phosphorylates MICAL2 at Tyr445, Tyr463, and Tyr488. Substitution of the Tyr445 or Tyr463 residue of purified recombinant MICAL2-redox with phenylalanine (generating a non-phosphorylatable mutant) abolishes the enhanced MICAL2-mediated F-actin disassembly induced by ARG. Consistently, ectopic expression of non-phosphorylatable MICAL2 mutants (MICAL2Y445F and MICAL2Y463F, not MICAL2Y488F) failed to ameliorate HNSCC cell growth, whereas expression of wild-type MICAL2 or MICAL2Y488F rescued the impaired proliferation induced by MICAL2 knockdown. Moreover, CCG-1423, an inhibitor of MICAL2, was shown to inhibit HNSCC cell proliferation, invasion, and migration. Taken together, our findings indicate that phosphorylation of MICAL2 at Tyr445 and Tyr463 by ARG mediates F-actin disassembly and promotes HNSCC progression.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ruoyan Liu
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yafei Wang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yun Wang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yanjie Shuai
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chuangwu Ke
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Rui Jin
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jingtao Luo
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
27
|
Abstract
Actin filaments and microtubules are cytoskeletal polymers that participate in many vital cell functions including division, morphogenesis, phagocytosis, and motility. Despite the persistent dogma that actin filament and microtubule networks are distinct in localization, structure, and function, a growing body of evidence shows that these elements are choreographed through intricate mechanisms sensitive to either polymer. Many proteins and cellular signals that mediate actin–microtubule interactions have already been identified. However, the impact of these regulators is typically assessed with actin filament or microtubule polymers alone, independent of the other system. Further, unconventional modes and regulators coordinating actin–microtubule interactions are still being discovered. Here we examine several methods of actin–microtubule crosstalk with an emphasis on the molecular links between both polymer systems and their higher-order interactions.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Jessica L Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210.,Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
28
|
Huang SC, Vu LV, Yu FH, Nguyen DT, Benz EJ. Multifunctional protein 4.1R regulates the asymmetric segregation of Numb during terminal erythroid maturation. J Biol Chem 2021; 297:101051. [PMID: 34364872 PMCID: PMC8408529 DOI: 10.1016/j.jbc.2021.101051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 10/25/2022] Open
Abstract
The asymmetric cell division of stem or progenitor cells generates daughter cells with distinct fates that balance proliferation and differentiation. Asymmetric segregation of Notch signaling regulatory protein Numb plays a crucial role in cell diversification. However, the molecular mechanism remains unclear. Here, we examined the unequal distribution of Numb in the daughter cells of murine erythroleukemia cells (MELCs) that undergo DMSO-induced erythroid differentiation. In contrast to the cytoplasmic localization of Numb during uninduced cell division, Numb is concentrated at the cell boundary in interphase, near the one-spindle pole in metaphase, and is unequally distributed to one daughter cell in anaphase in induced cells. The inheritance of Numb guides this daughter cell toward erythroid differentiation while the other cell remains a progenitor cell. Mitotic spindle orientation, critical for distribution of cell fate determinants, requires complex communication between the spindle microtubules and the cell cortex mediated by the NuMA-LGN-dynein/dynactin complex. Depletion of each individual member of the complex randomizes the position of Numb relative to the mitotic spindle. Gene replacement confirms that multifunctional erythrocyte protein 4.1R (4.1R) functions as a member of the NuMA-LGN-dynein/dynactin complex and is necessary for regulating spindle orientation, in which interaction between 4.1R and NuMA plays an important role. These results suggest that mispositioning of Numb is the result of spindle misorientation. Finally, disruption of the 4.1R-NuMA-LGN complex increases Notch signaling and decreases the erythroblast population. Together, our results identify a critical role for 4.1R in regulating the asymmetric segregation of Numb to mediate erythropoiesis.
Collapse
Affiliation(s)
- Shu-Ching Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| | - Long V Vu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Faye H Yu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Dan T Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Edward J Benz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Department of Pediatrics and Genetics, Harvard Medical School, Boston, Massachusetts, USA; Leukemia Program, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Abstract
The cytoskeleton - comprising actin filaments, microtubules and intermediate filaments - serves instructive roles in regulating cell function and behaviour during development. However, a key challenge in cell and developmental biology is to dissect how these different structures function and interact in vivo to build complex tissues, with the ultimate aim to understand these processes in a mammalian organism. The preimplantation mouse embryo has emerged as a primary model system for tackling this challenge. Not only does the mouse embryo share many morphological similarities with the human embryo during its initial stages of life, it also permits the combination of genetic manipulations with live-imaging approaches to study cytoskeletal dynamics directly within an intact embryonic system. These advantages have led to the discovery of novel cytoskeletal structures and mechanisms controlling lineage specification, cell-cell communication and the establishment of the first forms of tissue architecture during development. Here we highlight the diverse organization and functions of each of the three cytoskeletal filaments during the key events that shape the early mammalian embryo, and discuss how they work together to perform key developmental tasks, including cell fate specification and morphogenesis of the blastocyst. Collectively, these findings are unveiling a new picture of how cells in the early embryo dynamically remodel their cytoskeleton with unique spatial and temporal precision to drive developmental processes in the rapidly changing in vivo environment.
