1
|
Atajanova T, Kang EM, Postnikova A, Price AL, Doerr S, Du M, Ugenti A, Ragkousi K. Lateral cell polarization drives organization of epithelia in sea anemone embryos and embryonic cell aggregates. Proc Natl Acad Sci U S A 2024; 121:e2408763121. [PMID: 39471210 PMCID: PMC11573592 DOI: 10.1073/pnas.2408763121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024] Open
Abstract
One of the first organizing processes during animal development is the assembly of embryonic cells into epithelia. Common features unite epithelialization across select bilaterians, however, we know less about the molecular and cellular mechanisms that drive epithelial emergence in early branching nonbilaterians. In sea anemones, epithelia emerge both during embryonic development and after cell aggregation of dissociated tissues. Although adhesion is required to keep cells together, it is not clear whether cell polarization plays a role as epithelia emerge from disordered aggregates. Here, we use the embryos of the sea anemone Nematostella vectensis to investigate the evolutionary origins of epithelial development. We demonstrate that lateral cell polarization is essential for epithelial organization in both embryos and aggregates. With disrupted lateral polarization, cell contact in the aggregate is not sufficient to trigger epithelialization and further tissue development. Specifically, knockdown of the conserved lateral polarity and tumor suppressor protein Lethal giant larvae (Lgl) disrupts epithelia in developing embryos and impairs the capacity of dissociated cells to epithelialize from aggregates. In contrast to other systems, cells in Nematostella lgl knockdown embryos do not undergo excessive proliferation. Cells with reduced Lgl levels lose their columnar shape and proper positioning of their mitotic spindles and basal bodies. Due to misoriented divisions and aberrant shapes, cells arrange nonuniformly without forming a monolayer. Together our data show that, in Nematostella, Lgl drives epithelialization in embryos and cell aggregates through its effect on cell shape and organelle localization.
Collapse
Affiliation(s)
| | | | | | | | - Sophia Doerr
- Department of Biology, Amherst College, Amherst, MA 01002
| | - Michael Du
- Department of Biology, Amherst College, Amherst, MA 01002
| | - Alicia Ugenti
- Department of Biology, Amherst College, Amherst, MA 01002
| | | |
Collapse
|
2
|
Atajanova T, Kang EM, Postnikova A, Price AL, Doerr S, Du M, Ugenti A, Ragkousi K. Lateral cell polarization drives organization of epithelia in sea anemone embryos and embryonic cell aggregates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.07.588493. [PMID: 38645007 PMCID: PMC11030385 DOI: 10.1101/2024.04.07.588493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
One of the first organizing processes during animal development is the assembly of embryonic cells into epithelia. In certain animals, including Hydra and sea anemones, epithelia also emerge when cells from dissociated tissues are aggregated back together. Although cell adhesion is required to keep cells together, it is not clear whether cell polarization plays a role as epithelia emerge from disordered aggregates. Here, we demonstrate that lateral cell polarization is essential for epithelial organization in both embryos and aggregates of the sea anemone Nematostella vectensis. Specifically, knock down of the lateral polarity protein Lgl disrupts epithelia in developing embryos and impairs the capacity of dissociated cells to epithelialize from aggregates. Cells in lgl mutant epithelia lose their columnar shape and have mispositioned mitotic spindles and ciliary basal bodies. Together, our data suggest that in Nematostella, Lgl is required to establish lateral cell polarity and position cytoskeletal organelles in cells of embryos and aggregates during de novo epithelial organization.
Collapse
Affiliation(s)
- Tavus Atajanova
- Department of Biology, Amherst College, Amherst, MA 01002, USA
- Present address: Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Anna Postnikova
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | | | - Sophia Doerr
- Department of Biology, Amherst College, Amherst, MA 01002, USA
- Present address: Department of Biology, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Michael Du
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Alicia Ugenti
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | | |
Collapse
|
3
|
Endocytosis at the Crossroad of Polarity and Signaling Regulation: Learning from Drosophila melanogaster and Beyond. Int J Mol Sci 2022; 23:ijms23094684. [PMID: 35563080 PMCID: PMC9101507 DOI: 10.3390/ijms23094684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
Cellular trafficking through the endosomal–lysosomal system is essential for the transport of cargo proteins, receptors and lipids from the plasma membrane inside the cells and across membranous organelles. By acting as sorting stations, vesicle compartments direct the fate of their content for degradation, recycling to the membrane or transport to the trans-Golgi network. To effectively communicate with their neighbors, cells need to regulate their compartmentation and guide their signaling machineries to cortical membranes underlying these contact sites. Endosomal trafficking is indispensable for the polarized distribution of fate determinants, adaptors and junctional proteins. Conversely, endocytic machineries cooperate with polarity and scaffolding components to internalize receptors and target them to discrete membrane domains. Depending on the cell and tissue context, receptor endocytosis can terminate signaling responses but can also activate them within endosomes that act as signaling platforms. Therefore, cell homeostasis and responses to environmental cues rely on the dynamic cooperation of endosomal–lysosomal machineries with polarity and signaling cues. This review aims to address advances and emerging concepts on the cooperative regulation of endocytosis, polarity and signaling, primarily in Drosophila melanogaster and discuss some of the open questions across the different cell and tissue types that have not yet been fully explored.
Collapse
|
4
|
Ramalho JJ, Jones VAS, Mutte S, Weijers D. Pole position: How plant cells polarize along the axes. THE PLANT CELL 2022; 34:174-192. [PMID: 34338785 PMCID: PMC8774072 DOI: 10.1093/plcell/koab203] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/30/2021] [Indexed: 05/10/2023]
Abstract
Having a sense of direction is a fundamental cellular trait that can determine cell shape, division orientation, or function, and ultimately the formation of a functional, multicellular body. Cells acquire and integrate directional information by establishing discrete subcellular domains along an axis with distinct molecular profiles, a process known as cell polarization. Insight into the principles and mechanisms underlying cell polarity has been propelled by decades of extensive research mostly in yeast and animal models. Our understanding of cell polarity establishment in plants, which lack most of the regulatory molecules identified in other eukaryotes, is more limited, but significant progress has been made in recent years. In this review, we explore how plant cells coordinately establish stable polarity axes aligned with the organ axes, highlighting similarities in the molecular logic used to polarize both plant and animal cells. We propose a classification system for plant cell polarity events and nomenclature guidelines. Finally, we provide a deep phylogenetic analysis of polar proteins and discuss the evolution of polarity machineries in plants.
Collapse
Affiliation(s)
| | | | - Sumanth Mutte
- Laboratory of Biochemistry, Wageningen University, Stippeneng 4, 6703WE Wageningen, The Netherlands
| | | |
Collapse
|
5
|
Leng S, Xie F, Liu J, Shen J, Quan G, Wen T. LLGL2 Increases Ca 2+ Influx and Exerts Oncogenic Activities via PI3K/AKT Signaling Pathway in Hepatocellular Carcinoma. Front Oncol 2021; 11:683629. [PMID: 34178676 PMCID: PMC8223678 DOI: 10.3389/fonc.2021.683629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/20/2021] [Indexed: 02/05/2023] Open
Abstract
Background Lethal giant larvae (Lgl), scaffolding proteins, regulate the epithelial cell apicobasal polarity in Drosophila. They play important roles in asymmetric cell division, cell migration, and progenitor cells self-renewal as tumor suppressors. One of Lgl mammalian homologues proteins, LLGL2 overexpression has been reported in ER+ breast cancer and promotes tumor proliferation through regulating leucine uptake. Nonetheless, the role of LLGL2 in hepatocellular carcinoma (HCC) is still unknown. Methods TCGA dataset mining, qRT-PCR, Western blot along with immunohistochemistry assays were employed to explore LLGL2 expression in human HCC samples and cell lines. Moreover, the clinical value of LLGL2 was investigated in 156 HCC patients. Furthermore, the role as well as the molecular mechanism of LLGL2 in the progression of HCC was explored through a series of in vitro and in vivo experiments. Results LLGL2 was up-regulated in HCC tissues, which was related with certain clinicopathological features including tumor number, vascular invasion as well as advanced stage. High expression of LLGL2 predicted poor prognosis after hepatectomy. LLGL2 promoted HCC cells proliferation, migration and invasion through PI3K/ATK signaling by promoting calcium ion influx. Conclusion Our study identified that LLGL2 is a tumor promoter in HCC for the first time, which could potentially be utilized as a new biomarker and a therapeutic target for HCC.
