1
|
McLellan MM, Aerne BL, Banerjee Dhoul JJ, Holder MV, Auchynnikava T, Tapon N. Meru co-ordinates spindle orientation with cell polarity and cell cycle progression. EMBO J 2025; 44:2949-2975. [PMID: 40169811 DOI: 10.1038/s44318-025-00420-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
Correct mitotic spindle alignment is essential for tissue architecture and plays an important role in cell fate specification through asymmetric cell division. Spindle tethering factors such as Drosophila Mud (NuMA in mammals) are recruited to the cell cortex and capture astral microtubules, pulling the spindle in the correct orientation. However, how spindle tethering complexes read the cell polarity axis and how spindle attachment is coupled to mitotic progression remains poorly understood. We explore these questions in Drosophila sensory organ precursors (SOPs), which divide asymmetrically to give rise to epidermal mechanosensory bristles. We show that the scaffold protein Meru, which is enriched at the posterior cortex by the Frizzled/Dishevelled planar cell polarity complex, in turn recruits Mud, linking the spindle tethering and polarity machineries. Furthermore, Cyclin A/Cdk1 associates with Meru at the posterior cortex, promoting the formation of the Mud/Meru/Dsh complex via Meru and Dsh phosphorylation. Thus, Meru couples spindle orientation with cell polarity and provides a cell cycle-dependent cue for spindle tethering.
Collapse
Affiliation(s)
- Melissa M McLellan
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Birgit L Aerne
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Jennifer J Banerjee Dhoul
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Maxine V Holder
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Tania Auchynnikava
- Proteomics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Nicolas Tapon
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
2
|
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
|
3
|
Sutton PJ, Mosqueda N, Brownlee CW. Palmitoylated Importin α Regulates Mitotic Spindle Orientation Through Interaction with NuMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.25.620315. [PMID: 39484393 PMCID: PMC11527331 DOI: 10.1101/2024.10.25.620315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Regulation of cell division orientation is a fundamental process critical to differentiation and tissue homeostasis. Microtubules emanating from the mitotic spindle pole bind a conserved complex of proteins at the cell cortex which orients the spindle and ultimately the cell division plane. Control of spindle orientation is of particular importance in developing tissues, such as the developing brain. Misorientation of the mitotic spindle and thus subsequent division plane misalignment can contribute to improper segregation of cell fate determinants in developing neuroblasts, leading to a rare neurological disorder known as microcephaly. We demonstrate that the nuclear transport protein importin α, when palmitoylated, plays a critical role in mitotic spindle orientation through localizing factors, such as NuMA, to the cell cortex. We also observe craniofacial developmental defects in Xenopus laevis when importin α palmitoylation is abrogated, including smaller head and brains, a hallmark of spindle misorientation and microcephaly. These findings characterize not only a role for importin α in spindle orientation, but also a broader role for importin α palmitoylation which has significance for many cellular processes.
Collapse
|
4
|
Iannitti R, Mascanzoni F, Colanzi A, Spano D. The role of Golgi complex proteins in cell division and consequences of their dysregulation. Front Cell Dev Biol 2025; 12:1513472. [PMID: 39839669 PMCID: PMC11747491 DOI: 10.3389/fcell.2024.1513472] [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: 10/18/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
The GC (Golgi complex) plays a pivotal role in the trafficking and sorting of proteins and lipids until they reach their final destination. Additionally, the GC acts as a signalling hub to regulate a multitude of cellular processes, including cell polarity, motility, apoptosis, DNA repair and cell division. In light of these crucial roles, the GC has garnered increasing attention, particularly given the evidence that a dysregulation of GC-regulated signalling pathways may contribute to the onset of various pathological conditions. This review examines the functions of the GC and GC-localised proteins in regulating cell cycle progression, in both mitosis and meiosis. It reviews the involvement of GC-resident proteins in the formation and orientation of the spindle during cell division. In light of the roles played by the GC in controlling cell division, this review also addresses the involvement of the GC in cancer development. Furthermore, TCGA (The Cancer Genome Atlas) database has been queried in order to retrieve information on the genetic alterations and the correlation between the expression of GC-localised proteins and the survival of cancer patients. The data presented in this review highlight the relevance of the GC in regulating cell cycle progression, cellular differentiation and tumourigenesis.
Collapse
Affiliation(s)
| | | | | | - Daniela Spano
- Department of Biomedical Sciences (DSB), Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
5
|
Huang S, Fu M, Gu A, Zhao R, Liu Z, Hua W, Mao Y, Wen W. mInsc coordinates Par3 and NuMA condensates for assembly of the spindle orientation machinery in asymmetric cell division. Int J Biol Macromol 2024; 279:135126. [PMID: 39218187 DOI: 10.1016/j.ijbiomac.2024.135126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
As a fundamental process governing the self-renewal and differentiation of stem cells, asymmetric cell division is controlled by several conserved regulators, including the polarity protein Par3 and the microtubule-associated protein NuMA, which orchestrate the assembly and interplay of the Par3/Par6/mInsc/LGN complex at the apical cortex and the LGN/Gαi/NuMA/Dynein complex at the mitotic spindle to ensure asymmetric segregation of cell fate determinants. However, this model, which is well-supported by genetic studies, has been challenged by evidence of competitive interaction between NuMA and mInsc for LGN. Here, the solved crystal structure of the Par3/mInsc complex reveals that mInsc competes with Par6β for Par3, raising questions about how proteins assemble overlapping targets into functional macromolecular complexes. Unanticipatedly, we discover that Par3 can recruit both Par6β and mInsc by forming a dynamic condensate through phase separation. Similarly, the phase-separated NuMA condensate enables the coexistence of competitive NuMA and mInsc with LGN in the same compartment. Bridge by mInsc, Par3/Par6β and LGN/NuMA condensates coacervate, robustly enriching all five proteins both in vitro and within cells. These findings highlight the pivotal role of protein condensates in assembling multi-component signalosomes that incorporate competitive protein-protein interaction pairs, effectively overcoming stoichiometric constraints encountered in conventional protein complexes.
Collapse
Affiliation(s)
- Shijing Huang
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Aihong Gu
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ruiqian Zhao
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ziheng Liu
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wenyu Wen
- Department of Neurosurgery, Huashan Hospital, The Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
6
|
Wu Y, Lan Y, Ononiwu F, Poole A, Rasmussen K, Da Silva J, Shamil AW, Jao LE, Hehnly H. Specific Mitotic Events Drive Cytoskeletal Remodeling Required for Left-Right Organizer Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593765. [PMID: 38798489 PMCID: PMC11118341 DOI: 10.1101/2024.05.12.593765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Cellular proliferation is vital for tissue development, including the Left-Right Organizer (LRO), a transient organ critical for establishing the vertebrate LR body plan. This study investigates cell redistribution and the role of specific progenitor cells in LRO formation, focusing on cell lineage and behavior. Using zebrafish as a model, we mapped all mitotic events in Kupffer's Vesicle (KV), revealing an FGF-dependent, anteriorly enriched mitotic pattern. With a KV-specific fluorescent microtubule (MT) line, we observed that mitotic spindles align along the KV's longest axis until the rosette stage, spindles that form after spin, and are excluded from KV. Early aligned spindles assemble cytokinetic bridges that point MT bundles toward a tight junction where a rosette will initially form. Post-abscission, repurposed MT bundles remain targeted at the rosette center, facilitating actin recruitment. Additional cells, both cytokinetic and non-cytokinetic, are incorporated into the rosette, repurposing or assembling MT bundles before actin recruitment. These findings show that initial divisions are crucial for rosette assembly, MT patterning, and actin remodeling during KV development.
Collapse
Affiliation(s)
- Yan Wu
- Department of Biology, Syracuse University, Syracuse, 13244 USA
- BioInspired Institute, Syracuse University, Syracuse, 13244 USA
| | - Yiling Lan
- Department of Biology, Syracuse University, Syracuse, 13244 USA
- BioInspired Institute, Syracuse University, Syracuse, 13244 USA
| | - Favour Ononiwu
- Department of Biology, Syracuse University, Syracuse, 13244 USA
- BioInspired Institute, Syracuse University, Syracuse, 13244 USA
| | - Abigail Poole
- Worcester Polytechnic Institute, Worcester, 01609 MA
| | | | - Jonah Da Silva
- Department of Biology, Syracuse University, Syracuse, 13244 USA
| | | | - Li-En Jao
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Sacramento, 95817 USA
| | - Heidi Hehnly
- Department of Biology, Syracuse University, Syracuse, 13244 USA
- BioInspired Institute, Syracuse University, Syracuse, 13244 USA
| |
Collapse
|
7
|
Finegan TM, Cammarota C, Mendoza Andrade O, Garoutte AM, Bergstralh DT. Fas2EB112: a tale of two chromosomes. G3 (BETHESDA, MD.) 2024; 14:jkae047. [PMID: 38447284 PMCID: PMC11075550 DOI: 10.1093/g3journal/jkae047] [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: 02/08/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
The cell-cell adhesion molecule Fasciclin II (Fas2) has long been studied for its evolutionarily conserved role in axon guidance. It is also expressed in the follicular epithelium, where together with a similar protein, Neuroglian (Nrg), it helps to drive the reintegration of cells born out of the tissue plane. Remarkably, one Fas2 protein null allele, Fas2G0336, demonstrates a mild reintegration phenotype, whereas work with the classic null allele Fas2EB112 showed more severe epithelial disorganization. These observations raise the question of which allele (if either) causes a bona fide loss of Fas2 protein function. The problem is not only relevant to reintegration but fundamentally important to understanding what this protein does and how it works: Fas2EB112 has been used in at least 37 research articles, and Fas2G0336 in at least three. An obvious solution is that one of the two chromosomes carries a modifier that either suppresses (Fas2G0336) or enhances (Fas2EB112) phenotypic severity. We find not only the latter to be the case, but identify the enhancing mutation as Nrg14, also a classic null allele.
