1
|
van den Goor L, Iseler J, Koning KM, Miller AL. Mechanosensitive recruitment of Vinculin maintains junction integrity and barrier function at epithelial tricellular junctions. Curr Biol 2024; 34:4677-4691.e5. [PMID: 39341202 PMCID: PMC11496005 DOI: 10.1016/j.cub.2024.08.060] [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: 09/08/2023] [Revised: 07/26/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024]
Abstract
Apical cell-cell junctions, including adherens junctions and tight junctions, adhere epithelial cells to one another and regulate selective permeability at both bicellular junctions and tricellular junctions (TCJs). Although several specialized proteins are known to localize at TCJs, it remains unclear how actomyosin-mediated tension transmission at TCJs contributes to the maintenance of junction integrity and barrier function at these sites. Here, utilizing the embryonic epithelium of gastrula-stage Xenopus laevis embryos, we define a mechanism by which the mechanosensitive protein Vinculin helps anchor the actomyosin network at TCJs, thus maintaining TCJ integrity and barrier function. Using an optogenetic approach to acutely increase junctional tension, we find that Vinculin is mechanosensitively recruited to apical junctions immediately surrounding TCJs. In Vinculin knockdown (KD) embryos, junctional actomyosin intensity is decreased and becomes disorganized at TCJs. Using fluorescence recovery after photobleaching (FRAP), we show that Vinculin KD reduces actin stability at TCJs and destabilizes Angulin-1, a key tricellular tight junction protein involved in regulating barrier function at TCJs. When Vinculin KD embryos are subjected to increased tension, TCJ integrity is not maintained, filamentous actin (F-actin) morphology at TCJs is disrupted, and breaks in the signal of the tight junction protein ZO-1 signal are detected. Finally, using a live imaging barrier assay, we detect increased barrier leaks at TCJs in Vinculin KD embryos. Together, our findings show that Vinculin-mediated actomyosin organization is required to maintain junction integrity and barrier function at TCJs and reveal new information about the interplay between adhesion and barrier function at TCJs.
Collapse
Affiliation(s)
- Lotte van den Goor
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI 48109, USA
| | - Jolene Iseler
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI 48109, USA
| | - Katherine M Koning
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2
|
Ohashi K, Kunitomi A, Chiba S, Mizuno K. Roles of the Dbl family of RhoGEFs in mechanotransduction - a review. Front Cell Dev Biol 2024; 12:1485725. [PMID: 39479515 PMCID: PMC11521908 DOI: 10.3389/fcell.2024.1485725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Rho guanine nucleotide exchange factors (RhoGEFs) comprise a wide range of proteins with a common domain responsible for the activation of the Rho family of small GTPases and various domains in other regions. The evolutionary divergence of RhoGEFs enables actin cytoskeletal reorganization, leading to complex cellular responses in higher organisms. In this review, we address the involvement of RhoGEFs in the mechanical stress response of mammalian cells. The cellular mechanical stress response is essential for the proper and orderly regulation of cell populations, including the maintenance of homeostasis, tissue morphogenesis, and adaptation to the mechanical environment. In particular, this review focuses on the recent findings regarding the Dbl family of RhoGEFs involved in mechanical stress responses at the cell-cell and cell-substrate adhesion sites, and their molecular mechanisms underlying actin cytoskeleton remodeling and signal transduction.
Collapse
Affiliation(s)
- Kazumasa Ohashi
- Department of Molecular and Chemical Life Sciences, Laboratory of Molecular and Cellular Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | | | | | | |
Collapse
|
3
|
Lu M, Xu Z, Xu F, Yin C, Guo H, Cheng B. Mechanical network motifs as targets for mechanomedicine. Drug Discov Today 2024; 29:104145. [PMID: 39182599 DOI: 10.1016/j.drudis.2024.104145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The identification and analysis of network motifs has been widely used in the functional analysis of signaling components, disease discovery and other fields. The positive feedback loop (PFL) is a simple but important network motif. The formation of a PFL is regulated by mechanical cues such as substrate stiffness, fiber stretching and cell compression in the cell microenvironment. Here, we propose a new term, 'mechanical PFL', and analyze the mechanisms of mechanical PFLs at molecular, subcellular and cellular scales. More and more therapies are being targeted against mechanosignaling pathways at the experimental and preclinical stages, and exploring mechanical PFLs as potential mechanomedicine targets could be a new direction for disease treatment.
Collapse
Affiliation(s)
- Mengnan Lu
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Zhao Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China.
| | - Hui Guo
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China.