Collapse
Affiliation(s)
- Shusheng Leng
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China.,General Surgery Department, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu, China
| | - Fei Xie
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The First People's Hospital of Neijiang City, Neijiang, China
| | - Junyi Liu
- Central Laboratory, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu, China
| | - Junyi Shen
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China
| | - Guangqian Quan
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China
| | - Tianfu Wen
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Krautz R, Khalili D, Theopold U. Tissue-autonomous immune response regulates stress signaling during hypertrophy. eLife 2020; 9:64919. [PMID: 33377870 PMCID: PMC7880693 DOI: 10.7554/elife.64919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Postmitotic tissues are incapable of replacing damaged cells through proliferation, but need to rely on buffering mechanisms to prevent tissue disintegration. By constitutively activating the Ras/MAPK-pathway via RasV12-overexpression in the postmitotic salivary glands (SGs) of Drosophila larvae, we overrode the glands adaptability to growth signals and induced hypertrophy. The accompanied loss of tissue integrity, recognition by cellular immunity, and cell death are all buffered by blocking stress signaling through a genuine tissue-autonomous immune response. This novel, spatio-temporally tightly regulated mechanism relies on the inhibition of a feedback-loop in the JNK-pathway by the immune effector and antimicrobial peptide Drosomycin. While this interaction might allow growing SGs to cope with temporary stress, continuous Drosomycin expression in RasV12-glands favors unrestricted hypertrophy. These findings indicate the necessity to refine therapeutic approaches that stimulate immune responses by acknowledging their possible, detrimental effects in damaged or stressed tissues. Tissues and organs work hard to maintain balance in everything from taking up nutrients to controlling their growth. Ageing, wounding, sickness, and changes in the genetic code can all alter this balance, and cause the tissue or organ to lose some of its cells. Many tissues restore this loss by dividing their remaining cells to fill in the gaps. But some – like the salivary glands of fruit fly larvae – have lost this ability. Tissues like these rely on being able to sense and counteract problems as they arise so as to not lose their balance in the first place. The immune system and stress responses are crucial for this process. They trigger steps to correct the problem and interact with each other to find a common decision about the fate of the affected tissue. To better understand how the immune system and stress response work together, Krautz, Khalili and Theopold genetically manipulated cells in the salivary gland of fruit fly larvae. These modifications switched on signals that stimulate cells to keep growing, causing the salivary gland’s tissue to slowly lose its balance and trigger the stress and immune response. The experiments showed that while the stress response instructed the cells in the gland to die, a peptide released by the immune system called Drosomycin blocked this response and prevented the tissue from collapsing. The cells in the part of the gland not producing this immune peptide were consequently killed by the stress response. When all the cells in the salivary gland were forced to produce Drosomycin, none of the cells died and the whole tissue survived. But it also allowed the cells in the gland to grow uncontrollably, like a tumor, threatening the health of the entire organism. Mapping the interactions between immune and stress pathways could help to fine-tune treatments that can prevent tissue damage. Fruit flies share many genetic features and molecular pathways with humans. So, the next step towards these kinds of treatments would be to screen for similar mechanisms that block stress activation in damaged human tissues. But this research carries a warning: careless activation of the immune system to protect stressed tissues could lead to uncontrolled tissue growth, and might cause more harm than good.
Collapse
Affiliation(s)
- Robert Krautz
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Dilan Khalili
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| | - Ulrich Theopold
- Department of Molecular Biosciences, The Wenner-Gren Institute (MBW), Stockholm University, Stockholm, Sweden
| |
Collapse
|
7
|
Ventura G, Moreira S, Barros-Carvalho A, Osswald M, Morais-de-Sá E. Lgl cortical dynamics are independent of binding to the Scrib-Dlg complex but require Dlg-dependent restriction of aPKC. Development 2020; 147:dev.186593. [PMID: 32665243 DOI: 10.1242/dev.186593] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/02/2020] [Indexed: 01/06/2023]
Abstract
Apical-basal polarity underpins the formation of epithelial barriers that are crucial for metazoan physiology. Although apical-basal polarity is long known to require the basolateral determinants Lethal Giant Larvae (Lgl), Discs Large (Dlg) and Scribble (Scrib), mechanistic understanding of their function is limited. Lgl plays a role as an aPKC inhibitor, but it remains unclear whether Lgl also forms complexes with Dlg or Scrib. Using fluorescence recovery after photobleaching, we show that Lgl does not form immobile complexes at the lateral domain of Drosophila follicle cells. Optogenetic depletion of plasma membrane PIP2 or dlg mutants accelerate Lgl cortical dynamics. However, Dlg and Scrib are required only for Lgl localization and dynamic behavior in the presence of aPKC function. Furthermore, light-induced oligomerization of basolateral proteins indicates that Lgl is not part of the Scrib-Dlg complex in the follicular epithelium. Thus, Scrib and Dlg are necessary to repress aPKC activity in the lateral domain but do not provide cortical binding sites for Lgl. Our work therefore highlights that Lgl does not act in a complex but in parallel with Scrib-Dlg to antagonize apical determinants.
Collapse
Affiliation(s)
- Guilherme Ventura
- i3S (Instituto de Investigação e Inovação em Saúde, Universidade do Porto) and IBMC (Instituto de Biologia Molecular e Celular), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Sofia Moreira
- i3S (Instituto de Investigação e Inovação em Saúde, Universidade do Porto) and IBMC (Instituto de Biologia Molecular e Celular), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - André Barros-Carvalho
- i3S (Instituto de Investigação e Inovação em Saúde, Universidade do Porto) and IBMC (Instituto de Biologia Molecular e Celular), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Mariana Osswald
- i3S (Instituto de Investigação e Inovação em Saúde, Universidade do Porto) and IBMC (Instituto de Biologia Molecular e Celular), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Eurico Morais-de-Sá
- i3S (Instituto de Investigação e Inovação em Saúde, Universidade do Porto) and IBMC (Instituto de Biologia Molecular e Celular), Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| |
Collapse
|
8
|
Abedrabbo M, Ravid S. Scribble, Lgl1, and myosin II form a complex in vivo to promote directed cell migration. Mol Biol Cell 2020; 31:2234-2248. [PMID: 32697665 PMCID: PMC7550706 DOI: 10.1091/mbc.e19-11-0657] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Scribble (Scrib) and Lethal giant larvae 1 (Lgl1) are conserved polarity proteins that play important roles in different forms of cell polarity. The roles of Scrib and Lgl1 in apical-basal cell polarity have been studied extensively, but little is known about their roles in the cell polarity of migrating cells. Furthermore, the effect of Scrib and Lgl1 interaction on cell polarity is largely unknown. In this study, we show that Scrib, through its leucine-rich repeat domain, forms a complex in vivo with Lgl1. Scrib also forms a complex with myosin II, and Scrib, Lgl1, and myosin II colocalize at the leading edge of migrating cells. The cellular localization and the cytoskeletal association of Scrib and Lgl1 are interdependent, as depletion of either protein affects its counterpart. In addition, depletion of either Scrib or Lgl1 disrupts the cellular localization of myosin II. We show that depletion of either Scrib or Lgl1 affects cell adhesion through the inhibition of focal adhesion disassembly. Finally, we show that Scrib and Lgl1 are required for proper cell polarity of migrating cells. These results provide new insights into the mechanism regulating the cell polarity of migrating cells by Scrib, Lgl1, and myosin II.
Collapse
Affiliation(s)
- Maha Abedrabbo
- Department of Biochemistry and Molecular Biology, The Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Shoshana Ravid
- Department of Biochemistry and Molecular Biology, The Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
9
|
Carmena A. The Case of the Scribble Polarity Module in Asymmetric Neuroblast Division in Development and Tumorigenesis. Int J Mol Sci 2020; 21:ijms21082865. [PMID: 32325951 PMCID: PMC7215838 DOI: 10.3390/ijms21082865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/29/2022] Open
Abstract
The Scribble polarity module is composed by Scribble (Scrib), Discs large 1 (Dlg1) and Lethal (2) giant larvae (L(2)gl), a group of highly conserved neoplastic tumor suppressor genes (TSGs) from flies to humans. Even though the Scribble module has been profusely studied in epithelial cell polarity, the number of tissues and processes in which it is involved is increasingly growing. Here we discuss the role of the Scribble module in the asymmetric division of Drosophila neuroblasts (NBs), as well as the underlying mechanisms by which those TSGs act in this process. Finally, we also describe what we know about the consequences of mutating these genes in impairing the process of asymmetric NB division and promoting tumor-like overgrowth.
Collapse
Affiliation(s)
- Ana Carmena
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas/Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Alicante, Spain
| |
Collapse
|
10
|
Moreira S, Osswald M, Ventura G, Gonçalves M, Sunkel CE, Morais-de-Sá E. PP1-Mediated Dephosphorylation of Lgl Controls Apical-basal Polarity. Cell Rep 2020; 26:293-301.e7. [PMID: 30625311 DOI: 10.1016/j.celrep.2018.12.060] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/23/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022] Open
Abstract
Apical-basal polarity is a common trait that underlies epithelial function. Although the asymmetric distribution of cortical polarity proteins works in a functioning equilibrium, it also retains plasticity to accommodate cell division, during which the basolateral determinant Lgl is released from the cortex. Here, we investigated how Lgl restores its cortical localization to maintain the integrity of dividing epithelia. We show that cytoplasmic Lgl is reloaded to the cortex at mitotic exit in Drosophila epithelia. Lgl cortical localization depends on protein phosphatase 1, which dephosphorylates Lgl on the serines phosphorylated by aPKC and Aurora A kinases through a mechanism that relies on the regulatory subunit Sds22 and a PP1-interacting RVxF motif of Lgl. This mechanism maintains epithelial polarity and is of particular importance at mitotic exit to couple Lgl cortical reloading with the polarization of the apical domain. Hence, PP1-mediated dephosphorylation of Lgl preserves the apical-basal organization of proliferative epithelia.