Collapse
Affiliation(s)
- Tara M Finegan
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
- Division of Biological Sciences, University of Missouri, Columbia, MO 65203, USA
| | - Christian Cammarota
- Department of Physics and Astronomy, University of Rochester, Rochester, NY 14627, USA
| | | | - Audrey M Garoutte
- Division of Biological Sciences, University of Missouri, Columbia, MO 65203, USA
| | - Dan T Bergstralh
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
- Division of Biological Sciences, University of Missouri, Columbia, MO 65203, USA
- Department of Physics and Astronomy, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
8
|
Finegan TM, Cammarota C, Andrade OM, Garoutte AM, Bergstralh DT. Fas2EB112: A Tale of Two Chromosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.574100. [PMID: 38260405 PMCID: PMC10802346 DOI: 10.1101/2024.01.03.574100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The cell-cell adhesion molecule Fasciclin II (Fas2) has long been studied for its evolutionarily-conserved role in axon guidance. It is also expressed in the follicular epithelium, where together with a similar protein, Neuroglian (Nrg), it helps to drive the reintegration of cells born out of the tissue plane. Remarkably, one Fas2 protein null allele, Fas2G0336, demonstrates a mild reintegration phenotype, whereas work with the classic null allele Fas2EB112 showed more severe epithelial disorganization. These observations raise the question of which allele (if either) causes a bona fide loss of Fas2 protein function. The problem is not only relevant to reintegration but fundamentally important to understanding what this protein does and how it works: Fas2EB112 has been used in at least 37 research articles, and Fas2G0336 in at least three. An obvious solution is that one of the two chromosomes carries a modifier that either suppresses (Fas2G0336) or enhances (Fas2EB112) phenotypic severity. We find not only the latter to be the case, but identify the enhancing mutation as Nrg14, also a classic null allele.
Collapse
Affiliation(s)
- Tara M Finegan
- Departments of Biology, University of Rochester, Rochester NY, 14627, USA
- Division of Biological Sciences, University of Missouri, Columbia, MO, 65203, USA
| | - Christian Cammarota
- Departments of Physics & Astronomy, University of Rochester, Rochester NY, 14627, USA
| | | | - Audrey M Garoutte
- Division of Biological Sciences, University of Missouri, Columbia, MO, 65203, USA
| | - Dan T Bergstralh
- Departments of Biology, University of Rochester, Rochester NY, 14627, USA
- Departments of Physics & Astronomy, University of Rochester, Rochester NY, 14627, USA
- Division of Biological Sciences, University of Missouri, Columbia, MO, 65203, USA
| |
Collapse
|
9
|
Burda I, Martin AC, Roeder AHK, Collins MA. The dynamics and biophysics of shape formation: Common themes in plant and animal morphogenesis. Dev Cell 2023; 58:2850-2866. [PMID: 38113851 PMCID: PMC10752614 DOI: 10.1016/j.devcel.2023.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/19/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The emergence of tissue form in multicellular organisms results from the complex interplay between genetics and physics. In both plants and animals, cells must act in concert to pattern their behaviors. Our understanding of the factors sculpting multicellular form has increased dramatically in the past few decades. From this work, common themes have emerged that connect plant and animal morphogenesis-an exciting connection that solidifies our understanding of the developmental basis of multicellular life. In this review, we will discuss the themes and the underlying principles that connect plant and animal morphogenesis, including the coordination of gene expression, signaling, growth, contraction, and mechanical and geometric feedback.
Collapse
Affiliation(s)
- Isabella Burda
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Genetic Genomics and Development Program, Cornell University, Ithaca, NY 14853, USA
| | - Adam C Martin
- Biology Department, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Adrienne H K Roeder
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Genetic Genomics and Development Program, Cornell University, Ithaca, NY 14853, USA; School of Integrative Plant Sciences, Section of Plant Biology, Cornell University, Ithaca, NY 14850, USA.
| | - Mary Ann Collins
- Biology Department, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
10
|
Faienza F, Polverino F, Rajendraprasad G, Milletti G, Hu Z, Colella B, Gargano D, Strappazzon F, Rizza S, Vistesen MV, Luo Y, Antonioli M, Cianfanelli V, Ferraina C, Fimia GM, Filomeni G, De Zio D, Dengjel J, Barisic M, Guarguaglini G, Di Bartolomeo S, Cecconi F. AMBRA1 phosphorylation by CDK1 and PLK1 regulates mitotic spindle orientation. Cell Mol Life Sci 2023; 80:251. [PMID: 37584777 PMCID: PMC10432340 DOI: 10.1007/s00018-023-04878-6] [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: 12/31/2022] [Revised: 06/27/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023]
Abstract
AMBRA1 is a crucial factor for nervous system development, and its function has been mainly associated with autophagy. It has been also linked to cell proliferation control, through its ability to regulate c-Myc and D-type cyclins protein levels, thus regulating G1-S transition. However, it remains still unknown whether AMBRA1 is differentially regulated during the cell cycle, and if this pro-autophagy protein exerts a direct role in controlling mitosis too. Here we show that AMBRA1 is phosphorylated during mitosis on multiple sites by CDK1 and PLK1, two mitotic kinases. Moreover, we demonstrate that AMBRA1 phosphorylation at mitosis is required for a proper spindle function and orientation, driven by NUMA1 protein. Indeed, we show that the localization and/or dynamics of NUMA1 are strictly dependent on AMBRA1 presence, phosphorylation and binding ability. Since spindle orientation is critical for tissue morphogenesis and differentiation, our findings could account for an additional role of AMBRA1 in development and cancer ontogenesis.
Collapse
Affiliation(s)
- Fiorella Faienza
- Cell Stress and Survival Group, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Institute, Copenhagen, Denmark
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Federica Polverino
- Institute of Molecular Biology and Pathology, CNR National Research Council, Rome, Italy
| | | | - Giacomo Milletti
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- DNA Replication and Cancer Group, Danish Cancer Institute, 2100, Copenhagen, Denmark
| | - Zehan Hu
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Barbara Colella
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Deborah Gargano
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Flavie Strappazzon
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyogène, Univ Lyon, Univ Lyon 1, CNRS, INSERM, 69008, Lyon, France
| | - Salvatore Rizza
- Redox Biology Group, Danish Cancer Institute, Copenhagen, Denmark
| | - Mette Vixø Vistesen
- Cell Stress and Survival Group, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Institute, Copenhagen, Denmark
| | - Yonglun Luo
- Lars Bolund Institute of Regenerative Medicine and Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Shenzhen, China
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Manuela Antonioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- National Institute for Infectious Diseases, IRCSS "L. Spallanzani", Rome, Italy
| | - Valentina Cianfanelli
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Science, University "ROMA TRE", 00146, Rome, Italy
- Department of Woman and Child Health and Public Health, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Caterina Ferraina
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gian Maria Fimia
- National Institute for Infectious Diseases, IRCSS "L. Spallanzani", Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Redox Biology Group, Danish Cancer Institute, Copenhagen, Denmark
- Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Daniela De Zio
- Melanoma Research Team, Danish Cancer Institute, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University Of Copenhagen, Copenhagen, Denmark
| | - Joern Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Institute, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, CNR National Research Council, Rome, Italy
| | | | - Francesco Cecconi
- Cell Stress and Survival Group, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Institute, Copenhagen, Denmark.
- Università Cattolica del Sacro Cuore and Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
11
|
Ganguli S, Wyatt T, Nyga A, Lawson RH, Meyer T, Baum B, Matthews HK. Oncogenic Ras deregulates cell-substrate interactions during mitotic rounding and respreading to alter cell division orientation. Curr Biol 2023; 33:2728-2741.e3. [PMID: 37343559 PMCID: PMC7614879 DOI: 10.1016/j.cub.2023.05.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/21/2023] [Accepted: 05/25/2023] [Indexed: 06/23/2023]
Abstract
Oncogenic Ras has been shown to change the way cancer cells divide by increasing the forces generated during mitotic rounding. In this way, RasV12 enables cancer cells to divide across a wider range of mechanical environments than normal cells. Here, we identify a further role for oncogenic Ras-ERK signaling in division by showing that RasV12 expression alters the shape, division orientation, and respreading dynamics of cells as they exit mitosis. Many of these effects appear to result from the impact of RasV12 signaling on actomyosin contractility, because RasV12 induces the severing of retraction fibers that normally guide spindle positioning and provide a memory of the interphase cell shape. In support of this idea, the RasV12 phenotype is reversed by inhibition of actomyosin contractility and can be mimicked by the loss of cell-substrate adhesion during mitosis. Finally, we show that RasV12 activation also perturbs division orientation in cells cultured in 2D epithelial monolayers and 3D spheroids. Thus, the induction of oncogenic Ras-ERK signaling leads to rapid changes in division orientation that, along with the effects of RasV12 on cell growth and cell-cycle progression, are likely to disrupt epithelial tissue organization and contribute to cancer dissemination.
Collapse
Affiliation(s)
- Sushila Ganguli
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Tom Wyatt
- Laboratoirè Matiere et Systèmes Complexes, Université Paris Diderot, 10 rue Alice Domon et Léonie Duquet, Bâtiment Condorcet, 75013 Paris, France
| | - Agata Nyga
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Rachel H Lawson
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Tim Meyer
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Buzz Baum
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - Helen K Matthews
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| |
Collapse
|
12
|
Zheng H, Wen W. Protein phase separation: new insights into cell division. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1042-1051. [PMID: 37249333 PMCID: PMC10415187 DOI: 10.3724/abbs.2023093] [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: 12/31/2022] [Accepted: 02/15/2023] [Indexed: 05/31/2023] Open
Abstract
As the foundation for the development of multicellular organisms and the self-renewal of single cells, cell division is a highly organized event which segregates cellular components into two daughter cells equally or unequally, thus producing daughters with identical or distinct fates. Liquid-liquid phase separation (LLPS), an emerging biophysical concept, provides a new perspective for us to understand the mechanisms of a wide range of cellular events, including the organization of membrane-less organelles. Recent studies have shown that several key organelles in the cell division process are assembled into membrane-free structures via LLPS of specific proteins. Here, we summarize the regulatory functions of protein phase separation in centrosome maturation, spindle assembly and polarity establishment during cell division.