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
4
|
Shioka I, Morita R, Yagasaki R, Wuergezhen D, Yamashita T, Fujiwara H, Okuda S. Ex vivo SIM-AFM measurements reveal the spatial correlation of stiffness and molecular distributions in 3D living tissue. Acta Biomater 2024:S1742-7061(24)00539-7. [PMID: 39379233 DOI: 10.1016/j.actbio.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024]
Abstract
Living tissues each exhibit a distinct stiffness, which provides cells with key environmental cues that regulate their behaviors. Despite this significance, our understanding of the spatiotemporal dynamics and the biological roles of stiffness in three-dimensional tissues is currently limited due to a lack of appropriate measurement techniques. To address this issue, we propose a new method combining upright structured illumination microscopy (USIM) and atomic force microscopy (AFM) to obtain precisely coordinated stiffness maps and biomolecular fluorescence images of thick living tissue slices. Using mouse embryonic and adult skin as a representative tissue with mechanically heterogeneous structures inside, we validate the measurement principle of USIM-AFM. Live measurement of tissue stiffness distributions revealed the highly heterogeneous mechanical nature of skin, including nucleated/enucleated epithelium, mesenchyme, and hair follicle, as well as the role of collagens in maintaining its integrity. Furthermore, quantitative analysis comparing stiffness distributions in live tissue samples with those in preserved tissues, including formalin-fixed and cryopreserved tissue samples, unveiled the distinct impacts of preservation processes on tissue stiffness patterns. This series of experiments highlights the importance of live mechanical testing of tissue-scale samples to accurately capture the true spatiotemporal variations in mechanical properties. Our USIM-AFM technique provides a new methodology to reveal the dynamic nature of tissue stiffness and its correlation with biomolecular distributions in live tissues and thus could serve as a technical basis for exploring tissue-scale mechanobiology. STATEMENT OF SIGNIFICANCE: Stiffness, a simple mechanical parameter, has drawn attention in understanding the mechanobiological principles underlying the homeostasis and pathology of living tissues. To explore tissue-scale mechanobiology, we propose a technique integrating an upright structured illumination microscope and an atomic force microscope. This technique enables live measurements of stiffness distribution and fluorescent observation of thick living tissue slices. Experiments revealed the highly heterogeneous mechanical nature of mouse embryonic and adult skin in three dimensions and the previously unnoticed influences of preservation techniques on the mechanical properties of tissue at microscopic resolution. This study provides a new technical platform for live stiffness measurement and biomolecular observation of tissue-scale samples with micron-scale resolution, thus contributing to future studies of tissue- and organ-scale mechanobiology.
Collapse
Affiliation(s)
- Itsuki Shioka
- Graduate School of Frontier Science Initiative, Kanazawa University, Kanazawa 920-1192, Japan
| | - Ritsuko Morita
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Rei Yagasaki
- Nano Life Science Institute, Kanazawa University, Kanazawa 920-1192, Japan
| | - Duligengaowa Wuergezhen
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe 650-0047, Japan; Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Tadahiro Yamashita
- Department of System Design Engineering, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Hironobu Fujiwara
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe 650-0047, Japan; Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Satoru Okuda
- Nano Life Science Institute, Kanazawa University, Kanazawa 920-1192, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa 920-8640, Japan.
| |
Collapse
|
5
|
Uesugi K, Obata S, Nagayama K. Micro tensile tester measurement of biomechanical properties and adhesion force of microtubule-polymerization-inhibited cancer cells. J Mech Behav Biomed Mater 2024; 156:106586. [PMID: 38805872 DOI: 10.1016/j.jmbbm.2024.106586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/30/2024]
Abstract
Both mechanical and adhesion properties of cancer cells are complex and reciprocally related to migration, invasion, and metastasis with large cell deformation. Therefore, we evaluated these properties for human cervical cancer cells (HeLa) simultaneously using our previously developed micro tensile tester system. For efficient evaluation, we developed image analysis software to modify the system. The software can analyze the tensile force in real time. The modified system can evaluate the tensile stiffness of cells to which a large deformation is applied, also evaluate the adhesion strength of cancer cells that adhered to a culture substrate and were cultured for several days with their adhesion maturation. We used the modified system to simultaneously evaluate the stiffness of the cancer cells to which a large deformation was applied and their adhesion strength. The obtained results revealed that the middle phase of tensile stiffness and adhesion force of the microtubule-depolymerized group treated with colchicine (an anti-cancer drug) (stiffness, 13.4 ± 7.5 nN/%; adhesion force, 460.6 ± 258.2 nN) were over two times larger than those of the control group (stiffness, 5.0 ± 3.5 nN/%; adhesion force, 168.2 ± 98.0 nN). Additionally, the same trend was confirmed with the detailed evaluation of cell surface stiffness using an atomic force microscope. Confocal fluorescence microscope observations showed that the stress fibers (SFs) of colchicine-treated cells were aligned in the same direction, and focal adhesions (FAs) of the cells developed around both ends of the SFs and aligned parallel to the developed direction of the SFs. There was a possibility that the microtubule depolymerization by the colchicine treatment induced the development of SFs and FAs and subsequently caused an increment of cell stiffness and adhesion force. From the above results, we concluded the modified system would be applicable to cancer detection and anti-cancer drug efficacy tests.