Collapse
Affiliation(s)
- Sofia Moreira
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Mariana Osswald
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Guilherme Ventura
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Margarida Gonçalves
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Claudio E Sunkel
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Eurico Morais-de-Sá
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.
| |
Collapse
|
11
|
Peglion F, Goehring NW. Switching states: dynamic remodelling of polarity complexes as a toolkit for cell polarization. Curr Opin Cell Biol 2019; 60:121-130. [PMID: 31295650 PMCID: PMC6906085 DOI: 10.1016/j.ceb.2019.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/07/2019] [Accepted: 05/11/2019] [Indexed: 02/04/2023]
Abstract
Polarity is defined by the segregation of cellular components along a defined axis. To polarize robustly, cells must be able to break symmetry and subsequently amplify these nascent asymmetries. Finally, asymmetric localization of signaling molecules must be translated into functional regulation of downstream effector pathways. Central to these behaviors are a diverse set of cell polarity networks. Within these networks, molecules exhibit varied behaviors, dynamically switching among different complexes and states, active versus inactive, bound versus unbound, immobile versus diffusive. This ability to switch dynamically between states is intimately connected to the ability of molecules to generate asymmetric patterns within cells. Focusing primarily on polarity pathways governed by the conserved PAR proteins, we discuss strategies enabled by these dynamic behaviors that are used by cells to polarize. We highlight not only how switching between states is linked to the ability of polarity proteins to localize asymmetrically, but also how cells take advantage of 'state switching' to regulate polarity in time and space.
Collapse
Affiliation(s)
- Florent Peglion
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, F-75015, Paris, France
| | - Nathan W Goehring
- The Francis Crick Institute, London, UK; MRC Laboratory for Molecular Cell Biology, UCL, London, UK.
| |
Collapse
|
12
|
Carmena A. Non-muscle myosin II activation: adding a classical touch to ROCK. Small GTPases 2019; 12:161-166. [PMID: 31552778 DOI: 10.1080/21541248.2019.1671148] [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: 10/25/2022] Open
Abstract
Non-muscle myosin II molecules are actin-binding proteins with ATPase activity, this latter capacity providing the energy required for actin filament cross-linking and contraction. The activation of these molecular motors relies on direct phosphorylation at conserved sites through different protein kinases, including the Rho-associated coiled coil-containing kinase (ROCK). In the light of some recent results found in our lab, we comment on the necessity of additional regulatory mechanisms to control the subcellular distribution of non-muscle myosin II proteins to ensure their full activation.
Collapse
Affiliation(s)
- Ana Carmena
- Developmental Neurobiology Department, Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas/Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
13
|
Zhang T, Hou C, Zhang S, Liu S, Li Z, Gao J. Lgl1 deficiency disrupts hippocampal development and impairs cognitive performance in mice. GENES BRAIN AND BEHAVIOR 2019; 18:e12605. [PMID: 31415124 DOI: 10.1111/gbb.12605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 08/11/2019] [Accepted: 08/12/2019] [Indexed: 12/15/2022]
Abstract
Cellular polarity is crucial for brain development and morphogenesis. Lethal giant larvae 1 (Lgl1) plays a crucial role in the establishment of cell polarity from Drosophila to mammalian cells. Previous studies have found the importance of Lgl1 in the development of cerebellar, olfactory bulb, and cerebral cortex. However, the role of Lgl1 in hippocampal development during the embryonic stage and function in adult mice is still unknown. In our study, we created Lgl1-deficient hippocampus mice by using Emx1-Cre mice. Histological analysis showed that the Emx1-Lgl1-/- mice exhibited reduced size of the hippocampus with severe malformations of hippocampal cytoarchitecture. These defects mainly originated from the disrupted hippocampal neuroepithelium, including increased cell proliferation, abnormal interkinetic nuclear migration, reduced differentiation, increased apoptosis, gradual disruption of adherens junctions, and abnormal neuronal migration. The radial glial scaffold was disorganized in the Lgl1-deficient hippocampus. Thus, Lgl1 plays a distinct role in hippocampal neurogenesis. In addition, the Emx1-Lgl1-/- mice displayed impaired behavioral performance in the Morris water maze and fear conditioning test.
Collapse
Affiliation(s)
- Tingting Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Congzhe Hou
- Department of Reproductive medicine, Second Hospital of Shandong University, Jinan, Shandong, China
| | - Sen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Shuoyang Liu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Zhenzu Li
- Department of Bioengineering, Shandong Polytechnic, Jinan, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| |
Collapse
|
14
|
Osswald M, Santos AF, Morais-de-Sá E. Light-Induced Protein Clustering for Optogenetic Interference and Protein Interaction Analysis in Drosophila S2 Cells. Biomolecules 2019; 9:biom9020061. [PMID: 30759894 PMCID: PMC6406598 DOI: 10.3390/biom9020061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 12/19/2022] Open
Abstract
Drosophila Schneider 2 (S2) cells are a simple and powerful system commonly used in cell biology because they are well suited for high resolution microscopy and RNAi-mediated depletion. However, understanding dynamic processes, such as cell division, also requires methodology to interfere with protein function with high spatiotemporal control. In this research study, we report the adaptation of an optogenetic tool to Drosophila S2 cells. Light-activated reversible inhibition by assembled trap (LARIAT) relies on the rapid light-dependent heterodimerization between cryptochrome 2 (CRY2) and cryptochrome-interacting bHLH 1 (CIB1) to form large protein clusters. An anti-green fluorescent protein (GFP) nanobody fused with CRY2 allows this method to quickly trap any GFP-tagged protein in these light-induced protein clusters. We evaluated clustering kinetics in response to light for different LARIAT modules, and showed the ability of GFP-LARIAT to inactivate the mitotic protein Mps1 and to disrupt the membrane localization of the polarity regulator Lethal Giant Larvae (Lgl). Moreover, we validated light-induced co-clustering assays to assess protein-protein interactions in S2 cells. In conclusion, GFP-based LARIAT is a versatile tool to answer different biological questions, since it enables probing of dynamic processes and protein-protein interactions with high spatiotemporal resolution in Drosophila S2 cells.
Collapse
Affiliation(s)
- Mariana Osswald
- Epithelial Polarity and Cell Division, i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - A Filipa Santos
- Epithelial Polarity and Cell Division, i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Eurico Morais-de-Sá
- Epithelial Polarity and Cell Division, i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
15
|
Weisman NY. Genetic and Epigenetic Pathways of lethal (2) giant larvae Tumor Suppressor in Drosophila melanogaster. RUSS J GENET+ 2019. [DOI: 10.1134/s1022795419020145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Zhang T, Zhang S, Song X, Zhao X, Hou C, Li Z, Gao J. Loss of Lgl1 Disrupts the Radial Glial Fiber-guided Cortical Neuronal Migration and Causes Subcortical Band Heterotopia in Mice. Neuroscience 2018; 400:132-145. [PMID: 30597194 DOI: 10.1016/j.neuroscience.2018.12.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/16/2022]
Abstract
Radial glial cells (RGCs) are neuronal progenitors and function as scaffolds for neuronal radial migration in the developing cerebral cortex. These functions depend on a polarized radial glial scaffold, which is of fundamental importance for brain development. Lethal giant larvae 1 (Lgl1), a key regulator for cell polarity from Drosophila to mammals, plays a key role in tumorigenesis and brain development. To overcome neonatal lethality in Lgl1-null mice and clarify the role of Lgl1 in mouse cerebral cortex development and function, we created Lgl1 dorsal telencephalon-specific knockout mice mediated by Emx1-Cre. Lgl1Emx1 conditional knockout (CKO) mice had normal life spans and could be used for function research. Histology results revealed that the mutant mice displayed an ectopic cortical mass in the dorsolateral hemispheric region between the normotopic cortex and the subcortical white matter, resembling human subcortical band heterotopia (SBH). The Lgl1Emx1 CKO cortex showed disrupted adherens junctions (AJs), which were accompanied by ectopic RGCs and intermediate progenitors, and disorganization of the radial glial fiber system. The early- and late-born neurons failed to reach the destined position along the disrupted radial glial fiber scaffold and instead accumulated in ectopic positions and formed SBH. Additionally, the absence of Lgl1 led to severe abnormalities in RGCs, including hyperproliferation, impaired differentiation, and increased apoptosis. Lgl1Emx1 CKO mice also displayed deficiencies in anxiety-related behaviors. We concluded that Lgl1 is essential for RGC development and neural migration during cerebral cortex development.