Collapse
|
30
|
Ikushima YM, Awazawa M, Kobayashi N, Osonoi S, Takemiya S, Kobayashi H, Suwanai H, Morimoto Y, Soeda K, Adachi J, Muratani M, Charron J, Mizukami H, Takahashi N, Ueki K. MEK/ERK Signaling in β-Cells Bifunctionally Regulates β-Cell Mass and Glucose-Stimulated Insulin Secretion Response to Maintain Glucose Homeostasis. Diabetes 2021; 70:1519-1535. [PMID: 33906910 DOI: 10.2337/db20-1295] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/23/2021] [Indexed: 11/13/2022]
Abstract
In diabetic pathology, insufficiency in β-cell mass, unable to meet peripheral insulin demand, and functional defects of individual β-cells in production of insulin are often concurrently observed, collectively causing hyperglycemia. Here we show that the phosphorylation of ERK1/2 is significantly decreased in the islets of db/db mice as well as in those of a cohort of subjects with type 2 diabetes. In mice with abrogation of ERK signaling in pancreatic β-cells through deletion of Mek1 and Mek2, glucose intolerance aggravates under high-fat diet-feeding conditions due to insufficient insulin production with lower β-cell proliferation and reduced β-cell mass, while in individual β-cells dampening of the number of insulin exocytosis events is observed, with the molecules involved in insulin exocytosis being less phosphorylated. These data reveal bifunctional roles for MEK/ERK signaling in β-cells for glucose homeostasis, i.e., in regulating β-cell mass as well as in controlling insulin exocytosis in individual β-cells, thus providing not only a novel perspective for the understanding of diabetes pathophysiology but also a potential clue for new drug development for diabetes treatment.
Collapse
Affiliation(s)
- Yoshiko Matsumoto Ikushima
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Motoharu Awazawa
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Naoki Kobayashi
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Sho Osonoi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Seiichi Takemiya
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroshi Kobayashi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hirotsugu Suwanai
- Department of Diabetes, Metabolism and Endocrinology, Tokyo Medical University, Tokyo, Japan
| | - Yuichi Morimoto
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| | - Kotaro Soeda
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jun Adachi
- Laboratory of Proteome Research, Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Jean Charron
- Centre de Recherche sur le Cancer de l'Université Laval, L'Hôtel-Dieu de Québec, Quebec City, Quebec, Canada
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Noriko Takahashi
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kohjiro Ueki
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Molecular Diabetology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
31
|
Bouvrais H, Chesneau L, Le Cunff Y, Fairbrass D, Soler N, Pastezeur S, Pécot T, Kervrann C, Pécréaux J. The coordination of spindle-positioning forces during the asymmetric division of the Caenorhabditis elegans zygote. EMBO Rep 2021; 22:e50770. [PMID: 33900015 DOI: 10.15252/embr.202050770] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022] Open
Abstract
In Caenorhabditis elegans zygote, astral microtubules generate forces essential to position the mitotic spindle, by pushing against and pulling from the cortex. Measuring microtubule dynamics there, we revealed the presence of two populations, corresponding to pulling and pushing events. It offers a unique opportunity to study, under physiological conditions, the variations of both spindle-positioning forces along space and time. We propose a threefold control of pulling force, by polarity, spindle position and mitotic progression. We showed that the sole anteroposterior asymmetry in dynein on-rate, encoding pulling force imbalance, is sufficient to cause posterior spindle displacement. The positional regulation, reflecting the number of microtubule contacts in the posterior-most region, reinforces this imbalance only in late anaphase. Furthermore, we exhibited the first direct proof that dynein processivity increases along mitosis. It reflects the temporal control of pulling forces, which strengthens at anaphase onset following mitotic progression and independently from chromatid separation. In contrast, the pushing force remains constant and symmetric and contributes to maintaining the spindle at the cell centre during metaphase.
Collapse
Affiliation(s)
| | | | - Yann Le Cunff
- CNRS, IGDR - UMR 6290, University of Rennes, Rennes, France
| | | | - Nina Soler
- CNRS, IGDR - UMR 6290, University of Rennes, Rennes, France
| | | | - Thierry Pécot
- INRIA, Centre Rennes - Bretagne Atlantique, Rennes, France
| | | | | |
Collapse
|
32
|
Nunes V, Ferreira JG. From the cytoskeleton to the nucleus: An integrated view on early spindle assembly. Semin Cell Dev Biol 2021; 117:42-51. [PMID: 33726956 DOI: 10.1016/j.semcdb.2021.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/01/2022]
Abstract
Accurate chromosome segregation requires a complete restructuring of cellular organization. Microtubules remodel to assemble a mitotic spindle and the actin cytoskeleton rearranges to form a stiff actomyosin cortex. These cytoplasmic events must be spatially and temporally coordinated with mitotic chromosome condensation and nuclear envelope permeabilization, in order to ensure mitotic timing and fidelity. Here, we discuss the main cytoskeletal and nuclear events that occur during mitotic entry in proliferating animal cells, focusing on their coordinated contribution for early mitotic spindle assembly. We will also explore recent progress in understanding their regulatory biochemical and mechanical pathways.