Collapse
Affiliation(s)
- Kaoru Uesugi
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan
| | - Shota Obata
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan
| | - Kazuaki Nagayama
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan.
| |
Collapse
|
6
|
Lechuga S, Marino-Melendez A, Davis A, Zalavadia A, Khan A, Longworth MS, Ivanov AI. Coactosin-like protein 1 regulates integrity and repair of model intestinal epithelial barriers via actin binding dependent and independent mechanisms. Front Cell Dev Biol 2024; 12:1405454. [PMID: 39040043 PMCID: PMC11260685 DOI: 10.3389/fcell.2024.1405454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
The actin cytoskeleton regulates the integrity and repair of epithelial barriers by mediating the assembly of tight junctions (TJs), and adherens junctions (AJs), and driving epithelial wound healing. Actin filaments undergo a constant turnover guided by numerous actin-binding proteins, however, the roles of actin filament dynamics in regulating intestinal epithelial barrier integrity and repair remain poorly understood. Coactosin-like protein 1 (COTL1) is a member of the ADF/cofilin homology domain protein superfamily that binds and stabilizes actin filaments. COTL1 is essential for neuronal and cancer cell migration, however, its functions in epithelia remain unknown. The goal of this study is to investigate the roles of COTL1 in regulating the structure, permeability, and repair of the epithelial barrier in human intestinal epithelial cells (IEC). COTL1 was found to be enriched at apical junctions in polarized IEC monolayers in vitro. The knockdown of COTL1 in IEC significantly increased paracellular permeability, impaired the steady state TJ and AJ integrity, and attenuated junctional reassembly in a calcium-switch model. Consistently, downregulation of COTL1 expression in Drosophila melanogaster increased gut permeability. Loss of COTL1 attenuated collective IEC migration and decreased cell-matrix attachment. The observed junctional abnormalities in COTL1-depleted IEC were accompanied by the impaired assembly of the cortical actomyosin cytoskeleton. Overexpression of either wild-type COTL1 or its actin-binding deficient mutant tightened the paracellular barrier and activated junction-associated myosin II. Furthermore, the actin-uncoupled COTL1 mutant inhibited epithelial migration and matrix attachment. These findings highlight COTL1 as a novel regulator of the intestinal epithelial barrier integrity and repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
7
|
Kunitomi A, Chiba S, Higashitani N, Higashitani A, Sato S, Mizuno K, Ohashi K. Solo regulates the localization and activity of PDZ-RhoGEF for actin cytoskeletal remodeling in response to substrate stiffness. Mol Biol Cell 2024; 35:ar87. [PMID: 38656797 PMCID: PMC11238083 DOI: 10.1091/mbc.e23-11-0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
Recent findings indicate that Solo, a RhoGEF, is involved in cellular mechanical stress responses. However, the mechanism of actin cytoskeletal remodeling via Solo remains unclear. Therefore, this study aimed to identify Solo-interacting proteins using the BioID, a proximal-dependent labeling method, and elucidate the molecular mechanisms of function of Solo. We identified PDZ-RhoGEF (PRG) as a Solo-interacting protein. PRG colocalized with Solo in the basal area of cells, depending on Solo localization, and enhanced actin polymerization at the Solo accumulation sites. Additionally, Solo and PRG interaction was necessary for actin cytoskeletal remodeling. Furthermore, the purified Solo itself had little or negligible GEF activity, even its GEF-inactive mutant directly activated the GEF activity of PRG through interaction. Moreover, overexpression of the Solo and PRG binding domains, respectively, had a dominant-negative effect on actin polymerization and actin stress fiber formation in response to substrate stiffness. Therefore, Solo restricts the localization of PRG and regulates actin cytoskeletal remodeling in synergy with PRG in response to the surrounding mechanical environment.