Collapse
Affiliation(s)
- Tingting Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Sen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Xinli Song
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Xiaohan Zhao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Congzhe Hou
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Zhenzu Li
- Department of Bioengineering, Shandong Polytechnic, Jinan 250104, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China.
| |
Collapse
|
17
|
Polarized Organization of the Cytoskeleton: Regulation by Cell Polarity Proteins. J Mol Biol 2018; 430:3565-3584. [DOI: 10.1016/j.jmb.2018.06.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/09/2018] [Accepted: 06/13/2018] [Indexed: 01/02/2023]
|
18
|
Gamblin CL, Parent-Prévost F, Jacquet K, Biehler C, Jetté A, Laprise P. Oligomerization of the FERM-FA protein Yurt controls epithelial cell polarity. J Cell Biol 2018; 217:3853-3862. [PMID: 30082297 PMCID: PMC6219725 DOI: 10.1083/jcb.201803099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/24/2022] Open
Abstract
Drosophila melanogaster Yurt (Yrt) and its mammalian orthologue EPB41L5 limit apical membrane growth in polarized epithelia. EPB41L5 also supports epithelial-mesenchymal transition and metastasis. Yrt and EPB41L5 contain a four-point-one, ezrin, radixin, and moesin (FERM) domain and a FERM-adjacent (FA) domain. The former contributes to the quaternary structure of 50 human proteins, whereas the latter defines a subfamily of 14 human FERM proteins and fulfills unknown roles. In this study, we show that both Yrt and EPB41L5 oligomerize. Our data also establish that the FERM-FA unit forms an oligomeric interface and that multimerization of Yrt is crucial for its function in epithelial cell polarity regulation. Finally, we demonstrate that aPKC destabilizes the Yrt oligomer to repress its functions, thereby revealing a mechanism through which this kinase supports apical domain formation. Overall, our study highlights a conserved biochemical property of fly and human Yrt proteins, describes a novel function of the FA domain, and further characterizes the molecular mechanisms sustaining epithelial cell polarity.
Collapse
Affiliation(s)
- Clémence L Gamblin
- Centre de Recherche sur le Cancer de l'Université Laval, and Axe Oncologie du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Canada
| | - Frédérique Parent-Prévost
- Centre de Recherche sur le Cancer de l'Université Laval, and Axe Oncologie du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Canada
| | - Kévin Jacquet
- Centre de Recherche sur le Cancer de l'Université Laval, and Axe Oncologie du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Canada
| | - Cornélia Biehler
- Centre de Recherche sur le Cancer de l'Université Laval, and Axe Oncologie du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Canada
| | - Alexandra Jetté
- Centre de Recherche sur le Cancer de l'Université Laval, and Axe Oncologie du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Canada
| | - Patrick Laprise
- Centre de Recherche sur le Cancer de l'Université Laval, and Axe Oncologie du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Canada
| |
Collapse
|
19
|
Stephens R, Lim K, Portela M, Kvansakul M, Humbert PO, Richardson HE. The Scribble Cell Polarity Module in the Regulation of Cell Signaling in Tissue Development and Tumorigenesis. J Mol Biol 2018; 430:3585-3612. [PMID: 29409995 DOI: 10.1016/j.jmb.2018.01.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 01/22/2023]
Abstract
The Scribble cell polarity module, comprising Scribbled (Scrib), Discs-large (Dlg) and Lethal-2-giant larvae (Lgl), has a tumor suppressive role in mammalian epithelial cancers. The Scribble module proteins play key functions in the establishment and maintenance of different modes of cell polarity, as well as in the control of tissue growth, differentiation and directed cell migration, and therefore are major regulators of tissue development and homeostasis. Whilst molecular details are known regarding the roles of Scribble module proteins in cell polarity regulation, their precise mode of action in the regulation of other key cellular processes remains enigmatic. An accumulating body of evidence indicates that Scribble module proteins play scaffolding roles in the control of various signaling pathways, which are linked to the control of tissue growth, differentiation and cell migration. Multiple Scrib, Dlg and Lgl interacting proteins have been discovered, which are involved in diverse processes, however many function in the regulation of cellular signaling. Herein, we review the components of the Scrib, Dlg and Lgl protein interactomes, and focus on the mechanism by which they regulate cellular signaling pathways in metazoans, and how their disruption leads to cancer.
Collapse
Affiliation(s)
- Rebecca Stephens
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Krystle Lim
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Marta Portela
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute (CSIC), Avenida Doctor Arce, 37, Madrid 28002, Spain
| | - Marc Kvansakul
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Patrick O Humbert
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia; Department of Biochemistry & Molecular Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Helena E Richardson
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia; Department of Biochemistry & Molecular Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Anatomy & Neurobiology, University of Melbourne, Melbourne, Victoria 3010, Australia.
| |
Collapse
|
20
|
Daniel SG, Russ AD, Guthridge KM, Raina AI, Estes PS, Parsons LM, Richardson HE, Schroeder JA, Zarnescu DC. miR-9a mediates the role of Lethal giant larvae as an epithelial growth inhibitor in Drosophila. Biol Open 2018; 7:bio.027391. [PMID: 29361610 PMCID: PMC5829493 DOI: 10.1242/bio.027391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Drosophila lethal giant larvae (lgl) encodes a conserved tumor suppressor with established roles in cell polarity, asymmetric division, and proliferation control. Lgl's human orthologs, HUGL1 and HUGL2, are altered in human cancers, however, its mechanistic role as a tumor suppressor remains poorly understood. Based on a previously established connection between Lgl and Fragile X protein (FMRP), a miRNA-associated translational regulator, we hypothesized that Lgl may exert its role as a tumor suppressor by interacting with the miRNA pathway. Consistent with this model, we found that lgl is a dominant modifier of Argonaute1 overexpression in the eye neuroepithelium. Using microarray profiling we identified a core set of ten miRNAs that are altered throughout tumorigenesis in Drosophila lgl mutants. Among these are several miRNAs previously linked to human cancers including miR-9a, which we found to be downregulated in lgl neuroepithelial tissues. To determine whether miR-9a can act as an effector of Lgl in vivo, we overexpressed it in the context of lgl knock-down by RNAi and found it able to reduce the overgrowth phenotype caused by Lgl loss in epithelia. Furthermore, cross-comparisons between miRNA and mRNA profiling in lgl mutant tissues and human breast cancer cells identified thrombospondin (tsp) as a common factor altered in both fly and human breast cancer tumorigenesis models. Our work provides the first evidence of a functional connection between Lgl and the miRNA pathway, demonstrates that miR-9a mediates Lgl's role in restricting epithelial proliferation, and provides novel insights into pathways controlled by Lgl during tumor progression. Summary: Mir-9a overexpression can suppress the overgrowth phenotype caused by Lgl knock-down in epithelia. Gene profiling identifies pathways dysregulated in lgl mutants and shared features between flies and human cancer cells.
Collapse
Affiliation(s)
- Scott G Daniel
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Atlantis D Russ
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA.,Genetics Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| | - Kathryn M Guthridge
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Center, Melbourne, Victoria 3000, Australia
| | - Ammad I Raina
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Patricia S Estes
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Linda M Parsons
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Center, Melbourne, Victoria 3000, Australia.,Department of Genetics, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Helena E Richardson
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Center, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, University of Melbourne, Melbourne, Victoria 3000, Australia.,Department of Biochemistry & Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Joyce A Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA.,Genetics Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| | - Daniela C Zarnescu
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA .,Genetics Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
21
|
Lang CF, Munro E. The PAR proteins: from molecular circuits to dynamic self-stabilizing cell polarity. Development 2017; 144:3405-3416. [PMID: 28974638 DOI: 10.1242/dev.139063] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PAR proteins constitute a highly conserved network of scaffolding proteins, adaptors and enzymes that form and stabilize cortical asymmetries in response to diverse inputs. They function throughout development and across the metazoa to regulate cell polarity. In recent years, traditional approaches to identifying and characterizing molecular players and interactions in the PAR network have begun to merge with biophysical, theoretical and computational efforts to understand the network as a pattern-forming biochemical circuit. Here, we summarize recent progress in the field, focusing on recent studies that have characterized the core molecular circuitry, circuit design and spatiotemporal dynamics. We also consider some of the ways in which the PAR network has evolved to polarize cells in different contexts and in response to different cues and functional constraints.
Collapse
Affiliation(s)
- Charles F Lang
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA.,Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Edwin Munro
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA .,Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
22
|
Sokol SY. At the Crossroads between Cell Polarity and Adhesion in Neocortical Development. Dev Cell 2017; 41:453-454. [PMID: 28586640 DOI: 10.1016/j.devcel.2017.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In this issue of Developmental Cell, Jossin et al. (2017) show that the tumor suppressor protein Lgl1 interacts with N-cadherin to stabilize apical junctions in brain stem cells. Neural progenitors lacking Lgl1 had decreased N-cadherin internalization and abnormal cell junctions, generating an ectopic neuronal layer that resembles cerebral cortex abnormalities in humans.
Collapse
Affiliation(s)
- Sergei Y Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
23
|
Moreira S, Morais-de-Sá E. Spatiotemporal phosphoregulation of Lgl: Finding meaning in multiple on/off buttons. BIOARCHITECTURE 2016; 6:29-38. [PMID: 26919260 DOI: 10.1080/19490992.2016.1149290] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intracellular asymmetries, often termed cell polarity, determine how cells organize and divide to ultimately control cell fate and shape animal tissues. The tumor suppressor Lethal giant larvae (Lgl) functions at the core of the evolutionarily conserved cell polarity machinery that controls apico-basal polarization. This function relies on its restricted basolateral localization via phosphorylation by aPKC. Here, we summarize the spatial and temporal control of Lgl during the cell cycle, highlighting two ideas that emerged from our recent findings: 1) Aurora A directly phosphorylates Lgl during symmetric division to couple reorganization of epithelial polarity with the cell cycle; 2) Phosphorylation of Lgl within three conserved serines controls its localization and function in a site-specific manner. Considering the importance of phosphorylation to regulate the concentration of Lgl at the plasma membrane, we will further discuss how it may work as an on-off switch for the interaction with cortical binding partners, with implications on epithelial polarization and spindle orientation.