Collapse
Affiliation(s)
- Hongdan Zheng
- />Department of NeurosurgeryHuashan Hospitalthe Shanghai Key Laboratory of Medical EpigeneticsState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceNational Center for Neurological DisordersInstitutes of Biomedical SciencesSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Wenyu Wen
- />Department of NeurosurgeryHuashan Hospitalthe Shanghai Key Laboratory of Medical EpigeneticsState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceNational Center for Neurological DisordersInstitutes of Biomedical SciencesSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| |
Collapse
|
13
|
Dawney NS, Cammarota C, Jia Q, Shipley A, Glichowski JA, Vasandani M, Finegan TM, Bergstralh DT. A novel tool for the unbiased characterization of epithelial monolayer development in culture. Mol Biol Cell 2023; 34:ar25. [PMID: 36696175 PMCID: PMC10092640 DOI: 10.1091/mbc.e22-04-0121] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 12/09/2022] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
The function of an epithelial tissue is intertwined with its architecture. Epithelial tissues are often described as pseudo-two-dimensional, but this view may be partly attributed to experimental bias: many model epithelia, including cultured cell lines, are easiest to image from the "top-down." We measured the three-dimensional architecture of epithelial cells in culture and found that it varies dramatically across cultured regions, presenting a challenge for reproducibility and cross-study comparisons. We therefore developed a novel tool (Automated Layer Analysis, "ALAn") to characterize architecture in an unbiased manner. Using ALAn, we find that cultured epithelial cells can organize into four distinct architectures and that architecture correlates with cell density. Cells exhibit distinct biological properties in each architecture. Organization in the apical-basal axis is determined early in monolayer development by substrate availability, while disorganization in the apical-basal axis arises from an inability to form substrate connections. Our work highlights the need to carefully control for three-dimensional architecture when using cell culture as a model system for epithelial cell biology and introduces a novel tool, built on a set of rules that can be widely applied to epithelial cell culture.
Collapse
Affiliation(s)
- Nicole S. Dawney
- Department of Biology, University of Rochester, Rochester, NY 14627
| | - Christian Cammarota
- Department of Physics & Astronomy, University of Rochester, Rochester, NY 14627
| | - Qingyuan Jia
- Department of Biology, University of Rochester, Rochester, NY 14627
| | - Alicia Shipley
- Department of Biology, University of Rochester, Rochester, NY 14627
| | | | | | - Tara M. Finegan
- Department of Biology, University of Rochester, Rochester, NY 14627
| | - Dan T. Bergstralh
- Department of Biology, University of Rochester, Rochester, NY 14627
- Department of Physics & Astronomy, University of Rochester, Rochester, NY 14627
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14627
| |
Collapse
|
14
|
Neville KE, Finegan TM, Lowe N, Bellomio PM, Na D, Bergstralh DT. The Drosophila mitotic spindle orientation machinery requires activation, not just localization. EMBO Rep 2023; 24:e56074. [PMID: 36629398 PMCID: PMC9986814 DOI: 10.15252/embr.202256074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
The orientation of the mitotic spindle at metaphase determines the placement of the daughter cells. Spindle orientation in animals typically relies on an evolutionarily conserved biological machine comprised of at least four proteins - called Pins, Gαi, Mud, and Dynein in flies - that exerts a pulling force on astral microtubules and reels the spindle into alignment. The canonical model for spindle orientation holds that the direction of pulling is determined by asymmetric placement of this machinery at the cell cortex. In most cell types, this placement is thought to be mediated by Pins, and a substantial body of literature is therefore devoted to identifying polarized cues that govern localized cortical enrichment of Pins. In this study we revisit the canonical model and find that it is incomplete. Spindle orientation in the Drosophila follicular epithelium and embryonic ectoderm requires not only Pins localization but also direct interaction between Pins and the multifunctional protein Discs large. This requirement can be over-ridden by interaction with another Pins interacting protein, Inscuteable.
Collapse
Affiliation(s)
| | - Tara M Finegan
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Nicholas Lowe
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | | | - Daxiang Na
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Dan T Bergstralh
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
- Department of Physics & AstronomyUniversity of RochesterRochesterNew YorkUSA
- Department of Biomedical GeneticsUniversity of Rochester Medical CenterRochesterNew YorkUSA
| |
Collapse
|
15
|
Guevara-Garcia A, Soleilhac M, Minc N, Delacour D. Regulation and functions of cell division in the intestinal tissue. Semin Cell Dev Biol 2023:S1084-9521(23)00004-6. [PMID: 36702722 DOI: 10.1016/j.semcdb.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/16/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023]
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing epithelial tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs to ensure correct organ formation and functioning. In these processes, proliferation rates and division orientation regulate the speed, timing and direction of tissue expansion but also its proper patterning. Moreover, tissue homeostasis relies on spatio-temporal modulations of daughter cell behavior and arrangement. These aspects are particularly crucial in the intestine, which is one of the most proliferative tissues in adults, making it a very attractive adult organ system to study the role of cell division on epithelial morphogenesis and organ function. Although epithelial cell division has been the subject of intense research for many years in multiple models, it still remains in its infancy in the context of the intestinal tissue. In this review, we focus on the current knowledge on cell division and regulatory mechanisms at play in the intestinal epithelial tissue, as well as their importance in developmental biology and physiopathology.
Collapse
Affiliation(s)
| | - Matis Soleilhac
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Nicolas Minc
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Delphine Delacour
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France.
| |
Collapse
|
16
|
Chann AS, Chen Y, Kinwel T, Humbert PO, Russell SM. Scribble and E-cadherin cooperate to control symmetric daughter cell positioning by multiple mechanisms. J Cell Sci 2023; 136:286705. [PMID: 36661138 DOI: 10.1242/jcs.260547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/25/2022] [Indexed: 01/21/2023] Open
Abstract
The fate of the two daughter cells is intimately connected to their positioning, which is in turn regulated by cell junction remodelling and orientation of the mitotic spindle. How multiple cues are integrated to dictate the ultimate positioning of daughters is not clear. Here, we identify novel mechanisms of regulation of daughter positioning in single MCF10A cells. The polarity protein, Scribble cooperates with E-cadherin for sequential roles in daughter positioning. First Scribble stabilises E-cadherin at the mitotic cortex as well as the retraction fibres, to mediate spindle orientation. Second, Scribble re-locates to the junction between the two daughters to allow a new E-cadherin-based-interface to form between them, influencing the width of the nascent daughter-daughter junction and subsequent cell positioning. Thus, E-cadherin and Scribble dynamically relocate to different intracellular sites during cell division to orient the mitotic spindle and control placement of the daughter cells after cell division. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Anchi S Chann
- Optical Sciences Centre, School of Science, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia.,Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000Australia
| | - Ye Chen
- Optical Sciences Centre, School of Science, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia.,Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000Australia
| | - Tanja Kinwel
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Patrick O Humbert
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia.,Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, Victoria 3086, Australia.,Department of Biochemistry & Pharmacology, University of Melbourne, Melbourne, Victoria 3010, Australia.,Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Sarah M Russell
- Optical Sciences Centre, School of Science, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia.,Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
17
|
Fankhaenel M, Hashemi FSG, Mourao L, Lucas E, Hosawi MM, Skipp P, Morin X, Scheele CLGJ, Elias S. Annexin A1 is a polarity cue that directs mitotic spindle orientation during mammalian epithelial morphogenesis. Nat Commun 2023; 14:151. [PMID: 36631478 PMCID: PMC9834401 DOI: 10.1038/s41467-023-35881-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Oriented cell divisions are critical for the formation and maintenance of structured epithelia. Proper mitotic spindle orientation relies on polarised anchoring of force generators to the cell cortex by the evolutionarily conserved protein complex formed by the Gαi subunit of heterotrimeric G proteins, the Leucine-Glycine-Asparagine repeat protein (LGN) and the nuclear mitotic apparatus protein. However, the polarity cues that control cortical patterning of this ternary complex remain largely unknown in mammalian epithelia. Here we identify the membrane-associated protein Annexin A1 (ANXA1) as an interactor of LGN in mammary epithelial cells. Annexin A1 acts independently of Gαi to instruct the accumulation of LGN and nuclear mitotic apparatus protein at the lateral cortex to ensure cortical anchoring of Dynein-Dynactin and astral microtubules and thereby planar alignment of the mitotic spindle. Loss of Annexin A1 randomises mitotic spindle orientation, which in turn disrupts epithelial architecture and luminogenesis in three-dimensional cultures of primary mammary epithelial cells. Our findings establish Annexin A1 as an upstream cortical cue that regulates LGN to direct planar cell divisions during mammalian epithelial morphogenesis.
Collapse
Affiliation(s)
- Maria Fankhaenel
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Farahnaz S Golestan Hashemi
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Larissa Mourao
- VIB-KULeuven Center for Cancer Biology, Herestraat 49, 3000, Leuven, Belgium
| | - Emily Lucas
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Manal M Hosawi
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Paul Skipp
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Centre for Proteomic Research, University of Southampton, Southampton, SO17 1BJ, UK
| | - Xavier Morin
- Ecole Normale Supérieure, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), PSL Research University, Paris, France
| | | | - Salah Elias
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK. .,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
18
|
De la Cruz G, Nikolaishvili Feinberg N, Williams SE. Automated Immunofluorescence Staining for Analysis of Mitotic Stages and Division Orientation in Brain Sections. Methods Mol Biol 2023; 2583:63-79. [PMID: 36418726 DOI: 10.1007/978-1-0716-2752-5_7] [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] [Indexed: 06/16/2023]
Abstract
Microcephaly often results from mitotic defects in neuronal progenitors, frequently by decreasing proliferation rates or shifting cell fates. During neurogenesis, oriented cell division-the molecular control of mitotic spindle positioning to control the axis of division-represents an important mechanism to balance expansion of the progenitor pool with generating cellular diversity. While mostly studied in the context of cortical development, more recently, spindle orientation has emerged as a key player in the formation of other brain regions such as the cerebellum. Here we describe methods to perform automated dual-color fluorescent immunohistochemistry on murine cerebellar sections using the mitotic markers phospho-Histone H3 and Survivin, and detail analytical and statistical approaches to display and compare division orientation datasets.