Collapse
Affiliation(s)
- Vanessa Nunes
- Instituto de Investigação e Inovação em Saúde - i3S, University of Porto, Porto, Portugal; BiotechHealth PhD Programe, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Jorge G Ferreira
- Instituto de Investigação e Inovação em Saúde - i3S, University of Porto, Porto, Portugal; Departamento de Biomedicina, Faculdade de Medicina, University of Porto, Porto, Portugal.
| |
Collapse
|
33
|
Mechanochemical control of epidermal stem cell divisions by B-plexins. Nat Commun 2021; 12:1308. [PMID: 33637728 PMCID: PMC7910479 DOI: 10.1038/s41467-021-21513-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 01/28/2021] [Indexed: 01/31/2023] Open
Abstract
The precise spatiotemporal control of cell proliferation is key to the morphogenesis of epithelial tissues. Epithelial cell divisions lead to tissue crowding and local changes in force distribution, which in turn suppress the rate of cell divisions. However, the molecular mechanisms underlying this mechanical feedback are largely unclear. Here, we identify a critical requirement of B-plexin transmembrane receptors in the response to crowding-induced mechanical forces during embryonic skin development. Epidermal stem cells lacking B-plexins fail to sense mechanical compression, resulting in disinhibition of the transcriptional coactivator YAP, hyperproliferation, and tissue overgrowth. Mechanistically, we show that B-plexins mediate mechanoresponses to crowding through stabilization of adhesive cell junctions and lowering of cortical stiffness. Finally, we provide evidence that the B-plexin-dependent mechanochemical feedback is also pathophysiologically relevant to limit tumor growth in basal cell carcinoma, the most common type of skin cancer. Our data define a central role of B-plexins in mechanosensation to couple cell density and cell division in development and disease.
Collapse
|
34
|
Cristofani R, Piccolella M, Crippa V, Tedesco B, Montagnani Marelli M, Poletti A, Moretti RM. The Role of HSPB8, a Component of the Chaperone-Assisted Selective Autophagy Machinery, in Cancer. Cells 2021; 10:335. [PMID: 33562660 PMCID: PMC7915307 DOI: 10.3390/cells10020335] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The cellular response to cancer-induced stress is one of the major aspects regulating cancer development and progression. The Heat Shock Protein B8 (HSPB8) is a small chaperone involved in chaperone-assisted selective autophagy (CASA). CASA promotes the selective degradation of proteins to counteract cell stress such as tumor-induced stress. HSPB8 is also involved in (i) the cell division machinery regulating chromosome segregation and cell cycle arrest in the G0/G1 phase and (ii) inflammation regulating dendritic cell maturation and cytokine production. HSPB8 expression and role are tumor-specific, showing a dual and opposite role. Interestingly, HSPB8 may be involved in the acquisition of chemoresistance to drugs. Despite the fact the mechanisms of HSPB8-mediated CASA activation in tumors need further studies, HSPB8 could represent an important factor in cancer induction and progression and it may be a potential target for anticancer treatment in specific types of cancer. In this review, we will discuss the molecular mechanism underlying HSPB8 roles in normal and cancer conditions. The basic mechanisms involved in anti- and pro-tumoral activities of HSPB8 are deeply discussed together with the pathways that modulate HSPB8 expression, in order to outline molecules with a beneficial effect for cancer cell growth, migration, and death.
Collapse
|
35
|
Bloomfield M, Chen J, Cimini D. Spindle Architectural Features Must Be Considered Along With Cell Size to Explain the Timing of Mitotic Checkpoint Silencing. Front Physiol 2021; 11:596263. [PMID: 33584330 PMCID: PMC7877541 DOI: 10.3389/fphys.2020.596263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/23/2020] [Indexed: 11/25/2022] Open
Abstract
Mitosis proceeds through a defined series of events that is largely conserved, but the amount of time needed for their completion can vary in different cells and organisms. In many systems, mitotic duration depends on the time required to satisfy and silence the spindle assembly checkpoint (SAC), also known as the mitotic checkpoint. Because SAC silencing involves trafficking SAC molecules among kinetochores, spindle, and cytoplasm, the size and geometry of the spindle relative to cell volume are expected to affect mitotic duration by influencing the timing of SAC silencing. However, the relationship between SAC silencing, cell size, and spindle dimensions is unclear. To investigate this issue, we used four DLD-1 tetraploid (4N) clones characterized by small or large nuclear and cell size. We found that the small 4N clones had longer mitotic durations than the parental DLD-1 cells and that this delay was due to differences in their metaphase duration. Leveraging a previous mathematical model for spatiotemporal regulation of SAC silencing, we show that the difference in metaphase duration, i.e., SAC silencing time, can be explained by the distinct spindle microtubule densities and sizes of the cell, spindle, and spindle poles in the 4N clones. Lastly, we demonstrate that manipulating spindle geometry can alter mitotic and metaphase duration, consistent with a model prediction. Our results suggest that spindle size does not always scale with cell size in mammalian cells and cell size is not sufficient to explain the differences in metaphase duration. Only when a number of spindle architectural features are considered along with cell size can the kinetics of SAC silencing, and hence mitotic duration, in the different clones be explained.