Collapse
Affiliation(s)
- Sofia Moreira
- a IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto , Porto , Portugal.,b I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal
| | - Eurico Morais-de-Sá
- a IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto , Porto , Portugal.,b I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto , Portugal
| |
Collapse
|
24
|
aPKC regulates apical localization of Lgl to restrict elongation of microridges in developing zebrafish epidermis. Nat Commun 2016; 7:11643. [PMID: 27249668 PMCID: PMC4895443 DOI: 10.1038/ncomms11643] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 04/15/2016] [Indexed: 12/05/2022] Open
Abstract
Epithelial cells exhibit apical membrane protrusions, which confer specific functions to epithelial tissues. Microridges are short actin protrusions that are laterally long and form a maze-like pattern in the apical domain. They are widely found on vertebrate squamous epithelia including epidermis and have functions in mucous retention, membrane storage and abrasion resistance. It is largely unknown how the formation of these laterally long actin projections is regulated. Here, we show that antagonistic interactions between aPKC and Lgl–regulators of apical and basolateral domain identity, respectively,–control the length of microridges in the zebrafish periderm, the outermost layer of the epidermis. aPKC regulates the levels of Lgl and the active form of non-muscle myosinII at the apical cortex to prevent actin polymerization-dependent precocious fusion and elongation of microridges. Our data unravels the functional significance of exclusion of Lgl from the apical domain in epithelial cells. Squamous epithelia present actin-rich microridges on the apical surface, but the mechanism of their formation is not known. Here the authors show that, in zebrafish epidermis, the exclusion of the basolateral regulator Lgl from the apical domain by atypical protein kinase C prevents precocious elongation and fusion of microridges.
Collapse
|
25
|
Bailey MJ, Prehoda KE. Establishment of Par-Polarized Cortical Domains via Phosphoregulated Membrane Motifs. Dev Cell 2015; 35:199-210. [PMID: 26481050 DOI: 10.1016/j.devcel.2015.09.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/03/2015] [Accepted: 09/23/2015] [Indexed: 01/06/2023]
Abstract
The Par polarity complex creates mutually exclusive cortical domains in diverse animal cells. Activity of the atypical protein kinase C (aPKC) is a key output of the Par complex as phosphorylation removes substrates from the Par domain. Here, we investigate how diverse, apparently unrelated Par substrates couple phosphorylation to cortical displacement. Each protein contains a basic and hydrophobic (BH) motif that interacts directly with phospholipids and also overlaps with aPKC phosphorylation sites. Phosphorylation alters the electrostatic character of the sequence, inhibiting interaction with phospholipids and the cell cortex. We searched for overlapping BH and aPKC phosphorylation site motifs (i.e., putative phosphoregulated BH motifs) in several animal proteomes. Candidate proteins with strong PRBH signals associated with the cell cortex but were displaced into the cytoplasm by aPKC. These findings demonstrate a potentially general mechanism for exclusion of proteins from the Par cortical domain in polarized cells.
Collapse
Affiliation(s)
- Matthew J Bailey
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Kenneth E Prehoda
- Department of Chemistry and Biochemistry, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
26
|
Liu X, Lu D, Ma P, Liu H, Cao Y, Sang B, Zhu X, Shi Q, Hu J, Yu R, Zhou X. Hugl-1 inhibits glioma cell growth in intracranial model. J Neurooncol 2015; 125:113-21. [PMID: 26341367 DOI: 10.1007/s11060-015-1901-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 08/29/2015] [Indexed: 12/28/2022]
Abstract
Drosophila lethal (2) giant larvae (lgl) has been reported as a tumor suppressor and could regulate the Drosophila hippo signaling. Human giant larvae-1(Hugl-1), one human homologue of Drosophila lgl, also has been reported to be involved in the development of some human cancers. However, whether Hugl-1 is associated with the pathogenesis of malignant gliomas remains poorly understood. In the present work, we examined the effect of Hugl-1 on glioma cell growth both in vitro and in vivo. Firstly, we found that Hugl-1 protein levels decreased in the human glioma tissues, suggesting that Hugl-1 is involved in glioma progression. Unfortunately, either stably or transiently over-expressing Hugl-1 did not affect glioma cell proliferation in vitro. In addition, Hugl-1 over-expression did not regulate hippo signaling pathway. Interestingly, over-expression of Hugl-1 not only inhibited gliomagenesis but also markedly inhibited cell proliferation and promoted the apoptosis of U251 cells in an orthotopic model of nude mice. Taken together, this study provides the evidence that Hugl-1 inhibits glioma cell growth in intracranial model of nude mice, suggesting that Hugl-1 might be a potential tumor target for glioma therapy.
Collapse
Affiliation(s)
- Xuejiao Liu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Dong Lu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Peng Ma
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Huaqiang Liu
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Yuewen Cao
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Ben Sang
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Xianlong Zhu
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Qiong Shi
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jinxia Hu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Ravid S. The tumor suppressor Lgl1 regulates front-rear polarity of migrating cells. Cell Adh Migr 2015; 8:378-83. [PMID: 25482644 DOI: 10.4161/cam.29387] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cell migration is a highly integrated, multistep process that plays an important role in physiological and pathological processes. The migrating cell is highly polarized, with complex regulatory pathways that integrate its component processes spatially and temporally. The Drosophila tumor suppressor, Lethal (2) giant larvae (Lgl), regulates apical-basal polarity in epithelia and asymmetric cell division. But little is known about the role of Lgl in establishing cell polarity in migrating cells. Recently, we showed that the mammalian Lgl1 interacts directly with non-muscle myosin IIA (NMIIA), inhibiting its ability to assemble into filaments in vitro. Lgl1 also regulates the cellular localization of NMIIA, the maturation of focal adhesions, and cell migration. We further showed that phosphorylation of Lgl1 by aPKCζ prevents its interaction with NMIIA and is important for Lgl1 and acto-NMII cytoskeleton cellular organization. Lgl is a critical downstream target of the Par6-aPKC cell polarity complex; we showed that Lgl1 forms two distinct complexes in vivo, Lgl1-NMIIA and Lgl1-Par6-aPKCζ in different cellular compartments. We further showed that aPKCζ and NMIIA compete to bind directly to Lgl1 through the same domain. These data provide new insights into the role of Lgl1, NMIIA, and Par6-aPKCζ in establishing front-rear polarity in migrating cells. In this commentary, I discuss the role of Lgl1 in the regulation of the acto-NMII cytoskeleton and its regulation by the Par6-aPKCζ polarity complex, and how Lgl1 activity may contribute to the establishment of front-rear polarity in migrating cells.
Collapse
Affiliation(s)
- Shoshana Ravid
- a Department of Biochemistry and Molecular Biology; The Institute of Medical Research Israel-Canada ; The Hebrew University-Hadassah Medical School ; Jerusalem , Israel
| |
Collapse
|
28
|
Watson K, Rossi G, Temple B, Brennwald P. Structural basis for recognition of the Sec4 Rab GTPase by its effector, the Lgl/tomosyn homologue, Sro7. Mol Biol Cell 2015. [PMID: 26202462 PMCID: PMC4569318 DOI: 10.1091/mbc.e15-04-0228] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Members of the tomosyn/Lgl/Sro7 family play important roles in vesicle trafficking and cell polarity in eukaryotic cells. The yeast homologue, Sro7, is believed to act as a downstream effector of the Sec4 Rab GTPase to promote soluble N-ethylmaleimide-sensitive factor adaptor protein receptor (SNARE) assembly during Golgi-to-cell surface vesicle transport. Here we describe the identification of a Sec4 binding site on the surface of Sro7 that is contained within a cleft created by the junction of two adjacent β-propellers that form the core structure of Sro7. Computational docking experiments suggested four models for interaction of GTP-Sec4 with the Sro7 binding cleft. Further mutational and biochemical analyses confirmed that only one of the four docking arrangements is perfectly consistent with our genetic and biochemical interaction data. Close examination of this docking model suggests a structural basis for the high substrate and nucleotide selectivity in effector binding by Sro7. Finally, analysis of the surface variation within the homologous interaction site on tomosyn-1 and Lgl-1 structural models suggests a possible conserved Rab GTPase effector function in tomosyn vertebrate homologues.