Collapse
Affiliation(s)
- Gabriela De la Cruz
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Pathology Services Core, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nana Nikolaishvili Feinberg
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Pathology Services Core, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott E Williams
- Department of Pathology & Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
19
|
Abstract
A signature feature of the animal kingdom is the presence of epithelia: sheets of polarized cells that both insulate the organism from its environment and mediate interactions with it. Epithelial cells display a marked apico-basal polarity, which is highly conserved across the animal kingdom, both in terms of morphology and of molecular regulators. How did this architecture first evolve? Although the last eukaryotic common ancestor almost certainly possessed a simple form of apico-basal polarity (marked by the presence of one or several flagella at a single cellular pole), comparative genomics and evolutionary cell biology reveal that the polarity regulators of animal epithelial cells have a surprisingly complex and stepwise evolutionary history. Here, we retrace their evolutionary assembly. We suggest that the "polarity network" that polarized animal epithelial cells evolved by integration of initially independent cellular modules that evolved at distinct steps of our evolutionary ancestry. The first module dates back to the last common ancestor of animals and amoebozoans and involved Par1, extracellular matrix proteins, and the integrin-mediated adhesion complex. Other regulators, such as Cdc42, Dlg, Par6 and cadherins evolved in ancient unicellular opisthokonts, and might have first been involved in F-actin remodeling and filopodial dynamics. Finally, the bulk of "polarity proteins" as well as specialized adhesion complexes evolved in the metazoan stem-line, in concert with the newly evolved intercellular junctional belts. Thus, the polarized architecture of epithelia can be understood as a palimpsest of components of distinct histories and ancestral functions, which have become tightly integrated in animal tissues.
Collapse
|
20
|
Wright BA, Kvansakul M, Schierwater B, Humbert PO. Cell polarity signalling at the birth of multicellularity: What can we learn from the first animals. Front Cell Dev Biol 2022; 10:1024489. [PMID: 36506100 PMCID: PMC9729800 DOI: 10.3389/fcell.2022.1024489] [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: 08/21/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
The innovation of multicellularity has driven the unparalleled evolution of animals (Metazoa). But how is a multicellular organism formed and how is its architecture maintained faithfully? The defining properties and rules required for the establishment of the architecture of multicellular organisms include the development of adhesive cell interactions, orientation of division axis, and the ability to reposition daughter cells over long distances. Central to all these properties is the ability to generate asymmetry (polarity), coordinated by a highly conserved set of proteins known as cell polarity regulators. The cell polarity complexes, Scribble, Par and Crumbs, are considered to be a metazoan innovation with apicobasal polarity and adherens junctions both believed to be present in all animals. A better understanding of the fundamental mechanisms regulating cell polarity and tissue architecture should provide key insights into the development and regeneration of all animals including humans. Here we review what is currently known about cell polarity and its control in the most basal metazoans, and how these first examples of multicellular life can inform us about the core mechanisms of tissue organisation and repair, and ultimately diseases of tissue organisation, such as cancer.
Collapse
Affiliation(s)
- Bree A. Wright
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Marc Kvansakul
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia,Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, VIC, Australia
| | - Bernd Schierwater
- Institute of Animal Ecology and Evolution, University of Veterinary Medicine Hannover, Foundation, Bünteweg, Hannover, Germany
| | - Patrick O. Humbert
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia,Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, VIC, Australia,Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC, Australia,Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia,*Correspondence: Patrick O. Humbert,
| |
Collapse
|
21
|
Zucca F, Visintin C, Li J, Gygi SP, Visintin R. APC/CCdc20-mediated degradation of Clb4 prompts astral microtubule stabilization at anaphase onset. J Cell Biol 2022; 222:213563. [PMID: 36269172 PMCID: PMC9595209 DOI: 10.1083/jcb.202203089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/12/2022] [Accepted: 10/05/2022] [Indexed: 11/22/2022] Open
Abstract
Key for accurate chromosome partitioning to the offspring is the ability of mitotic spindle microtubules to respond to different molecular signals and remodel their dynamics accordingly. Spindle microtubules are conventionally divided into three classes: kinetochore, interpolar, and astral microtubules (kMTs, iMTs, and aMTs, respectively). Among all, aMT regulation remains elusive. Here, we show that aMT dynamics are tightly regulated. aMTs remain unstable up to metaphase and are stabilized at anaphase onset. This switch in aMT dynamics, important for proper spindle orientation, specifically requires the degradation of the mitotic cyclin Clb4 by the Anaphase Promoting Complex bound to its activator subunit Cdc20 (APC/CCdc20). These data highlight a unique role for mitotic cyclin Clb4 in controlling aMT regulating factors, of which Kip2 is a prime candidate, provide a framework to understand aMT regulation in vertebrates, and uncover mechanistic principles of how the APC/CCdc20 choreographs the timing of late mitotic events by sequentially impacting on the three classes of spindle microtubules.
Collapse
Affiliation(s)
- Federico Zucca
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Clara Visintin
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Jiaming Li
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Rosella Visintin
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy,Correspondence to Rosella Visintin:
| |
Collapse
|
22
|
Sana S, Rajeevan A, Kotak S. Membrane compartmentalization of Ect2/Cyk4/Mklp1 and NuMA/dynein regulates cleavage furrow formation. J Biophys Biochem Cytol 2022; 221:213522. [PMID: 36197340 PMCID: PMC9539458 DOI: 10.1083/jcb.202203127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/09/2022] [Accepted: 09/02/2022] [Indexed: 12/13/2022] Open
Abstract
In animal cells, spindle elongation during anaphase is temporally coupled with cleavage furrow formation. Spindle elongation during anaphase is regulated by NuMA/dynein/dynactin complexes that occupy the polar region of the cell membrane and are excluded from the equatorial membrane. How NuMA/dynein/dynactin are excluded from the equatorial membrane and the biological significance of this exclusion remains unknown. Here, we show that the centralspindlin (Cyk4/Mklp1) and its interacting partner RhoGEF Ect2 are required for NuMA/dynein/dynactin exclusion from the equatorial cell membrane. The Ect2-based (Ect2/Cyk4/Mklp1) and NuMA-based (NuMA/dynein/dynactin) complexes occupy mutually exclusive membrane surfaces during anaphase. The equatorial membrane enrichment of Ect2-based complexes is essential for NuMA/dynein/dynactin exclusion and proper spindle elongation. Conversely, NuMA-based complexes at the polar region of the cell membrane ensure spatially confined localization of Ect2-based complexes and thus RhoA. Overall, our work establishes that membrane compartmentalization of NuMA-based and Ect2-based complexes at the two distinct cell surfaces restricts dynein/dynactin and RhoA for coordinating spindle elongation with cleavage furrow formation.
Collapse
Affiliation(s)
- Shrividya Sana
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), Bangalore, India
| | - Ashwathi Rajeevan
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), Bangalore, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology (MCB), Indian Institute of Science (IISc), Bangalore, India,Correspondence to Sachin Kotak:
| |
Collapse
|
23
|
Zhong T, Gongye X, Wang M, Yu J. Understanding the underlying mechanisms governing spindle orientation: How far are we from there? J Cell Mol Med 2022; 26:4904-4910. [PMID: 36029193 PMCID: PMC9549511 DOI: 10.1111/jcmm.17526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Proper spindle orientation is essential for cell fate determination and tissue morphogenesis. Recently, accumulating studies have elucidated several factors that regulate spindle orientation, including geometric, internal and external cues. Abnormality in these factors generally leads to defects in the physiological functions of various organs and the development of severe diseases. Herein, we first review models that are commonly used for studying spindle orientation. We then review a conservative heterotrimeric complex critically involved in spindle orientation regulation in different models. Finally, we summarize some cues that affect spindle orientation and explore whether we can establish a model that precisely elucidates the effects of spindle orientation without interfusing other spindle functions. We aim to summarize current models used in spindle orientation studies and discuss whether we can build a model that disturbs spindle orientation alone. This can substantially improve our understanding of how spindle orientation is regulated and provide insights to investigate this complex event.
Collapse
Affiliation(s)
- Tao Zhong
- Medical Integration and Practice Center, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| | - Xiaoxiao Gongye
- Medical Integration and Practice Center, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| | - Minglei Wang
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| | - Jinming Yu
- Medical Integration and Practice Center, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
24
|
Soffer A, Mahly A, Padmanabhan K, Cohen J, Adir O, Loushi E, Fuchs Y, Williams SE, Luxenburg C. Apoptosis and tissue thinning contribute to symmetric cell division in the developing mouse epidermis in a nonautonomous way. PLoS Biol 2022; 20:e3001756. [PMID: 35969606 PMCID: PMC9410552 DOI: 10.1371/journal.pbio.3001756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 08/25/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022] Open
Abstract
Mitotic spindle orientation (SO) is a conserved mechanism that governs cell fate and tissue morphogenesis. In the developing epidermis, a balance between self-renewing symmetric divisions and differentiative asymmetric divisions is necessary for normal development. While the cellular machinery that executes SO is well characterized, the extrinsic cues that guide it are poorly understood. Here, we identified the basal cell adhesion molecule (BCAM), a β1 integrin coreceptor, as a novel regulator of epidermal morphogenesis. In utero RNAi-mediated depletion of Bcam in the mouse embryo did not hinder β1 integrin distribution or cell adhesion and polarity. However, Bcam depletion promoted apoptosis, thinning of the epidermis, and symmetric cell division, and the defects were reversed by concomitant overexpression of the apoptosis inhibitor Xiap. Moreover, in mosaic epidermis, depletion of Bcam or Xiap induced symmetric divisions in neighboring wild-type cells. These results identify apoptosis and epidermal architecture as extrinsic cues that guide SO in the developing epidermis.