Collapse
Affiliation(s)
- Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Jing Chen
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Daniela Cimini
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
36
|
In Vitro Reconstitution of Dynamic Co-organization of Microtubules and Actin Filaments in Emulsion Droplets. Methods Mol Biol 2021. [PMID: 31879898 DOI: 10.1007/978-1-0716-0219-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
In vitro (or cell-free) reconstitution is a powerful tool to study the physical basis of cytoskeletal organization in eukaryotic cells. Cytoskeletal reconstitution studies have mostly been done for individual cytoskeleton systems in unconfined 3D or quasi-2D geometries, which lack complexity relative to a cellular environment. To increase the level of complexity, we present a method to study co-organization of two cytoskeletal components, namely microtubules and actin filaments, confined in cell-sized water-in-oil emulsion droplets. We show that centrosome-nucleated dynamic microtubules can be made to interact with actin filaments through a tip-tracking complex consisting of microtubule end-binding proteins and an actin-microtubule cytolinker. In addition to the protocols themselves, we discuss the optimization steps required in order to build these more complex in vitro model systems of cytoskeletal interactions.
Collapse
|
37
|
Machaliński B, Rogińska D, Wilk A, Szumilas K, Prowans P, Paczkowska E, Szumilas P, Stecewicz I, Zawodny P, Ziętek M, Wiszniewska B. Global Gene Expression of Cultured Human Dermal Fibroblasts: Focus on Cell Cycle and Proliferation Status in Improving the Condition of Face Skin. Int J Med Sci 2021; 18:1519-1531. [PMID: 33628110 PMCID: PMC7893558 DOI: 10.7150/ijms.46265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Chronological skin ageing is an inevitable physiological process that results in thin and sagging skin, fine wrinkles, and gradual dermal atrophy. The main therapeutic approaches to soft tissue augmentation involve using dermal fillers, where natural fillers, such as autologous fibroblasts, are involved in generating dermal matrix proteins. The aim of this study was to determine the global transcriptome profile of three passages of dermal autologous fibroblasts from a male volunteer, focusing on the processes of the cell cycle and cell proliferation status to estimate the optimal passage of the tested cells with respect to their reimplantation. We performed K-means clustering and validation of the expression of the selected mRNA by qRT-PCR. Ten genes were selected (ANLN, BUB1, CDC20, CCNA2, DLGAP5, MKI67, PLK1, PRC1, SPAG5, and TPX2) from the top five processes annotated to cluster 5. Detailed microarray analysis of the fibroblast genes indicated that the cell population of the third passage exhibited the highest number of upregulated genes involved in the cell cycle and cell proliferation. In all cases, the results of qRT-PCR confirmed the differences in expression of the selected mRNAs between fibroblasts from the primary culture (C0) and from the first (C1), second (C2), and third (C3) cell passage. Our results thus suggest that these cells might be useful for increasing fibroblast numbers after reimplantation into a recipient's skin, and the method used in this study seems to be an excellent tool for autologous transplantation allowing the rejuvenation of aging skin.
Collapse
Affiliation(s)
- Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Aleksandra Wilk
- Department of Histology and Embryology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Kamila Szumilas
- Department of Physiology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Piotr Prowans
- Department of General Pathology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Paweł Szumilas
- Department of Social Medicine and Public Health, Chair of Social Medicine, Pomeranian Medical University, Żołnierska 48, 71-210 Szczecin, Poland
| | - Iwona Stecewicz
- Department of General Pathology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Piotr Zawodny
- Department of General Pathology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Maciej Ziętek
- Department of General Pathology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| | - Barbara Wiszniewska
- Department of Histology and Embryology, Pomeranian Medical University, Powstanców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
38
|
Actin on and around the Nucleus. Trends Cell Biol 2020; 31:211-223. [PMID: 33376040 DOI: 10.1016/j.tcb.2020.11.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022]
Abstract
Actin plays roles in many important cellular processes, including cell motility, organelle movement, and cell signaling. The discovery of transmembrane actin-binding proteins at the outer nuclear membrane (ONM) raises the exciting possibility that actin can play a role in direct force transmission to the nucleus and the genome at its interior. Actin-dependent nucleus displacement was first described a decade ago. We are now gaining a more detailed understanding of its mechanisms, as well as new roles for actin during mitosis and meiosis, for gene expression, and in the cell's response to mechanical stimuli. Here we review these recent developments, the actin-binding proteins involved, the tissue specificity of these mechanisms, and methods developed to reconstitute and study this interaction in vitro.
Collapse
|
39
|
Zucker RM, Ortenzio J, Degn LL, Boyes WK. Detection of large extracellular silver nanoparticle rings observed during mitosis using darkfield microscopy. PLoS One 2020; 15:e0240268. [PMID: 33259485 PMCID: PMC7707489 DOI: 10.1371/journal.pone.0240268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/22/2020] [Indexed: 12/27/2022] Open
Abstract
During studies on the absorption and interactions between silver nanoparticles and mammalian cells grown in vitro it was observed that large extracellular rings of silver nanoparticles were deposited on the microscope slide, many located near post-mitotic cells. Silver nanoparticles (AgNP, 80nm), coated with citrate, were incubated at concentrations of 0.3 to 30 μg/ml with a human-derived culture of retinal pigment epithelial cells (ARPE-19) and observed using darkfield and fluorescent microscopy, 24 h after treatment. Approximately cell-sized extracellular rings of deposited AgNP were observed on the slides among a field of dispersed individual AgNP. The mean diameter of 45 nanoparticles circles was 62.5 +/-12 microns. Ring structures were frequently observed near what appeared to be post-mitotic daughter cells, giving rise to the possibility that cell membrane fragments were deposited on the slide during mitosis, and those fragments selectively attracted and retained silver nanoparticles from suspension in the cell culture medium. These circular structures were observable for the following technical reasons: 1) darkfield microscope could observe single nanoparticles below 100 nm in size, 2) a large concentration (108 and 109) of nanoparticles was used in these experiments 3) negatively charged nanoparticles were attracted to adhesion membrane proteins remaining on the slide from mitosis. The observation of silver nanoparticles attracted to apparent remnants of cellular mitosis could be a useful tool for the study of normal and abnormal mitosis.