Collapse
Affiliation(s)
- Kelly Watson
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599 Graduate Program in Cell and Developmental Biology, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Guendalina Rossi
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Brenda Temple
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC 27599 R. L. Juliano Structural Bioinformatics Core, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Patrick Brennwald
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599 Graduate Program in Cell and Developmental Biology, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| |
Collapse
|
29
|
Cao F, Miao Y, Xu K, Liu P. Lethal (2) giant larvae: an indispensable regulator of cell polarity and cancer development. Int J Biol Sci 2015; 11:380-9. [PMID: 25798058 PMCID: PMC4366637 DOI: 10.7150/ijbs.11243] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/21/2015] [Indexed: 01/04/2023] Open
Abstract
Cell polarity is one of the most basic properties of all normal cells and is essential for regulating numerous biological processes. Loss of polarity is considered a hallmark for cancer. Multiple polarity proteins are implicated in maintenance of cell polarity. Lethal (2) giant larvae (Lgl) is one of polarity proteins that plays an important role in regulating cell polarity, asymmetric division as well as tumorigenesis. Lgl proteins in different species have similar structures and conserved functions. Lgl acts as an indispensable regulator of cell biological function, including cell polarity and asymmetric division, through interplaying with other polarity proteins, regulating exocytosis, mediating cytoskeleton and being involved in signaling pathways. Furthermore, Lgl plays a role of a tumor suppressor, and the aberrant expression of Hugl, a human homologue of Lgl, contributes to multiple cancers. However, the exact functions of Lgl and the underlying mechanisms remain enigmatic. In this review, we will give an overview of the Lgl functions in cell polarity and cancer development, discuss the potential mechanisms underlying these functions, and raise our conclusion of previous studies and points of view about the future studies.
Collapse
Affiliation(s)
- Fang Cao
- 1. Center for Translational Medicine, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| | - Yi Miao
- 1. Center for Translational Medicine, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| | - Kedong Xu
- 2. Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| | - Peijun Liu
- 1. Center for Translational Medicine, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| |
Collapse
|
30
|
Dulyaninova NG, Bresnick AR. The heavy chain has its day: regulation of myosin-II assembly. BIOARCHITECTURE 2015; 3:77-85. [PMID: 24002531 DOI: 10.4161/bioa.26133] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Nonmuscle myosin-II is an actin-based motor that converts chemical energy into force and movement, and thus functions as a key regulator of the eukaryotic cytoskeleton. Although it is established that phosphorylation on the regulatory light chain increases the actin-activated MgATPase activity of the motor and promotes myosin-II filament assembly, studies have begun to characterize alternative mechanisms that regulate filament assembly and disassembly. These investigations have revealed that all three nonmuscle myosin-II isoforms are subject to additional regulatory controls, which impact diverse cellular processes. In this review, we discuss current knowledge on mechanisms that regulate the oligomerization state of nonmuscle myosin-II filaments by targeting the myosin heavy chain.
Collapse
|
31
|
Ye S, Huang Y, Joshi S, Zhang J, Yang F, Zhang G, Smyth SS, Li Z, Takai Y, Whiteheart SW. Platelet secretion and hemostasis require syntaxin-binding protein STXBP5. J Clin Invest 2014; 124:4517-28. [PMID: 25244094 DOI: 10.1172/jci75572] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 07/24/2014] [Indexed: 01/22/2023] Open
Abstract
Genome-wide association studies (GWAS) have linked genes encoding several soluble NSF attachment protein receptor (SNARE) regulators to cardiovascular disease risk factors. Because these regulatory proteins may directly affect platelet secretion, we used SNARE-containing complexes to affinity purify potential regulators from human platelet extracts. Syntaxin-binding protein 5 (STXBP5; also known as tomosyn-1) was identified by mass spectrometry, and its expression in isolated platelets was confirmed by RT-PCR analysis. Coimmunoprecipitation studies showed that STXBP5 interacts with core secretion machinery complexes, such as syntaxin-11/SNAP23 heterodimers, and fractionation studies suggested that STXBP5 also interacts with the platelet cytoskeleton. Platelets from Stxbp5 KO mice had normal expression of other key secretory components; however, stimulation-dependent secretion from each of the 3 granule types was markedly defective. Secretion defects in STXBP5-deficient platelets were confirmed via lumi-aggregometry and FACS analysis for P-selectin and LAMP-1 exposure. Interestingly, STXBP5-deficient platelets had altered granule cargo levels, despite having normal morphology and granule numbers. Consistent with secretion and cargo deficiencies, Stxbp5 KO mice showed dramatic bleeding in the tail transection model and defective hemostasis in the FeCl3-induced carotid injury model. Transplantation experiments indicated that these defects were due to loss of STXBP5 in BM-derived cells. Our data demonstrate that STXBP5 is required for normal arterial hemostasis, due to its contributions to platelet granule cargo packaging and secretion.
Collapse
|
32
|
Izumi Y, Furuse M. Molecular organization and function of invertebrate occluding junctions. Semin Cell Dev Biol 2014; 36:186-93. [PMID: 25239398 DOI: 10.1016/j.semcdb.2014.09.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/25/2014] [Accepted: 09/01/2014] [Indexed: 12/31/2022]
Abstract
Septate junctions (SJs) are specialized intercellular junctions that function as permeability barriers to restrict the free diffusion of solutes through the paracellular routes in invertebrate epithelia. SJs are subdivided into several morphological types that vary among different animal phyla. In several phyla, different types of SJ have been described in different epithelia within an individual. Arthropods have two types of SJs: pleated SJs (pSJs) and smooth SJs (sSJs), found in ectodermally and endodermally derived epithelia, respectively. Several lines of Drosophila research have identified and characterized a large number of pSJ-associated proteins. Two sSJ-specific proteins have been recently reported. Molecular dissection of SJs in Drosophila and animals in other phyla will lead to a better understanding of the functional differences among SJ types and of evolutionary aspects of these permeability barriers.
Collapse
Affiliation(s)
- Yasushi Izumi
- Division of Cell Biology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | - Mikio Furuse
- Division of Cell Biology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; Division of Cerebral Structure, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
33
|
Betapudi V. Life without double-headed non-muscle myosin II motor proteins. Front Chem 2014; 2:45. [PMID: 25072053 PMCID: PMC4083560 DOI: 10.3389/fchem.2014.00045] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 06/19/2014] [Indexed: 11/20/2022] Open
Abstract
Non-muscle myosin II motor proteins (myosin IIA, myosin IIB, and myosin IIC) belong to a class of molecular motor proteins that are known to transduce cellular free-energy into biological work more efficiently than man-made combustion engines. Nature has given a single myosin II motor protein for lower eukaryotes and multiple for mammals but none for plants in order to provide impetus for their life. These specialized nanomachines drive cellular activities necessary for embryogenesis, organogenesis, and immunity. However, these multifunctional myosin II motor proteins are believed to go awry due to unknown reasons and contribute for the onset and progression of many autosomal-dominant disorders, cataract, deafness, infertility, cancer, kidney, neuronal, and inflammatory diseases. Many pathogens like HIV, Dengue, hepatitis C, and Lymphoma viruses as well as Salmonella and Mycobacteria are now known to take hostage of these dedicated myosin II motor proteins for their efficient pathogenesis. Even after four decades since their discovery, we still have a limited knowledge of how these motor proteins drive cell migration and cytokinesis. We need to enrich our current knowledge on these fundamental cellular processes and develop novel therapeutic strategies to fix mutated myosin II motor proteins in pathological conditions. This is the time to think how to relieve the hijacked myosins from pathogens in order to provide a renewed impetus for patients' life. Understanding how to steer these molecular motors in proliferating and differentiating stem cells will improve stem cell based-therapeutics development. Given the plethora of cellular activities non-muscle myosin motor proteins are involved in, their importance is apparent for human life.
Collapse
Affiliation(s)
- Venkaiah Betapudi
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic Cleveland, OH, USA ; Department of Physiology and Biophysics, Case Western Reserve University Cleveland, OH, USA
| |
Collapse
|
34
|
Dahan I, Petrov D, Cohen-Kfir E, Ravid S. The tumor suppressor Lgl1 forms discrete complexes with NMII-A and Par6α-aPKCζ that are affected by Lgl1 phosphorylation. J Cell Sci 2013; 127:295-304. [PMID: 24213535 DOI: 10.1242/jcs.127357] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Non-muscle myosin IIA (NMII-A) and the tumor suppressor lethal giant larvae 1 (Lgl1) play a central role in the polarization of migrating cells. Mammalian Lgl1 interacts directly with NMII-A, inhibiting its ability to assemble into filaments in vitro. Lgl1 also regulates the cellular localization of NMII-A, the maturation of focal adhesions and cell migration. In Drosophila, phosphorylation of Lgl affects its association with the cytoskeleton. Here we show that phosphorylation of mammalian Lgl1 by aPKCζ prevents its interaction with NMII-A both in vitro and in vivo, and affects its inhibition of NMII-A filament assembly. Phosphorylation of Lgl1 affects its cellular localization and is important for the cellular organization of the acto-NMII cytoskeleton. We further show that Lgl1 forms two distinct complexes in vivo, Lgl1-NMIIA and Lgl1-Par6α-aPKCζ, and that the formation of these complexes is affected by the phosphorylation state of Lgl1. The complex Lgl1-Par6α-aPKCζ resides in the leading edge of the cell. Finally, we show that aPKCζ and NMII-A compete to bind directly to Lgl1 at the same domain. These results provide new insights into the mechanism regulating the interaction between Lgl1, NMII-A, Par6α and aPKCζ in polarized migrating cells.