Collapse
Affiliation(s)
- Arad Soffer
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adnan Mahly
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Krishnanand Padmanabhan
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan Cohen
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Adir
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eidan Loushi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaron Fuchs
- Department of Biology, Technion—Israel Institute of Technology, Haifa, Israel
| | - Scott E. Williams
- Departments of Pathology & Laboratory Medicine and Biology, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Chen Luxenburg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
25
|
Spindle motility skews division site determination during asymmetric cell division in Physcomitrella. Nat Commun 2022; 13:2488. [PMID: 35513464 PMCID: PMC9072379 DOI: 10.1038/s41467-022-30239-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 04/21/2022] [Indexed: 01/09/2023] Open
Abstract
Asymmetric cell division (ACD) underlies the development of multicellular organisms. In animal ACD, the cell division site is determined by active spindle-positioning mechanisms. In contrast, it is considered that the division site in plants is determined prior to mitosis by the microtubule-actin belt known as the preprophase band (PPB) and that the localization of the mitotic spindle is typically static and does not govern the division plane. However, in some plant species, ACD occurs in the absence of PPB. Here, we isolate a hypomorphic mutant of the conserved microtubule-associated protein TPX2 in the moss Physcomitrium patens (Physcomitrella) and observe spindle motility during PPB-independent cell division. This defect compromises the position of the division site and produces inverted daughter cell sizes in the first ACD of gametophore (leafy shoot) development. The phenotype is rescued by restoring endogenous TPX2 function and, unexpectedly, by depolymerizing actin filaments. Thus, we identify an active spindle-positioning mechanism that, reminiscent of acentrosomal ACD in animals, involves microtubules and actin filaments, and sets the division site in plants.
Collapse
|
26
|
Wang LL, Wan XY, Liu CQ, Zheng FM. NDR1 increases NOTCH1 signaling activity by impairing Fbw7 mediated NICD degradation to enhance breast cancer stem cell properties. Mol Med 2022; 28:49. [PMID: 35508987 PMCID: PMC9066784 DOI: 10.1186/s10020-022-00480-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/18/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The existence of breast cancer stem cells (BCSCs) causes tumor relapses, metastasis and resistance to conventional therapy in breast cancer. NDR1 kinase, a component of the Hippo pathway, plays important roles in multiple biological processes. However, its role in cancer stem cells has not been explored. The purpose of this study was to investigate the roles of NDR1 in modulating BCSCs. METHODS The apoptosis was detected by Annexin V/Propidium Iodide staining and analyzed by flow cytometry. BCSCs were detected by CD24/44 or ALDEFLUOR staining and analyzed by flow cytometry. The proliferation ability of BCSCs was evaluated by sphere formation assay. The expression of interested proteins was detected by western blot analysis. The expression of HES-1 and c-MYC was detected by real-time PCR. Notch1 signaling activation was detected by luciferase reporter assay. Protein interaction was evaluated by immunoprecipitation. Protein degradation was evaluated by ubiquitination analysis. The clinical relevance of NDR1 was analyzed by Kaplan-Meier Plotter. RESULTS NDR1 regulates apoptosis and drug resistance in breast cancer cells. The upregulation of NDR1 increases CD24low/CD44high or ALDEFLUORhigh population and sphere-forming ability in SUM149 and MCF-7 cells, while downregulation of NDR1 induces opposite effects. NDR1 increased the expression of the Notch1 intracellular domain (NICD) and activated the transcription of its downstream target (HES-1 and c-MYC). Critically, both suppression of Notch pathway activation by DAPT treatment or downregulation of Notch1 expression by shRNA reverses NDR1 enhanced BCSC properties. Mechanically, NDR1 interactes with both NICD or Fbw7 in a kinase activity-independent manner. NDR1 reduces the proteolytic turnover of NICD by competing with Fbw7 for NICD binding, thereby leading to Notch pathway activation. Furthermore, NDR1 might function as a hub to modulate IL-6, TNF-α or Wnt3a induced activation of Notch1 signaling pathway and enrichment of breast cancer stem cells. Moreover, we find that the elevation of NDR1 expression predictes poor survival (OS, RFS, DMFS and PPS) in breast cancer. CONCLUSION Our study revealed a novel function of NDR1 in regulating BCSC properties by activating the Notch pathway. These data might provide a potential strategy for eradicating BCSC to overcome tumor relapses, metastasis and drug resistance.
Collapse
Affiliation(s)
- Ling-Ling Wang
- Department of Medical Oncology of The Eastern Hospital, The First Affiliated Hospital, Sun Yat-Sen University, No.58, Zhong Shan Er Lu, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Yun Wan
- Department of Medical Oncology, Guangzhou Panyu Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chun-Qi Liu
- Department of Thoracic Surgery, Panyu Central Hospital, Guangzhou, China
| | - Fei-Meng Zheng
- Department of Medical Oncology of The Eastern Hospital, The First Affiliated Hospital, Sun Yat-Sen University, No.58, Zhong Shan Er Lu, Guangzhou, 510080, China. .,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
27
|
He Y, Peng L, Li J, Li Q, Chu Y, Lin Q, Rui R, Ju S. TPX2 deficiency leads to spindle abnormity and meiotic impairment in porcine oocytes. Theriogenology 2022; 187:164-172. [DOI: 10.1016/j.theriogenology.2022.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 10/18/2022]
|
28
|
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
|
29
|
Pomp O, Lim HYG, Skory RM, Moverley AA, Tetlak P, Bissiere S, Plachta N. A monoastral mitotic spindle determines lineage fate and position in the mouse embryo. Nat Cell Biol 2022; 24:155-167. [PMID: 35102267 DOI: 10.1038/s41556-021-00826-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022]
Abstract
During mammalian development, the first asymmetric cell divisions segregate cells into inner and outer positions of the embryo to establish the pluripotent and trophectoderm lineages. Typically, polarity components differentially regulate the mitotic spindle via astral microtubule arrays to trigger asymmetric division patterns. However, early mouse embryos lack centrosomes, the microtubule-organizing centres (MTOCs) that usually generate microtubule asters. Thus, it remains unknown whether spindle organization regulates lineage segregation. Here we find that heterogeneities in cell polarity in the early 8-cell-stage mouse embryo trigger the assembly of a highly asymmetric spindle organization. This spindle arises in an unusual modular manner, forming a single microtubule aster from an apically localized, non-centrosomal MTOC, before joining it to the rest of the spindle apparatus. When fully assembled, this 'monoastral' spindle triggers spatially asymmetric division patterns to segregate cells into inner and outer positions. Moreover, the asymmetric inheritance of spindle components causes differential cell polarization to determine pluripotent versus trophectoderm lineage fate.
Collapse
Affiliation(s)
- Oz Pomp
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hui Yi Grace Lim
- Institute of Molecular and Cell Biology, ASTAR, Singapore, Singapore
| | - Robin M Skory
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam A Moverley
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Piotr Tetlak
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie Bissiere
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas Plachta
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Alubaidi G, Hasan S. Stem cells: Biology, types, polarity, and asymmetric cell division: A review. MEDICAL JOURNAL OF BABYLON 2022. [DOI: 10.4103/mjbl.mjbl_34_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
31
|
Penisson M, Jin M, Wang S, Hirotsune S, Francis F, Belvindrah R. Lis1 mutation prevents basal radial glia-like cell production in the mouse. Hum Mol Genet 2021; 31:942-957. [PMID: 34635911 DOI: 10.1093/hmg/ddab295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 01/26/2023] Open
Abstract
Human cerebral cortical malformations are associated with progenitor proliferation and neuronal migration abnormalities. Progenitor cells include apical radial glia, intermediate progenitors and basal (or outer) radial glia (bRGs or oRGs). bRGs are few in number in lissencephalic species (e.g. the mouse) but abundant in gyrencephalic brains. The LIS1 gene coding for a dynein regulator, is mutated in human lissencephaly, associated also in some cases with microcephaly. LIS1 was shown to be important during cell division and neuronal migration. Here, we generated bRG-like cells in the mouse embryonic brain, investigating the role of Lis1 in their formation. This was achieved by in utero electroporation of a hominoid-specific gene TBC1D3 (coding for a RAB-GAP protein) at mouse embryonic day (E) 14.5. We first confirmed that TBC1D3 expression in wild-type (WT) brain generates numerous Pax6+ bRG-like cells that are basally localized. Second, using the same approach, we assessed the formation of these cells in heterozygote Lis1 mutant brains. Our novel results show that Lis1 depletion in the forebrain from E9.5 prevented subsequent TBC1D3-induced bRG-like cell amplification. Indeed, we observe perturbation of the ventricular zone (VZ) in the mutant. Lis1 depletion altered adhesion proteins and mitotic spindle orientations at the ventricular surface and increased the proportion of abventricular mitoses. Progenitor outcome could not be further altered by TBC1D3. We conclude that disruption of Lis1/LIS1 dosage is likely to be detrimental for appropriate progenitor number and position, contributing to lissencephaly pathogenesis.
Collapse
Affiliation(s)
- Maxime Penisson
- INSERM U 1270, Paris, France.,Sorbonne University, UMR-S 1270, F-75005 Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Mingyue Jin
- Osaka City University Graduate School of Medicine, Genetic Disease Research, Asahi-machi 1-4-3, Osaka, JP 545-8585
| | - Shengming Wang
- Osaka City University Graduate School of Medicine, Genetic Disease Research, Asahi-machi 1-4-3, Osaka, JP 545-8585
| | - Shinji Hirotsune
- Osaka City University Graduate School of Medicine, Genetic Disease Research, Asahi-machi 1-4-3, Osaka, JP 545-8585
| | - Fiona Francis
- INSERM U 1270, Paris, France.,Sorbonne University, UMR-S 1270, F-75005 Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Richard Belvindrah
- INSERM U 1270, Paris, France.,Sorbonne University, UMR-S 1270, F-75005 Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
32
|
Akhshi T, Trimble WS. A non-canonical Hedgehog pathway initiates ciliogenesis and autophagy. J Cell Biol 2021; 220:211568. [PMID: 33258871 PMCID: PMC7714386 DOI: 10.1083/jcb.202004179] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/19/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Primary cilia function as critical signaling hubs whose absence leads to severe disorders collectively known as ciliopathies; our knowledge of ciliogenesis remains limited. We show that Smo induces ciliogenesis through two distinct yet essential noncanonical Hh pathways in several cell types, including neurons. Surprisingly, ligand activation of Smo induces autophagy via an LKB1-AMPK axis to remove the satellite pool of OFD1. This is required, but not sufficient, for ciliogenesis. Additionally, Smo activates the Gαi-LGN-NuMA-dynein axis, causing accumulation of a portion of OFD1 at centrioles in early ciliogenesis. Both pathways are critical for redistribution of BBS4 from satellites to centrioles, which is also mediated by OFD1 centriolar translocation. Notably, different Smo agonists, which activate Smo distinctly, activate one or the other of these pathways; only in combination they recapitulate the activity of Hh ligand. These studies provide new insight into physiological stimuli (Hh) that activate autophagy and promote ciliogenesis and introduce a novel role for the Gαi-LGN-NuMA-dynein complex in this process.