Collapse
Affiliation(s)
- Robert M. Zucker
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Public Health and Integrated Toxicology Division, Reproductive and Developmental Toxicology Branch, Research Triangle Park, Durham, NC, United States of America
| | - Jayna Ortenzio
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Public Health and Integrated Toxicology Division, Reproductive and Developmental Toxicology Branch, Research Triangle Park, Durham, NC, United States of America
| | - Laura L. Degn
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Public Health and Integrated Toxicology Division, Reproductive and Developmental Toxicology Branch, Research Triangle Park, Durham, NC, United States of America
| | - William K. Boyes
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Public Health and Integrated Toxicology Division, Reproductive and Developmental Toxicology Branch, Research Triangle Park, Durham, NC, United States of America
| |
Collapse
|
40
|
Effects of Netarsudil on Actin-Driven Cellular Functions in Normal and Glaucomatous Trabecular Meshwork Cells: A Live Imaging Study. J Clin Med 2020; 9:jcm9113524. [PMID: 33142742 PMCID: PMC7693753 DOI: 10.3390/jcm9113524] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023] Open
Abstract
The actin cytoskeleton of trabecular meshwork (TM) cells is a therapeutic target for lowering intraocular pressure (IOP) in glaucoma patients. Netarsudil (the active ingredient in RhopressaTM) is a Rho-associated protein kinase inhibitor that induces disassembly of actin stress fibers. Here, we used live cell imaging of SiR-actin-labeled normal (NTM) and glaucomatous TM (GTM) cells to investigate actin dynamics during actin-driven biological processes with and without netarsudil treatment. Actin stress fibers were thicker in GTM than NTM cells and took longer (>120 min) to disassemble following addition of 1 µM netarsudil. Actin-rich extracellular vesicles (EVs) were derived by two mechanisms: exocytosis of intracellular-derived vesicles, and cleavage of filopodial tips, which detached the filopodia from the substratum, allowing them to retract to the cell body. While some phagocytosis was noted in untreated TM cells, netarsudil potently stimulated phagocytic uptake of EVs. Netarsudil treatment induced lateral fusion of tunneling nanotubes (TNTs) that connected adjacent TM cells; TNTs are important for TM cellular communication. Together, our results suggest that netarsudil may clear outflow channels in TM tissue by inducing phagocytosis and/or by modulating TM communication via EVs and TNTs. These cellular functions likely work together to regulate IOP in normal and glaucomatous TM.
Collapse
|
41
|
Kinase-Independent Functions of MASTL in Cancer: A New Perspective on MASTL Targeting. Cells 2020; 9:cells9071624. [PMID: 32640605 PMCID: PMC7407770 DOI: 10.3390/cells9071624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
Microtubule-associated serine/threonine kinase-like (MASTL; Greatwall) is a well-characterized kinase, whose catalytic role has been extensively studied in relation to cell-cycle acceleration. Importantly, MASTL has been implicated to play a substantial role in cancer progression and subsequent studies have shown that MASTL is a significant regulator of the cellular actomyosin cytoskeleton. Several kinases have non-catalytic properties, which are essential or even sufficient for their functions. Likewise, MASTL functions have been attributed both to kinase-dependent phosphorylation of downstream substrates, but also to kinase-independent regulation of the actomyosin contractile machinery. In this review, we aimed to highlight the catalytic and non-catalytic roles of MASTL in proliferation, migration, and invasion. Further, we discussed the implications of this dual role for therapeutic design.