Collapse
Affiliation(s)
- Inbal Dahan
- Department of Biochemistry and Molecular Biology, The Institute of Medical Research Israel-Canada, The Hebrew University - Hadassah Medical School, Jerusalem 91120, Israel
| | | | | | | |
Collapse
|
35
|
Tay HG, Schulze SK, Compagnon J, Foley FC, Heisenberg CP, Yost HJ, Abdelilah-Seyfried S, Amack JD. Lethal giant larvae 2 regulates development of the ciliated organ Kupffer's vesicle. Development 2013; 140:1550-9. [PMID: 23482490 DOI: 10.1242/dev.087130] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Motile cilia perform crucial functions during embryonic development and throughout adult life. Development of organs containing motile cilia involves regulation of cilia formation (ciliogenesis) and formation of a luminal space (lumenogenesis) in which cilia generate fluid flows. Control of ciliogenesis and lumenogenesis is not yet fully understood, and it remains unclear whether these processes are coupled. In the zebrafish embryo, lethal giant larvae 2 (lgl2) is expressed prominently in ciliated organs. Lgl proteins are involved in establishing cell polarity and have been implicated in vesicle trafficking. Here, we identified a role for Lgl2 in development of ciliated epithelia in Kupffer's vesicle, which directs left-right asymmetry of the embryo; the otic vesicles, which give rise to the inner ear; and the pronephric ducts of the kidney. Using Kupffer's vesicle as a model ciliated organ, we found that depletion of Lgl2 disrupted lumen formation and reduced cilia number and length. Immunofluorescence and time-lapse imaging of Kupffer's vesicle morphogenesis in Lgl2-deficient embryos suggested cell adhesion defects and revealed loss of the adherens junction component E-cadherin at lateral membranes. Genetic interaction experiments indicate that Lgl2 interacts with Rab11a to regulate E-cadherin and mediate lumen formation that is uncoupled from cilia formation. These results uncover new roles and interactions for Lgl2 that are crucial for both lumenogenesis and ciliogenesis and indicate that these processes are genetically separable in zebrafish.
Collapse
Affiliation(s)
- Hwee Goon Tay
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hoege C, Hyman AA. Principles of PAR polarity in Caenorhabditis elegans embryos. Nat Rev Mol Cell Biol 2013; 14:315-22. [PMID: 23594951 DOI: 10.1038/nrm3558] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A hallmark of cell polarity in metazoans is the distribution of partitioning defective (PAR) proteins into two domains on the membrane. Domain boundaries are set by the collective integration of mechanical, biochemical and biophysical signals, and the resulting PAR domains define areas of cytosol specialization. However, the complexity of the signals acting on PAR proteins has been a barrier to uncovering the general principles of PAR polarity. We propose that physical studies, when combined with genetic data, provide new understanding of the mechanisms of polarity establishment in the Caenorhabditis elegans embryo and other organisms.
Collapse
Affiliation(s)
- Carsten Hoege
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, D-01307 Dresden, Germany.
| | | |
Collapse
|
37
|
Polarity protein complex Scribble/Lgl/Dlg and epithelial cell barriers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 763:149-70. [PMID: 23397623 DOI: 10.1007/978-1-4614-4711-5_7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The Scribble polarity complex or module is one of the three polarity modules that regulate cell polarity in multiple epithelia including blood-tissue barriers. This protein complex is composed of Scribble, Lethal giant larvae (Lgl) and Discs large (Dlg), which are well conserved across species from fruitflies and worms to mammals. Originally identified in Drosophila and C. elegans where the Scribble complex was found to work with the Par-based and Crumbs-based polarity modules to regulate apicobasal polarity and asymmetry in cells and tissues during embryogenesis, their mammalian homologs have all been identified in recent years. Components of the Scribble complex are known to regulate multiple cellular functions besides cell polarity, which include cell proliferation, assembly and maintenance of adherens junction (AJ) and tight junction (TJ), and they are also tumor suppressors. Herein, we provide an update on the Scribble polarity complex and how this protein complex modulates cell adhesion with some emphasis on its role in Sertoli cell blood-testis barrier (BTB) function. It should be noted that this is a rapidly developing field, in particular the role of this protein module in blood-tissue barriers, and this short chapter attempts to provide the information necessary for investigators studying reproductive biology and blood-tissue barriers to design future studies. We also include results of recent studies from flies and worms since this information will be helpful in planning experiments for future functional studies in the testis to understand how Scribble-based proteins regulate BTB dynamics and spermatogenesis.
Collapse
|
38
|
The Scribble-Dlg-Lgl polarity module in development and cancer: from flies to man. Essays Biochem 2012; 53:141-68. [PMID: 22928514 DOI: 10.1042/bse0530141] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Scribble, Par and Crumbs modules were originally identified in the vinegar (fruit) fly, Drosophila melanogaster, as being critical regulators of apico-basal cell polarity. In the present chapter we focus on the Scribble polarity module, composed of Scribble, discs large and lethal giant larvae. Since the discovery of the role of the Scribble polarity module in apico-basal cell polarity, these proteins have also been recognized as having important roles in other forms of polarity, as well as regulation of the actin cytoskeleton, cell signalling and vesicular trafficking. In addition to these physiological roles, an important role for polarity proteins in cancer progression has also been uncovered, with loss of polarity and tissue architecture being strongly correlated with metastatic disease.
Collapse
|
39
|
Wan Q, Liu J, Zheng Z, Zhu H, Chu X, Dong Z, Huang S, Du Q. Regulation of myosin activation during cell-cell contact formation by Par3-Lgl antagonism: entosis without matrix detachment. Mol Biol Cell 2012; 23:2076-91. [PMID: 22496418 PMCID: PMC3364173 DOI: 10.1091/mbc.e11-11-0940] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Two polarity proteins, partitioning defective 3 homologue (Par3) and mammalian homologues of Drosophila lethal(2)giant larvae (Lgl1/2), antagonize each other in modulating myosin II activation during cell–cell contact formation in Madin-Darby canine kidney cells. Altering the counteraction between Par3 and Lgl1/2 leads to entosis without matrix detachment. Cell–cell contact formation following cadherin engagement requires actomyosin contraction along the periphery of cell–cell contact. The molecular mechanisms that regulate myosin activation during this process are not clear. In this paper, we show that two polarity proteins, partitioning defective 3 homologue (Par3) and mammalian homologues of Drosophila Lethal (2) Giant Larvae (Lgl1/2), antagonize each other in modulating myosin II activation during cell–cell contact formation in Madin-Darby canine kidney cells. While overexpression of Lgl1/2 or depletion of endogenous Par3 leads to enhanced myosin II activation, knockdown of Lgl1/2 does the opposite. Intriguingly, altering the counteraction between Par3 and Lgl1/2 induces cell–cell internalization during early cell–cell contact formation, which involves active invasion of the lateral cell–cell contact underneath the apical-junctional complexes and requires activation of the Rho–Rho-associated, coiled-coil containing protein kinase (ROCK)–myosin pathway. This is followed by predominantly nonapoptotic cell-in-cell death of the internalized cells and frequent aneuploidy of the host cells. Such effects are reminiscent of entosis, a recently described process observed when mammary gland epithelial cells were cultured in suspension. We propose that entosis could occur without matrix detachment and that overactivation of myosin or unbalanced myosin activation between contacting cells may be the driving force for entosis in epithelial cells.
Collapse
Affiliation(s)
- Qingwen Wan
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
In the Drosophila embryonic central nervous system, the neural precursor cells called neuroblasts undergo a number of asymmetric divisions along the apical-basal axis to give rise to different daughter cells of distinct fates. This review summarizes recent progress in understanding the mechanisms of these asymmetric cell divisions. We discuss proteins that are localized at distinct domains of cortex in the neuroblasts and their role in generating asymmetry. We also review uniformly cortical localized factors and actin cytoskeleton-associated motor proteins with regard to their potential role to serve as a link between distinct cortical domains in the neuroblasts. In this review, asymmetric divisions of sensory organ precursor and larval neuroblasts are also briefly discussed.
Collapse
Affiliation(s)
- Hongyan Wang
- Temasek Life Sciences Laboratory, 1 Research Link, The National University of Singapore, Singapore 117604
| | | |
Collapse
|
41
|
Dahan I, Yearim A, Touboul Y, Ravid S. The tumor suppressor Lgl1 regulates NMII-A cellular distribution and focal adhesion morphology to optimize cell migration. Mol Biol Cell 2012; 23:591-601. [PMID: 22219375 PMCID: PMC3279388 DOI: 10.1091/mbc.e11-01-0015] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The Drosophila tumor suppressor Lethal (2) giant larvae (Lgl) regulates the apical-basal polarity in epithelia and asymmetric cell division. However, little is known about the role of Lgl in cell polarity in migrating cells. In this study we show direct physiological interactions between the mammalian homologue of Lgl (Lgl1) and the nonmuscle myosin II isoform A (NMII-A). We demonstrate that Lgl1 and NMII-A form a complex in vivo and provide data that Lgl1 inhibits NMII-A filament assembly in vitro. Furthermore, depletion of Lgl1 results in the unexpected presence of NMII-A in the cell leading edge, a region that is not usually occupied by this protein, suggesting that Lgl1 regulates the cellular localization of NMII-A. Finally, we show that depletion of Lgl1 affects the size and number of focal adhesions, as well as cell polarity, membrane dynamics, and the rate of migrating cells. Collectively these findings indicate that Lgl1 regulates the polarity of migrating cells by controlling the assembly state of NMII-A, its cellular localization, and focal adhesion assembly.