Collapse
Affiliation(s)
- Tara Akhshi
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| | - William S Trimble
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
33
|
Kindt KS, Akturk A, Jarysta A, Day M, Beirl A, Flonard M, Tarchini B. EMX2-GPR156-Gαi reverses hair cell orientation in mechanosensory epithelia. Nat Commun 2021; 12:2861. [PMID: 34001891 PMCID: PMC8129141 DOI: 10.1038/s41467-021-22997-1] [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: 06/02/2020] [Accepted: 04/10/2021] [Indexed: 12/21/2022] Open
Abstract
Hair cells detect sound, head position or water movements when their mechanosensory hair bundle is deflected. Each hair bundle has an asymmetric architecture that restricts stimulus detection to a single axis. Coordinated hair cell orientations within sensory epithelia further tune stimulus detection at the organ level. Here, we identify GPR156, an orphan GPCR of unknown function, as a critical regulator of hair cell orientation. We demonstrate that the transcription factor EMX2 polarizes GPR156 distribution, enabling it to signal through Gαi and trigger a 180° reversal in hair cell orientation. GPR156-Gαi mediated reversal is essential to establish hair cells with mirror-image orientations in mouse otolith organs in the vestibular system and in zebrafish lateral line. Remarkably, GPR156-Gαi also instructs hair cell reversal in the auditory epithelium, despite a lack of mirror-image organization. Overall, our work demonstrates that conserved GPR156-Gαi signaling is integral to the framework that builds directional responses into mechanosensory epithelia.
Collapse
MESH Headings
- Animals
- Cell Polarity/genetics
- Epithelium/metabolism
- Female
- GTP-Binding Protein alpha Subunits, Gi-Go/genetics
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Hair Cells, Auditory/cytology
- Hair Cells, Auditory/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal/methods
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Zebrafish/metabolism
- Mice
Collapse
Affiliation(s)
- Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Alisha Beirl
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | | | - Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME, USA.
- Department of Medicine, Tufts University, Boston, MA, USA.
- Graduate School of Biomedical Science and Engineering (GSBSE), University of Maine, Orono, ME, USA.
| |
Collapse
|
34
|
Vakilian M, Ghaedi K. A new hypothetical model for pancreatic development based on change in the cell division orientation. Gene 2021; 785:145607. [PMID: 33775847 DOI: 10.1016/j.gene.2021.145607] [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: 10/16/2020] [Revised: 03/05/2021] [Accepted: 03/19/2021] [Indexed: 11/15/2022]
Abstract
Although lifelong renewal and additional compensatory growth in response to demand are undeniable facts, so far, no specific stem cells have been found for pancreatic cells. According to the consensus model, the development of pancreas results from the hierarchical differentiation of pluripotent stem cells towards the appearance of the first endocrine and exocrine cells at approximately 7.5 to 8th gestation week (GW) of human embryo. However, the primitive endocrine cells arising from the embryonic phase of development do not appear to be mature or fully functional. Asymmetric localization of cellular components, such as Numb, partition protein complexes (PAR), planar cell polarity components, and certain mRNAs on the apical and basal sides of epithelial cells, causes cellular polarization. According to our model, the equal distribution of cellular components during symmetric cell division yields similar daughter cells that are associated with duct expansion. In contrast, asymmetric cell division is associated with uneven distribution of cellular components among daughter cells, resulting in different fates. Asymmetric cell division leads to duct branching and the development of acinar and stellate cells by a daughter cell, as well as the development of islet progenitor cells through partial epithelial-to-mesenchymal transition (EMT) and delamination of another daughter cell. Recently, we have developed an efficient method to obtain insulin-secreting cells from the transdifferentiation of hESC-derived ductal cells inducing a partial EMT by treatment with Wnt3A and activin A in a hypoxic environment. Similar models can be offered for other tissues and organs such as mammary glands, lungs, prostate, liver, etc. This model may open a new horizon in the field of regenerative medicine and be useful in explaining the cause of certain abnormalities, such as the occurrence of certain cysts and tumors.
Collapse
Affiliation(s)
- Mehrdad Vakilian
- Department of Cell Regeneration and Advanced Therapies, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain; Department of Cell Biology, Genetics and Physiology, University of Malaga (UMA), The Institute of Biomedical Research in Malaga (IBIMA), Málaga, Spain
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science & Technology, University of Isfahan, Hezar Jerib Ave., Azadi Sq., Isfahan, Iran.
| |
Collapse
|
35
|
Guo H, Wei JH, Zhang Y, Seemann J. Importin α phosphorylation promotes TPX2 activation by GM130 to control astral microtubules and spindle orientation. J Cell Sci 2021; 134:jcs.258356. [PMID: 33526712 DOI: 10.1242/jcs.258356] [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: 12/27/2020] [Accepted: 01/11/2021] [Indexed: 01/10/2023] Open
Abstract
Spindle orientation is important in multiple developmental processes as it determines cell fate and function. The orientation of the spindle depends on the assembly of a proper astral microtubule network. Here, we report that the spindle assembly factor TPX2 regulates astral microtubules. TPX2 in the spindle pole area is activated by GM130 (GOLGA2) on Golgi membranes to promote astral microtubule growth. GM130 relieves TPX2 inhibition by competing for importin α1 (KPNA2) binding. Mitotic phosphorylation of importin α at serine 62 (S62) by CDK1 switches its substrate preference from TPX2 to GM130, thereby enabling competition-based activation. Importin α S62A mutation impedes local TPX2 activation and compromises astral microtubule formation, ultimately resulting in misoriented spindles. Blocking the GM130-importin α-TPX2 pathway impairs astral microtubule growth. Our results reveal a novel role for TPX2 in the organization of astral microtubules. Furthermore, we show that the substrate preference of the important mitotic modulator importin α is regulated by CDK1-mediated phosphorylation.
Collapse
Affiliation(s)
- Haijing Guo
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jen-Hsuan Wei
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yijun Zhang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Joachim Seemann
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
36
|
Godard BG, Dumollard R, Munro E, Chenevert J, Hebras C, McDougall A, Heisenberg CP. Apical Relaxation during Mitotic Rounding Promotes Tension-Oriented Cell Division. Dev Cell 2020; 55:695-706.e4. [PMID: 33207225 DOI: 10.1016/j.devcel.2020.10.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/09/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
Global tissue tension anisotropy has been shown to trigger stereotypical cell division orientation by elongating mitotic cells along the main tension axis. Yet, how tissue tension elongates mitotic cells despite those cells undergoing mitotic rounding (MR) by globally upregulating cortical actomyosin tension remains unclear. We addressed this question by taking advantage of ascidian embryos, consisting of a small number of interphasic and mitotic blastomeres and displaying an invariant division pattern. We found that blastomeres undergo MR by locally relaxing cortical tension at their apex, thereby allowing extrinsic pulling forces from neighboring interphasic blastomeres to polarize their shape and thus division orientation. Consistently, interfering with extrinsic forces by reducing the contractility of interphasic blastomeres or disrupting the establishment of asynchronous mitotic domains leads to aberrant mitotic cell division orientations. Thus, apical relaxation during MR constitutes a key mechanism by which tissue tension anisotropy controls stereotypical cell division orientation.
Collapse
Affiliation(s)
- Benoit G Godard
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France; Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Rémi Dumollard
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | - Edwin Munro
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Janet Chenevert
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | - Céline Hebras
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | - Alex McDougall
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | | |
Collapse
|
37
|
Singh D, Schmidt N, Müller F, Bange T, Bird AW. Destabilization of Long Astral Microtubules via Cdk1-Dependent Removal of GTSE1 from Their Plus Ends Facilitates Prometaphase Spindle Orientation. Curr Biol 2020; 31:766-781.e8. [PMID: 33333009 DOI: 10.1016/j.cub.2020.11.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/25/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022]
Abstract
The precise regulation of microtubule dynamics over time and space in dividing cells is critical for several mitotic mechanisms that ultimately enable cell proliferation, tissue organization, and development. Astral microtubules, which extend from the centrosome toward the cell cortex, must be present for the mitotic spindle to properly orient, as well as for the faithful execution of anaphase and cytokinesis. However, little is understood about how the dynamic properties of astral microtubules are regulated spatiotemporally, or the contribution of astral microtubule dynamics to spindle positioning. The mitotic regulator Cdk1-CyclinB promotes destabilization of centrosomal microtubules and increased microtubule dynamics as cells enter mitosis, but how Cdk1 activity modulates astral microtubule stability, and whether it impacts spindle positioning, is unknown. Here, we uncover a mechanism revealing that Cdk1 destabilizes astral microtubules in prometaphase and thereby influences spindle reorientation. Phosphorylation of the EB1-dependent microtubule plus-end tracking protein GTSE1 by Cdk1 in early mitosis abolishes its interaction with EB1 and recruitment to microtubule plus ends. Loss of Cdk1 activity, or mutation of phosphorylation sites in GTSE1, induces recruitment of GTSE1 to growing microtubule plus ends in mitosis. This decreases the catastrophe frequency of astral microtubules and causes an increase in the number of long astral microtubules reaching the cell cortex, which restrains the ability of cells to reorient spindles along the long cellular axis in early mitosis. Astral microtubules thus must not only be present but also dynamic to allow the spindle to reorient, a state assisted by selective destabilization of long astral microtubules via Cdk1.