Collapse
|
42
|
Rainey L, Deevi RK, McClements J, Khawaja H, Watson CJ, Roudier M, Van Schaeybroeck S, Campbell FC. Fundamental control of grade-specific colorectal cancer morphology by Src regulation of ezrin-centrosome engagement. J Pathol 2020; 251:310-322. [PMID: 32315081 DOI: 10.1002/path.5452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/27/2020] [Accepted: 04/07/2020] [Indexed: 11/11/2022]
Abstract
The phenotypic spectrum of colorectal cancer (CRC) is remarkably diverse, with seemingly endless variations in cell shape, mitotic figures and multicellular configurations. Despite this morphological complexity, histological grading of collective phenotype patterns provides robust prognostic stratification in CRC. Although mechanistic understanding is incomplete, previous studies have shown that the cortical protein ezrin controls diversification of cell shape, mitotic figure geometry and multicellular architecture, in 3D organotypic CRC cultures. Because ezrin is a substrate of Src tyrosine kinase that is frequently overexpressed in CRC, we investigated Src regulation of ezrin and morphogenic growth in 3D CRC cultures. Here we show that Src perturbations disrupt CRC epithelial spatial organisation. Aberrant Src activity suppresses formation of the cortical ezrin cap that anchors interphase centrosomes. In CRC cells with a normal centrosome number, these events lead to mitotic spindle misorientation, perturbation of cell cleavage, abnormal epithelial stratification, apical membrane misalignment, multilumen formation and evolution of cribriform multicellular morphology, a feature of low-grade cancer. In isogenic CRC cells with centrosome amplification, aberrant Src signalling promotes multipolar mitotic spindle formation, pleomorphism and morphological features of high-grade cancer. Translational studies in archival human CRC revealed associations between Src intensity, multipolar mitotic spindle frequency and high-grade cancer morphology. Collectively, our study reveals Src regulation of CRC morphogenic growth via ezrin-centrosome engagement and uncovers combined perturbations underlying transition to high-grade CRC morphology. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lisa Rainey
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Ravi K Deevi
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Jane McClements
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Hajrah Khawaja
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Chris J Watson
- Wellcome Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Martine Roudier
- Molecular Pathology Laboratory, AstraZeneca Oncology Translational Science, Cambridge, UK
| | - Sandra Van Schaeybroeck
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Frederick C Campbell
- Centre for Cancer Research and Cell Biology, Queen's University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| |
Collapse
|
43
|
ALK Inhibitors-Induced M Phase Delay Contributes to the Suppression of Cell Proliferation. Cancers (Basel) 2020; 12:cancers12041054. [PMID: 32344689 PMCID: PMC7226408 DOI: 10.3390/cancers12041054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/22/2020] [Indexed: 12/26/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK), a receptor-type tyrosine kinase, is involved in the pathogenesis of several cancers. ALK has been targeted with small molecule inhibitors for the treatment of different cancers, but absolute success remains elusive. In the present study, the effects of ALK inhibitors on M phase progression were evaluated. Crizotinib, ceritinib, and TAE684 suppressed proliferation of neuroblastoma SH-SY5Y cells in a concentration-dependent manner. At approximate IC50 concentrations, these inhibitors caused misorientation of spindles, misalignment of chromosomes and reduction in autophosphorylation. Similarly, knockdown of ALK caused M phase delay, which was rescued by re-expression of ALK. Time-lapse imaging revealed that anaphase onset was delayed. The monopolar spindle 1 (MPS1) inhibitor, AZ3146, and MAD2 knockdown led to a release from inhibitor-induced M phase delay, suggesting that spindle assembly checkpoint may be activated in ALK-inhibited cells. H2228 human lung carcinoma cells that express EML4-ALK fusion showed M phase delay in the presence of TAE684 at about IC50 concentrations. These results suggest that ALK plays a role in M phase regulation and ALK inhibition may contribute to the suppression of cell proliferation in ALK-expressing cancer cells.
Collapse
|
44
|
Wang Y, Stear JH, Swain A, Xu X, Bryce NS, Carnell M, Alieva IB, Dugina VB, Cripe TP, Stehn J, Hardeman EC, Gunning PW. Drug Targeting the Actin Cytoskeleton Potentiates the Cytotoxicity of Low Dose Vincristine by Abrogating Actin-Mediated Repair of Spindle Defects. Mol Cancer Res 2020; 18:1074-1087. [PMID: 32269073 DOI: 10.1158/1541-7786.mcr-19-1122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/09/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022]
Abstract
Antimicrotubule vinca alkaloids are widely used in the clinic but their toxicity is often dose limiting. Strategies that enhance their effectiveness at lower doses are needed. We show that combining vinca alkaloids with compounds that target a specific population of actin filaments containing the cancer-associated tropomyosin Tpm3.1 result in synergy against a broad range of tumor cell types. We discovered that low concentrations of vincristine alone induce supernumerary microtubule asters that form transient multi-polar spindles in early mitosis. Over time these asters can be reconstructed into functional bipolar spindles resulting in cell division and survival. These microtubule asters are organized by the nuclear mitotic apparatus protein (NuMA)-dynein-dynactin complex without involvement of centrosomes. However, anti-Tpm3.1 compounds at nontoxic concentrations inhibit this rescue mechanism resulting in delayed onset of anaphase, formation of multi-polar spindles, and apoptosis during mitosis. These findings indicate that drug targeting actin filaments containing Tpm3.1 potentiates the anticancer activity of low-dose vincristine treatment. IMPLICATIONS: Simultaneously inhibiting Tpm3.1-containing actin filaments and microtubules is a promising strategy to potentiate the anticancer activity of low-dose vincristine.