Collapse
Affiliation(s)
- Inbal Dahan
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | | | | | | |
Collapse
|
42
|
Izumi Y, Yanagihashi Y, Furuse M. A novel protein complex, mesh-ssk, is required for septate junction formation in drosophila midgut. J Cell Sci 2012; 125:4923-33. [DOI: 10.1242/jcs.112243] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Septate junctions (SJs) are specialized intercellular junctions that restrict the free diffusion of solutes through the paracellular route in invertebrate epithelia. In arthropods, two morphologically different types of SJs have been reported: pleated SJs and smooth SJs (sSJs), which are found in ectodermally and endodermally derived epithelia, respectively. However, the molecular and functional differences between these SJ types have not been elucidated. Here we report that a novel sSJ-specific component, a single-pass transmembrane protein, termed ‘Mesh’ is highly concentrated in Drosophila sSJs. Compromised mesh expression causes defects in the organization of sSJs, in the localizations of other sSJ proteins, and in the barrier function of the midgut. Ectopic expression of Mesh in cultured cells induces cell-cell adhesion. Mesh forms a complex with Ssk, another sSJ-specific protein, and these proteins are mutually interdependent for their localization. Thus, a novel protein complex comprising Mesh and Ssk plays a significant role in sSJ formation and in intestinal barrier function in Drosophila.
Collapse
|
43
|
Froldi F, Ziosi M, Tomba G, Parisi F, Garoia F, Pession A, Grifoni D. Drosophila lethal giant larvae neoplastic mutant as a genetic tool for cancer modeling. Curr Genomics 2011; 9:147-54. [PMID: 19440511 PMCID: PMC2679652 DOI: 10.2174/138920208784340786] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Revised: 03/23/2008] [Accepted: 03/28/2008] [Indexed: 11/30/2022] Open
Abstract
Drosophila lethal giant larvae (lgl) is a tumour suppressor gene whose function in establishing apical-basal cell polarity as well as in exerting proliferation control in epithelial tissues is conserved between flies and mammals. Individuals bearing lgl null mutations show a gradual loss of tissue architecture and an extended larval life in which cell proliferation never ceases and no differentiation occurs, resulting in prepupal lethality. When tissues from those individuals are transplanted into adult normal recipients, a subset of cells, possibly the cancer stem units, are again able to proliferate and give rise to metastases which migrate to distant sites killing the host. This phenotype closely resembles that of mammalian epithelial cancers, in which loss of cell polarity is one of the hallmarks of a malignant, metastatic behaviour associated with poor prognosis. Lgl protein shares with its human counterpart Human giant larvae-1 (Hugl-1) significant stretches of sequence similarity that we demonstrated to translate into a complete functional conservation, pointing out a role in cell proliferation control and tumorigenesis also for the human homologue. The functional conservation and the power of fly genetics, that allows the researcher to manipulate the fly genome at a level of precision that exceeds that of any other multicellular genetic system, make this Drosophila mutant a very suitable model in which to investigate the mechanisms underlying epithelial tumour formation, progression and metastatisation. In this review, we will summarise the results obtained in these later years using this model for the study of cancer biology. Moreover, we will discuss how recent advances in developmental genetics techniques have succeeded in enhancing the similarities between fly and human tumorigenesis, giving Drosophila a pivotal role in the study of such a complex genetic disease.
Collapse
Affiliation(s)
- F Froldi
- Alma Mater Studiorum, Departments of Biologia Evoluzionistica Sperimentale and Patologia Sperimentale, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Laprise P, Tepass U. Novel insights into epithelial polarity proteins in Drosophila. Trends Cell Biol 2011; 21:401-8. [DOI: 10.1016/j.tcb.2011.03.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/18/2011] [Accepted: 03/21/2011] [Indexed: 01/04/2023]
|
45
|
Bukharina TA, Furman DP. Asymmetric cell division in the morphogenesis of Drosophila melanogaster macrochaetae. Russ J Dev Biol 2011. [DOI: 10.1134/s1062360411010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
46
|
Abstract
Cell polarity is essential for cells to divide asymmetrically, form spatially restricted subcellular structures and participate in three-dimensional multicellular organization. PAR proteins are conserved polarity regulators that function by generating cortical landmarks that establish dynamic asymmetries in the distribution of effector proteins. Here, we review recent findings on the role of PAR proteins in cell polarity in C. elegans and Drosophila, and emphasize the links that exist between PAR networks and cytoskeletal proteins that both regulate PAR protein localization and act as downstream effectors to elaborate polarity within the cell.
Collapse
Affiliation(s)
- Jeremy Nance
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute for Biomolecular Medicine, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA.
| | | |
Collapse
|
47
|
Knoblich JA. Asymmetric cell division: recent developments and their implications for tumour biology. Nat Rev Mol Cell Biol 2010; 11:849-60. [PMID: 21102610 DOI: 10.1038/nrm3010] [Citation(s) in RCA: 434] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ability of cells to divide asymmetrically is essential for generating diverse cell types during development. The past 10 years have seen tremendous progress in our understanding of this important biological process. We have learned that localized phosphorylation events are responsible for the asymmetric segregation of cell fate determinants in mitosis and that centrosomes and microtubules play important parts in this process. The relevance of asymmetric cell division for stem cell biology has added a new dimension to the field, and exciting connections between asymmetric cell division and tumorigenesis have begun to emerge.
Collapse
Affiliation(s)
- Juergen A Knoblich
- Institute of Molecular Biotechnology of Austrian Academy of Science, Doktor Bohr-Gasse 3, 1030 Vienna, Austria.
| |
Collapse
|
48
|
Abstract
The protein Lethal giant larvae (LGL) regulates cell polarity in diverse animal models. Now, an LGL orthologue has been identified in the worm Caenorhabditis elegans and is shown to function redundantly with a worm-specific polarity protein, PAR-2.
Collapse
Affiliation(s)
- Kenneth E Prehoda
- Institute of Molecular Biology, University of Oregon, 1370 Franklin Boulevard, Eugene, OR 97403, USA
| | | |
Collapse
|
49
|
Beatty A, Morton D, Kemphues K. The C. elegans homolog of Drosophila Lethal giant larvae functions redundantly with PAR-2 to maintain polarity in the early embryo. Development 2010; 137:3995-4004. [PMID: 21041363 DOI: 10.1242/dev.056028] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Polarity is essential for generating cell diversity. The one-cell C. elegans embryo serves as a model for studying the establishment and maintenance of polarity. In the early embryo, a myosin II-dependent contraction of the cortical meshwork asymmetrically distributes the highly conserved PDZ proteins PAR-3 and PAR-6, as well as an atypical protein kinase C (PKC-3), to the anterior. The RING-finger protein PAR-2 becomes enriched on the posterior cortex and prevents these three proteins from returning to the posterior. In addition to the PAR proteins, other proteins are required for polarity in many metazoans. One example is the conserved Drosophila tumor-suppressor protein Lethal giant larvae (Lgl). In Drosophila and mammals, Lgl contributes to the maintenance of cell polarity and plays a role in asymmetric cell division. We have found that the C. elegans homolog of Lgl, LGL-1, has a role in polarity but is not essential. It localizes asymmetrically to the posterior of the early embryo in a PKC-3-dependent manner, and functions redundantly with PAR-2 to maintain polarity. Furthermore, overexpression of LGL-1 is sufficient to rescue loss of PAR-2 function. LGL-1 negatively regulates the accumulation of myosin (NMY-2) on the posterior cortex, representing a possible mechanism by which LGL-1 might contribute to polarity maintenance.
Collapse
Affiliation(s)
- Alexander Beatty
- Department of Molecular Biology and Genetics, Cornell University, 433 Biotechnology Building, Ithaca, NY 14850, USA
| | | | | |
Collapse
|
50
|
Papagiannouli F, Mechler BM. Discs large in the Drosophila testis: an old player on a new task. Fly (Austin) 2010; 4:294-8. [PMID: 20798604 DOI: 10.4161/fly.4.4.13149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gamete development requires a coordinated soma-germ line interaction that ensures renewal and differentiation of germline and somatic stem cells. The physical contact between the germline and somatic cell populations is crucial because it allows the exchange of diffusible signals among them. The tumor suppressor gene discs large (dlg) encodes a septate junction protein with functions in epithelial cell polarity, asymmetric neuroblast division and formation of neuromuscular junctions. Our recent work reveals a new role of dlg in the Drosophila testis, as mutations in dlg lead to testis defects and cell death. Dlg is required throughout spermatogenesis in the somatic lineage and its localization changes from a uniform distribution along the plasma membrane of somatic cells in the testis apex, to a restricted localization on the distally located somatic cell in growing cysts. The extensive defects in dlg testis underline the importance of the somatic cells in the establishment and maintenance of the male stem cell niche and somatic cell differentiation. Here, we discuss our latest findings on the role of dlg in the Drosophila testis, supporting the view that junction proteins are dynamic structures, which can provide guiding cues to recruit scaffold proteins or other signaling molecules.
Collapse
Affiliation(s)
- Fani Papagiannouli
- Cell Networks-Cluster of Excellence and BIOQUANT Center, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|