Collapse
Affiliation(s)
- Divya Singh
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Nadine Schmidt
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Franziska Müller
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Tanja Bange
- Department for Systems Chronobiology, Institute of Medical Psychology, LMU Munich, Goethestrasse 31/ I, 80336 Munich, Germany
| | - Alexander W Bird
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany.
| |
Collapse
|
38
|
Lagos-Cabré R, Ivanova A, Taylor CW. Ca 2+ Release by IP 3 Receptors Is Required to Orient the Mitotic Spindle. Cell Rep 2020; 33:108483. [PMID: 33326774 PMCID: PMC7758162 DOI: 10.1016/j.celrep.2020.108483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 09/23/2020] [Accepted: 11/13/2020] [Indexed: 11/30/2022] Open
Abstract
The mitotic spindle distributes chromosomes evenly to daughter cells during mitosis. The orientation of the spindle, guided by internal and external cues, determines the axis of cell division and thereby contributes to tissue morphogenesis. Progression through mitosis requires local Ca2+ signals at critical steps, and because store-operated Ca2+ entry is inhibited during mitosis, those signals probably require Ca2+ release through inositol 1,4,5-trisphosphate receptors (IP3Rs). In cells without IP3Rs, astral microtubules around the daughter centrosome are shorter than those at the mother centrosome, and the mitotic spindle fails to align with the substratum during metaphase. The misalignment is due to the spindle ineffectively detecting internal cues rather than a failure of cells to recognize the substratum. Expression of type 3 IP3R is sufficient to rescue spindle alignment, but only if the IP3R has a functional pore. We conclude that Ca2+ signals evoked by IP3Rs are required to orient the mitotic spindle. IP3 receptors are required for mitotic spindle orientation Only IP3 receptors with a functional channel restore spindle orientation Ca2+ release through IP3 receptors is required for spindle orientation
Collapse
Affiliation(s)
- Raul Lagos-Cabré
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Adelina Ivanova
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Colin W Taylor
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| |
Collapse
|
39
|
Ling J, Sckaff M, Tiwari M, Chen Y, Li J, Jones J, Sen GL. RAS-mediated suppression of PAR3 and its effects on SCC initiation and tissue architecture occur independently of hyperplasia. J Cell Sci 2020; 133:jcs.249102. [PMID: 33172988 DOI: 10.1242/jcs.249102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Proper epithelial development and homeostasis depends on strict control of oriented cell division. Current evidence shows that this process is regulated by intrinsic polarity factors and external spatial cues. Owing to the lack of an appropriate model system that can recapitulate the architecture of the skin, deregulation of spindle orientation in human epithelial carcinoma has never been investigated. Here, using an inducible model of human squamous cell carcinoma (SCC), we demonstrate that RAS-dependent suppression of PAR3 (encoded by PARD3) accelerates epithelial disorganization during early tumorigenesis. Diminished PAR3 led to loss of E-cadherin-mediated cell adhesion, which in turn contributed to misoriented cell division. Pharmacological inhibition of the MAPK pathway downstream of RAS activation reversed the defects in PAR3 expression, E-cadherin-mediated cell adhesion and mitotic spindle orientation. Thus, temporal analysis of human neoplasia provides a powerful approach to study cellular and molecular transformations during early oncogenesis, which allowed identification of PAR3 as a critical regulator of tissue architecture during initial human SCC development.
Collapse
Affiliation(s)
- Ji Ling
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Maria Sckaff
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Manisha Tiwari
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Yifang Chen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Jingting Li
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Jackson Jones
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - George L Sen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| |
Collapse
|
40
|
Keshri R, Rajeevan A, Kotak S. PP2A--B55γ counteracts Cdk1 and regulates proper spindle orientation through the cortical dynein adaptor NuMA. J Cell Sci 2020; 133:jcs243857. [PMID: 32591484 PMCID: PMC7406356 DOI: 10.1242/jcs.243857] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
Proper orientation of the mitotic spindle is critical for accurate development and morphogenesis. In human cells, spindle orientation is regulated by the evolutionarily conserved protein NuMA, which interacts with dynein and enriches it at the cell cortex. Pulling forces generated by cortical dynein orient the mitotic spindle. Cdk1-mediated phosphorylation of NuMA at threonine 2055 (T2055) negatively regulates its cortical localization. Thus, only NuMA not phosphorylated at T2055 localizes at the cell cortex. However, the identity and the mechanism of action of the phosphatase complex involved in T2055 dephosphorylation remains elusive. Here, we characterized the PPP2CA-B55γ (PPP2R2C)-PPP2R1B complex that counteracts Cdk1 to orchestrate cortical NuMA for proper spindle orientation. In vitro reconstitution experiments revealed that this complex is sufficient for T2055 dephosphorylation. Importantly, we identified polybasic residues in NuMA that are critical for T2055 dephosphorylation, and for maintaining appropriate cortical NuMA levels for accurate spindle elongation. Furthermore, we found that Cdk1-mediated phosphorylation and PP2A-B55γ-mediated dephosphorylation at T2055 are reversible events. Altogether, this study uncovers a novel mechanism by which Cdk1 and its counteracting PP2A-B55γ complex orchestrate spatiotemporal levels of cortical force generators for flawless mitosis.
Collapse
Affiliation(s)
- Riya Keshri
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| | - Ashwathi Rajeevan
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| | - Sachin Kotak
- Department of Microbiology and Cell Biology, Indian Institute of Science, 560012 Bangalore, India
| |
Collapse
|
41
|
Wu X, Cai Q, Feng Z, Zhang M. Liquid-Liquid Phase Separation in Neuronal Development and Synaptic Signaling. Dev Cell 2020; 55:18-29. [PMID: 32726576 DOI: 10.1016/j.devcel.2020.06.012] [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: 04/09/2020] [Revised: 05/27/2020] [Accepted: 06/09/2020] [Indexed: 01/09/2023]
Abstract
Formation of biomolecular condensates that are not enclosed by membranes via liquid-liquid phase separation (LLPS) is a general strategy that cells adopt to organize membraneless subcellular compartments for diverse functions. Neurons are highly polarized with elaborate branching and functional compartmentalization of their neurites, thus, raising additional demand for the proper subcellular localization of both membraneless and membrane-based organelles. Recent studies have provided evidence that several protein assemblies involved in the establishment of neuronal stem cell (NSC) polarity and in the asymmetric division of NSCs form distinct molecular condensates via LLPS. In synapses of adult neurons, molecular apparatuses controlling presynaptic neurotransmitter release and postsynaptic signaling transmission are also likely formed via LLPS. These molecular condensates, though not enclosed by lipid bilayers, directly associate with plasma membranes or membrane-based organelles, indicating that direct communication between membraneless and membrane-based organelles is a common theme in neurons and other types of cells.
Collapse
Affiliation(s)
- Xiandeng Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Qixu Cai
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zhe Feng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
42
|
Wavreil FDM, Yajima M. Diversity of activator of G-protein signaling (AGS)-family proteins and their impact on asymmetric cell division across taxa. Dev Biol 2020; 465:89-99. [PMID: 32687894 DOI: 10.1016/j.ydbio.2020.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 11/18/2022]
Abstract
Asymmetric cell division (ACD) is a cellular process that forms two different cell types through a cell division and is thus critical for the development of all multicellular organisms. Not all but many of the ACD processes are mediated by proper orientation of the mitotic spindle, which segregates the fate determinants asymmetrically into daughter cells. In many cell types, the evolutionarily conserved protein complex of Gαi/AGS-family protein/NuMA-like protein appears to play critical roles in orienting the spindle and/or generating the polarized cortical forces to regulate ACD. Studies in various organisms reveal that this conserved protein complex is slightly modified in each phylum or even within species. In particular, AGS-family proteins appear to be modified with a variable number of motifs in their functional domains across taxa. This apparently creates different molecular interactions and mechanisms of ACD in each developmental program, ultimately contributing to developmental diversity across species. In this review, we discuss how a conserved ACD machinery has been modified in each phylum over the course of evolution with a major focus on the molecular evolution of AGS-family proteins and its impact on ACD regulation.
Collapse
Affiliation(s)
- Florence D M Wavreil
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02906, USA
| | - Mamiko Yajima
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02906, USA.
| |
Collapse
|
43
|
Nakamoto A, Kumano G. Dynein-Mediated Regional Cell Division Reorientation Shapes a Tailbud Embryo. iScience 2020; 23:100964. [PMID: 32199290 PMCID: PMC7082557 DOI: 10.1016/j.isci.2020.100964] [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: 08/14/2019] [Revised: 01/17/2020] [Accepted: 03/03/2020] [Indexed: 11/17/2022] Open
Abstract
Regulation of cell division orientation controls the spatial distribution of cells during development and is essential for one-directional tissue transformation, such as elongation. However, little is known about whether it plays a role in other types of tissue morphogenesis. Using an ascidian Halocynthia roretzi, we found that differently oriented cell divisions in the epidermis of the future trunk (anterior) and tail (posterior) regions create an hourglass-like epithelial bending between the two regions to shape the tailbud embryo. Our results show that posterior epidermal cells are polarized with dynein protein anteriorly localized, undergo dynein-dependent spindle rotation, and divide along the anteroposterior axis. This cell division facilitates constriction around the embryo's circumference only in the posterior region and epithelial bending formation. Our findings, therefore, provide an important insight into the role of oriented cell division in tissue morphogenesis.
Collapse
Affiliation(s)
- Ayaki Nakamoto
- Asamushi Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, 9 Sakamoto, Asamushi, Aomori 039-3501, Japan.
| | - Gaku Kumano
- Asamushi Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, 9 Sakamoto, Asamushi, Aomori 039-3501, Japan
| |
Collapse
|
44
|
Lough KJ, Byrd KM, Descovich CP, Spitzer DC, Bergman AJ, Beaudoin GM, Reichardt LF, Williams SE. Telophase correction refines division orientation in stratified epithelia. eLife 2019; 8:49249. [PMID: 31833472 PMCID: PMC6959978 DOI: 10.7554/elife.49249] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
During organogenesis, precise control of spindle orientation balances proliferation and differentiation. In the developing murine epidermis, planar and perpendicular divisions yield symmetric and asymmetric fate outcomes, respectively. Classically, division axis specification involves centrosome migration and spindle rotation, events occurring early in mitosis. Here, we identify a novel orientation mechanism which corrects erroneous anaphase orientations during telophase. The directionality of reorientation correlates with the maintenance or loss of basal contact by the apical daughter. While the scaffolding protein LGN is known to determine initial spindle positioning, we show that LGN also functions during telophase to reorient oblique divisions toward perpendicular. The fidelity of telophase correction also relies on the tension-sensitive adherens junction proteins vinculin, α-E-catenin, and afadin. Failure of this corrective mechanism impacts tissue architecture, as persistent oblique divisions induce precocious, sustained differentiation. The division orientation plasticity provided by telophase correction may enable progenitors to adapt to local tissue needs.