Collapse
Affiliation(s)
- Yao Wang
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jeffrey H Stear
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Ashleigh Swain
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Xing Xu
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Nicole S Bryce
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Michael Carnell
- Biomedical Imaging Facility, Mark Wainwright Analytical Center, University of New South Wales, Sydney, New South Wales, Australia
| | - Irina B Alieva
- Department of Electron Microscopy, A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vera B Dugina
- Department of Mathematical Methods in Biology, A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Justine Stehn
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Peter W Gunning
- Cellular and Genetic Medicine Unit, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
45
|
Aguilar‐Aragon M, Bonello TT, Bell GP, Fletcher GC, Thompson BJ. Adherens junction remodelling during mitotic rounding of pseudostratified epithelial cells. EMBO Rep 2020; 21:e49700. [PMID: 32030856 PMCID: PMC7132200 DOI: 10.15252/embr.201949700] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/10/2019] [Accepted: 01/15/2020] [Indexed: 12/26/2022] Open
Abstract
Epithelial cells undergo cortical rounding at the onset of mitosis to enable spindle orientation in the plane of the epithelium. In cuboidal epithelia in culture, the adherens junction protein E-cadherin recruits Pins/LGN/GPSM2 and Mud/NuMA to orient the mitotic spindle. In the pseudostratified columnar epithelial cells of Drosophila, septate junctions recruit Mud/NuMA to orient the spindle, while Pins/LGN/GPSM2 is surprisingly dispensable. We show that these pseudostratified epithelial cells downregulate E-cadherin as they round up for mitosis. Preventing cortical rounding by inhibiting Rho-kinase-mediated actomyosin contractility blocks downregulation of E-cadherin during mitosis. Mitotic activation of Rho-kinase depends on the RhoGEF ECT2/Pebble and its binding partners RacGAP1/MgcRacGAP/CYK4/Tum and MKLP1/KIF23/ZEN4/Pav. Cell cycle control of these Rho activators is mediated by the Aurora A and B kinases, which act redundantly during mitotic rounding. Thus, in Drosophila pseudostratified epithelia, disruption of adherens junctions during mitosis necessitates planar spindle orientation by septate junctions to maintain epithelial integrity.
Collapse
Affiliation(s)
| | - Teresa T Bonello
- EMBL AustraliaThe John Curtin School of Medical ResearchThe Australian National UniversityActonACTAustralia
| | - Graham P Bell
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
| | | | - Barry J Thompson
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
- EMBL AustraliaThe John Curtin School of Medical ResearchThe Australian National UniversityActonACTAustralia
| |
Collapse
|
46
|
Pham QL, Tong A, Rodrigues LN, Zhao Y, Surblyte M, Ramos D, Brito J, Rahematpura A, Voronov RS. Ranking migration cue contributions to guiding individual fibroblasts faced with a directional decision in simple microfluidic bifurcations. Integr Biol (Camb) 2020; 11:208-220. [PMID: 31251334 DOI: 10.1093/intbio/zyz018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 04/04/2019] [Accepted: 05/21/2019] [Indexed: 01/02/2023]
Abstract
Directed cell migration in complex micro-environments, such as in vivo pores, is important for predicting locations of artificial tissue growth and optimizing scaffold architectures. Yet, the directional decisions of cells facing multiple physiochemical cues have not been characterized. Hence, we aim to provide a ranking of the relative importance of the following cues to the decision-making of individual fibroblast cells: chemoattractant concentration gradient, channel width, mitosis, and contact-guidance. In this study, bifurcated micro-channels with branches of different widths were created. Fibroblasts were then allowed to travel across these geometries by following a gradient of platelet-derived growth factor-BB (PDGF-BB) established inside the channels. Subsequently, a combination of statistical analysis and image-based diffusion modeling was used to report how the presence of multiple complex migration cues, including cell-cell influences, affect the fibroblast decision-making. It was found that the cells prefer wider channels over a higher chemoattractant gradient when choosing between asymmetric bifurcated branches. Only when the branches were symmetric in width did the gradient become predominant in directing which path the cell will take. Furthermore, when both the gradient and the channels were symmetric, contact guidance became important for guiding the cells in making directional choices. Based on these results we were able to rank these directional cues from most influential to the least as follows: mitosis > channel width asymmetry > chemoattractant gradient difference > and contact-guidance. It is expected that these results will benefit the fields of regenerative medicine, wound healing and developmental biology.
Collapse
Affiliation(s)
- Quang Long Pham
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Anh Tong
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Lydia N Rodrigues
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Yang Zhao
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Migle Surblyte
- Ying Wu College of Computing Sciences, Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Diomar Ramos
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - John Brito
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Adwik Rahematpura
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Roman S Voronov
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| |
Collapse
|
47
|
Garg R, Koo CY, Infante E, Giacomini C, Ridley AJ, Morris JDH. Rnd3 interacts with TAO kinases and contributes to mitotic cell rounding and spindle positioning. J Cell Sci 2020; 133:jcs235895. [PMID: 32041905 DOI: 10.1242/jcs.235895] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/27/2020] [Indexed: 01/22/2023] Open
Abstract
Rnd3 is an atypical Rho family protein that is constitutively GTP bound, and acts on membranes to induce loss of actin stress fibers and cell rounding. Phosphorylation of Rnd3 promotes 14-3-3 binding and its relocation to the cytosol. Here, we show that Rnd3 binds to the thousand-and-one amino acid kinases TAOK1 and TAOK2 in vitro and in cells. TAOK1 and TAOK2 can phosphorylate serine residues 210, 218 and 240 near the C-terminus of Rnd3, and induce Rnd3 translocation from the plasma membrane to the cytosol. TAOKs are activated catalytically during mitosis and Rnd3 phosphorylation on serine 210 increases in dividing cells. Rnd3 depletion by RNAi inhibits mitotic cell rounding and spindle centralization, and delays breakdown of the intercellular bridge between two daughter cells. Our results show that TAOKs bind, phosphorylate and relocate Rnd3 to the cytosol and that Rnd3 contributes to mitotic cell rounding, spindle positioning and cytokinesis. Rnd3 can therefore participate in the regulation of early and late mitosis and may also act downstream of TAOKs to affect the cytoskeleton.