Collapse
Affiliation(s)
- Kendall J Lough
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Kevin M Byrd
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States.,Department of Oral & Craniofacial Health Sciences, The University of North Carolina School of Dentistry, Chapel Hill, United States
| | - Carlos P Descovich
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Danielle C Spitzer
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Abby J Bergman
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Gerard Mj Beaudoin
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States.,Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Louis F Reichardt
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States.,Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Scott E Williams
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| |
Collapse
|
45
|
Takayanagi H, Hayase J, Kamakura S, Miyano K, Chishiki K, Yuzawa S, Sumimoto H. Intramolecular interaction in LGN, an adaptor protein that regulates mitotic spindle orientation. J Biol Chem 2019; 294:19655-19666. [PMID: 31732560 DOI: 10.1074/jbc.ra119.011457] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/07/2019] [Indexed: 12/11/2022] Open
Abstract
Proper mitotic spindle orientation requires that astral microtubules are connected to the cell cortex by the microtubule-binding protein NuMA, which is recruited from the cytoplasm. Cortical recruitment of NuMA is at least partially mediated via direct binding to the adaptor protein LGN. LGN normally adopts a closed conformation via an intramolecular interaction between its N-terminal NuMA-binding domain and its C-terminal region that contains four GoLoco (GL) motifs, each capable of binding to the membrane-anchored Gαi subunit of heterotrimeric G protein. Here we show that the intramolecular association with the N-terminal domain in LGN involves GL3, GL4, and a region between GL2 and GL3, whereas GL1 and GL2 do not play a major role. This conformation renders GL1 but not the other GL motifs in a state easily accessible to Gαi To interact with full-length LGN in a closed state, NuMA requires the presence of Gαi; both NuMA and Gαi are essential for cortical recruitment of LGN in mitotic cells. In contrast, mInsc, a protein that competes with NuMA for binding to LGN and regulates mitotic spindle orientation in asymmetric cell division, efficiently binds to full-length LGN without Gαi and induces its conformational change, enhancing its association with Gαi In nonpolarized symmetrically dividing HeLa cells, disruption of the LGN-NuMA interaction by ectopic expression of mInsc results in a loss of cortical localization of NuMA during metaphase and anaphase and promotes mitotic spindle misorientation and a delayed anaphase progression. These findings highlight a specific role for LGN-mediated cell cortex recruitment of NuMA.
Collapse
Affiliation(s)
- Hiroki Takayanagi
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Junya Hayase
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Sachiko Kamakura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kei Miyano
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kanako Chishiki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Satoru Yuzawa
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| |
Collapse
|
46
|
Abstract
Asymmetric cell division (ACD) is a conserved strategy for achieving cell diversity. A cell can undergo an intrinsic ACD through asymmetric segregation of cell fate determinants or cellular organelles. Recently, a new biophysical concept known as biomolecular phase separation, through which proteins and/or RNAs autonomously form a highly concentrated non-membrane-enclosed compartment via multivalent interactions, has provided new insights into the assembly and regulation of many membrane-less or membrane-attached organelles. Intriguingly, biomolecular phase separation is suggested to drive asymmetric condensation of cell fate determinants during ACD as well as organization of cellular organelles involved in ACD. In this Perspective, I first summarize recent findings on the molecular basis governing intrinsic ACD. Then I will discuss how ACD might be regulated by formation of dense molecular assemblies via phase separation.
Collapse
Affiliation(s)
- Wenyu Wen
- Department of Neurosurgery, Huashan Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences , Shanghai Medical College of Fudan University , Shanghai 200032 , China
| |
Collapse
|
47
|
Franco M, Carmena A. Eph signaling in mitotic spindle orientation: what´s your angle here? Cell Cycle 2019; 18:2590-2597. [PMID: 31475621 DOI: 10.1080/15384101.2019.1658479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The orientation of the mitotic spindle is a crucial process during development and adult tissue homeostasis and multiple mechanisms have been shown to intrinsically regulate this process. However, much less is known about the extrinsic cues involved in modulating spindle orientation. We have recently uncovered a novel function of Eph intercellular signaling in regulating spindle alignment by ultimately ensuring the correct cortical distribution of central components within the intrinsic spindle orientation machinery. Here, we comment on these results, novel questions that they open and potential additional research to address in the future.
Collapse
Affiliation(s)
- Maribel Franco
- Developmental Neurobiology Unit, Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas/Universidad Miguel Hernández , Alicante , Spain
| | - Ana Carmena
- Developmental Neurobiology Unit, Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas/Universidad Miguel Hernández , Alicante , Spain
| |
Collapse
|
48
|
Porter AP, White GRM, Mack NA, Malliri A. The interaction between CASK and the tumour suppressor Dlg1 regulates mitotic spindle orientation in mammalian epithelia. J Cell Sci 2019; 132:jcs230086. [PMID: 31289196 PMCID: PMC6679578 DOI: 10.1242/jcs.230086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/14/2019] [Indexed: 12/30/2022] Open
Abstract
Oriented cell divisions are important for the formation of normal epithelial structures. Dlg1, a tumour suppressor, is required for mitotic spindle orientation in Drosophila epithelia and chick neuroepithelia, but how Dlg1 is localised to the membrane and its importance in mammalian epithelia are unknown. We show that Dlg1 is required in non-transformed mammalian epithelial cells for oriented cell divisions and normal lumen formation. We demonstrate that the MAGUK protein CASK, a membrane-associated scaffold, is the factor responsible for Dlg1 membrane localisation during spindle orientation, thereby identifying a new cellular function for CASK. Depletion of CASK leads to misoriented divisions in 3D, and to the formation of multilumen structures in cultured kidney and breast epithelial cells. Blocking the CASK-Dlg1 interaction with an interfering peptide, or by deletion of the CASK-interaction domain of Dlg1, disrupts spindle orientation and causes multilumen formation. We show that the CASK-Dlg1 interaction is important for localisation of the canonical LGN-NuMA complex known to be required for spindle orientation. These results establish the importance of the CASK-Dlg1 interaction in oriented cell division and epithelial integrity.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Andrew P Porter
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Macclesfield SK10 4TG, UK
| | - Gavin R M White
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Macclesfield SK10 4TG, UK
| | - Natalie A Mack
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Macclesfield SK10 4TG, UK
| | - Angeliki Malliri
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Macclesfield SK10 4TG, UK
| |
Collapse
|
49
|
Wolf B, Busso C, Gönczy P. Live imaging screen reveals that TYRO3 and GAK ensure accurate spindle positioning in human cells. Nat Commun 2019; 10:2859. [PMID: 31253758 PMCID: PMC6599018 DOI: 10.1038/s41467-019-10446-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 04/29/2019] [Indexed: 12/28/2022] Open
Abstract
Proper spindle positioning is crucial for spatial cell division control. Spindle positioning in human cells relies on a ternary complex comprising Gαi1-3, LGN and NuMA, which anchors dynein at the cell cortex, thus enabling pulling forces to be exerted on astral microtubules. We develop a live imaging siRNA-based screen using stereotyped fibronectin micropatterns to uncover components modulating spindle positioning in human cells, testing 1280 genes, including all kinases and phosphatases. We thus discover 16 components whose inactivation dramatically perturbs spindle positioning, including tyrosine receptor kinase 3 (TYRO3) and cyclin G associated kinase (GAK). TYRO3 depletion results in excess NuMA and dynein at the cortex during metaphase, similar to the effect of blocking the TYRO3 downstream target phosphatidylinositol 3-kinase (PI3K). Furthermore, depletion of GAK leads to impaired astral microtubules, similar to the effect of downregulating the GAK-interactor Clathrin. Overall, our work uncovers components and mechanisms governing spindle positioning in human cells.
Collapse
Affiliation(s)
- Benita Wolf
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015, Lausanne, Switzerland.
| |
Collapse
|
50
|
Hart M, Zulkipli I, Shrestha RL, Dang D, Conti D, Gul P, Kujawiak I, Draviam VM. MARK2/Par1b kinase present at centrosomes and retraction fibres corrects spindle off-centring induced by actin disassembly. Open Biol 2019; 9:180263. [PMID: 31238822 PMCID: PMC6597755 DOI: 10.1098/rsob.180263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tissue maintenance and development requires a directed plane of cell division. While it is clear that the division plane can be determined by retraction fibres that guide spindle movements, the precise molecular components of retraction fibres that control spindle movements remain unclear. We report MARK2/Par1b kinase as a novel component of actin-rich retraction fibres. A kinase-dead mutant of MARK2 reveals MARK2's ability to monitor subcellular actin status during interphase. During mitosis, MARK2's localization at actin-rich retraction fibres, but not the rest of the cortical membrane or centrosome, is dependent on its activity, highlighting a specialized spatial regulation of MARK2. By subtly perturbing the actin cytoskeleton, we reveal MARK2's role in correcting mitotic spindle off-centring induced by actin disassembly. We propose that MARK2 provides a molecular framework to integrate cortical signals and cytoskeletal changes in mitosis and interphase.
Collapse
Affiliation(s)
- Madeleine Hart
- 1 School of Biological and Chemical Sciences, Queen Mary University of London , London , UK
| | - Ihsan Zulkipli
- 2 Department of Genetics, University of Cambridge , Cambridge , UK
| | | | - David Dang
- 1 School of Biological and Chemical Sciences, Queen Mary University of London , London , UK.,3 Department of Informatics, King's College, London , London , UK
| | - Duccio Conti
- 1 School of Biological and Chemical Sciences, Queen Mary University of London , London , UK
| | - Parveen Gul
- 1 School of Biological and Chemical Sciences, Queen Mary University of London , London , UK
| | - Izabela Kujawiak
- 2 Department of Genetics, University of Cambridge , Cambridge , UK
| | - Viji M Draviam
- 1 School of Biological and Chemical Sciences, Queen Mary University of London , London , UK
| |
Collapse
|