Collapse
Affiliation(s)
- Ritu Garg
- King's College London, School of Cancer and Pharmaceutical Sciences, New Hunt's House, Guy's Campus, London SE1 1UL, UK
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Chuay-Yeng Koo
- King's College London, School of Cancer and Pharmaceutical Sciences, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Elvira Infante
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Caterina Giacomini
- King's College London, School of Cancer and Pharmaceutical Sciences, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Anne J Ridley
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, UK
- School of Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Jonathan D H Morris
- King's College London, School of Cancer and Pharmaceutical Sciences, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
48
|
Winnicki K. The Winner Takes It All: Auxin-The Main Player during Plant Embryogenesis. Cells 2020; 9:E606. [PMID: 32138372 PMCID: PMC7140527 DOI: 10.3390/cells9030606] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 12/11/2022] Open
Abstract
In plants, the first asymmetrical division of a zygote leads to the formation of two cells with different developmental fates. The establishment of various patterns relies on spatial and temporal gene expression, however the precise mechanism responsible for embryonic patterning still needs elucidation. Auxin seems to be the main player which regulates embryo development and controls expression of various genes in a dose-dependent manner. Thus, local auxin maxima and minima which are provided by polar auxin transport underlie cell fate specification. Diverse auxin concentrations in various regions of an embryo would easily explain distinct cell identities, however the question about the mechanism of cellular patterning in cells exposed to similar auxin concentrations still remains open. Thus, specification of cell fate might result not only from the cell position within an embryo but also from events occurring before and during mitosis. This review presents the impact of auxin on the orientation of the cell division plane and discusses the mechanism of auxin-dependent cytoskeleton alignment. Furthermore, close attention is paid to auxin-induced calcium fluxes, which regulate the activity of MAPKs during postembryonic development and which possibly might also underlie cellular patterning during embryogenesis.
Collapse
Affiliation(s)
- Konrad Winnicki
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lódź, Poland
| |
Collapse
|
49
|
Rizzelli F, Malabarba MG, Sigismund S, Mapelli M. The crosstalk between microtubules, actin and membranes shapes cell division. Open Biol 2020; 10:190314. [PMID: 32183618 PMCID: PMC7125961 DOI: 10.1098/rsob.190314] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
Mitotic progression is orchestrated by morphological and mechanical changes promoted by the coordinated activities of the microtubule (MT) cytoskeleton, the actin cytoskeleton and the plasma membrane (PM). MTs assemble the mitotic spindle, which assists sister chromatid separation, and contact the rigid and tensile actomyosin cortex rounded-up underneath the PM. Here, we highlight the dynamic crosstalk between MTs, actin and cell membranes during mitosis, and discuss the molecular connections between them. We also summarize recent views on how MT traction forces, the actomyosin cortex and membrane trafficking contribute to spindle positioning in isolated cells in culture and in epithelial sheets. Finally, we describe the emerging role of membrane trafficking in synchronizing actomyosin tension and cell shape changes with cell-substrate adhesion, cell-cell contacts and extracellular signalling events regulating proliferation.
Collapse
Affiliation(s)
| | - Maria Grazia Malabarba
- IEO, Istituto Europeo di Oncologia IRCCS, Milan, Italy
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy
| | - Sara Sigismund
- IEO, Istituto Europeo di Oncologia IRCCS, Milan, Italy
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
50
|
Targeting Insulin-Like Growth Factor 1 Receptor Delays M-Phase Progression and Synergizes with Aurora B Inhibition to Suppress Cell Proliferation. Int J Mol Sci 2020; 21:ijms21031058. [PMID: 32033461 PMCID: PMC7037296 DOI: 10.3390/ijms21031058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/01/2020] [Indexed: 12/22/2022] Open
Abstract
The insulin-like growth factor 1 receptor (IGF1R) is a receptor-type tyrosine kinase that transduces signals related to cell proliferation, differentiation, and survival. IGF1R expression is often misregulated in tumor cells, but the relevance of this for cancer progression remains unclear. Here, we examined the impact of IGF1R inhibition on cell division. We found that siRNA-mediated knockdown of IGF1R from HeLa S3 cells leads to M-phase delays. Although IGF1R depletion causes partial exclusion of FoxM1 from the nucleus, quantitative real-time PCR revealed that the transcription of M-phase regulators is not affected by decreased levels of IGF1R. Moreover, a similar delay in M phase was observed following 2 h of incubation with the IGF1R inhibitors OSI-906 and NVP-ADW742. These results suggest that the M-phase delay observed in IGF1R-compromised cells is not caused by altered expression of mitotic regulators. Live-cell imaging revealed that both prolonged prometaphase and prolonged metaphase underlie the delay and this can be abrogated by the inhibition of Mps1 with AZ3146, suggesting activation of the Spindle Assembly Checkpoint when IGF1R is inhibited. Furthermore, incubation with the Aurora B inhibitor ZM447439 potentiated the IGF1R inhibitor-induced suppression of cell proliferation, opening up new possibilities for more effective cancer chemotherapy.
Collapse
|