Collapse
Affiliation(s)
- Aoi Kunitomi
- Laboratory of Molecular and Cellular Biology, Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shuhei Chiba
- Laboratory of Molecular and Cellular Biology, Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Nahoko Higashitani
- Laboratory of Molecular Genetics and Physiology, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Atsushi Higashitani
- Laboratory of Molecular Genetics and Physiology, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Shinichi Sato
- Laboratory of Bioactive Molecules, Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Kensaku Mizuno
- Laboratory of Molecular and Cellular Biology, Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Kazumasa Ohashi
- Laboratory of Molecular and Cellular Biology, Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
8
|
Nguyen TP, Otani T, Tsutsumi M, Kinoshita N, Fujiwara S, Nemoto T, Fujimori T, Furuse M. Tight junction membrane proteins regulate the mechanical resistance of the apical junctional complex. J Cell Biol 2024; 223:e202307104. [PMID: 38517380 PMCID: PMC10959758 DOI: 10.1083/jcb.202307104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/29/2024] [Accepted: 02/16/2024] [Indexed: 03/23/2024] Open
Abstract
Epithelia must be able to resist mechanical force to preserve tissue integrity. While intercellular junctions are known to be important for the mechanical resistance of epithelia, the roles of tight junctions (TJs) remain to be established. We previously demonstrated that epithelial cells devoid of the TJ membrane proteins claudins and JAM-A completely lack TJs and exhibit focal breakages of their apical junctions. Here, we demonstrate that apical junctions fracture when claudin/JAM-A-deficient cells undergo spontaneous cell stretching. The junction fracture was accompanied by actin disorganization, and actin polymerization was required for apical junction integrity in the claudin/JAM-A-deficient cells. Further deletion of CAR resulted in the disruption of ZO-1 molecule ordering at cell junctions, accompanied by severe defects in apical junction integrity. These results demonstrate that TJ membrane proteins regulate the mechanical resistance of the apical junctional complex in epithelial cells.
Collapse
Affiliation(s)
- Thanh Phuong Nguyen
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Kawaguchi, Japan
| | - Motosuke Tsutsumi
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Noriyuki Kinoshita
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Sachiko Fujiwara
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tomomi Nemoto
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
9
|
Morel A, Douat C, Blangy A, Vives V. Bone resorption by osteoclasts involves fine tuning of RHOA activity by its microtubule-associated exchange factor GEF-H1. Front Physiol 2024; 15:1342024. [PMID: 38312316 PMCID: PMC10834693 DOI: 10.3389/fphys.2024.1342024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Bone health is controlled by the balance between bone formation by osteoblasts and degradation by osteoclasts. A disequilibrium in favor of bone resorption leads to osteolytic diseases characterized by decreased bone density. Osteoclastic resorption is dependent on the assembly of an adhesion structure: the actin ring, also called podosome belt or sealing zone, which is composed of a unique patterning of podosomes stabilized by microtubules. A better understanding of the molecular mechanisms regulating the crosstalk between actin cytoskeleton and microtubules network is key to find new treatments to inhibit bone resorption. Evidence points to the importance of the fine tuning of the activity of the small GTPase RHOA for the formation and maintenance of the actin ring, but the underlying mechanism is not known. We report here that actin ring disorganization upon microtubule depolymerization is mediated by the activation of the RHOA-ROCK signaling pathway. We next show the involvement of GEF-H1, one of RHOA guanine exchange factor highly expressed in osteoclasts, which has the particularity of being negatively regulated by sequestration on microtubules. Using a CRISPR/Cas9-mediated GEF-H1 knock-down osteoclast model, we demonstrate that RHOA activation upon microtubule depolymerization is mediated by GEF-H1 release. Interestingly, although lower levels of GEF-H1 did not impact sealing zone formation in the presence of an intact microtubule network, sealing zone was smaller leading to impaired resorption. Altogether, these results suggest that a fine tuning of GEF-H1 through its association with microtubules, and consequently of RHOA activity, is essential for osteoclast sealing zone stability and resorption function.
Collapse
Affiliation(s)
- Anne Morel
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Christophe Douat
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Anne Blangy
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Virginie Vives
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| |
Collapse
|
10
|
Prakash A, Paunikar S, Webber M, McDermott E, Vellanki SH, Thompson K, Dockery P, Jahns H, Brown JAL, Hopkins AM, Bourke E. Centrosome amplification promotes cell invasion via cell-cell contact disruption and Rap-1 activation. J Cell Sci 2023; 136:jcs261150. [PMID: 37772773 PMCID: PMC10629695 DOI: 10.1242/jcs.261150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023] Open
Abstract
Centrosome amplification (CA) is a prominent feature of human cancers linked to tumorigenesis in vivo. Here, we report mechanistic contributions of CA induction alone to tumour architecture and extracellular matrix (ECM) remodelling. CA induction in non-tumorigenic breast cells MCF10A causes cell migration and invasion, with underlying disruption of epithelial cell-cell junction integrity and dysregulation of expression and subcellular localisation of cell junction proteins. CA also elevates expression of integrin β-3, its binding partner fibronectin-1 and matrix metalloproteinase enzymes, promoting cell-ECM attachment, ECM degradation, and a migratory and invasive cell phenotype. Using a chicken embryo xenograft model for in vivo validation, we show that CA-induced (+CA) MCF10A cells invade into the chick mesodermal layer, with inflammatory cell infiltration and marked focal reactions between chorioallantoic membrane and cell graft. We also demonstrate a key role of small GTPase Rap-1 signalling through inhibition using GGTI-298, which blocked various CA-induced effects. These insights reveal that in normal cells, CA induction alone (without additional oncogenic alterations) is sufficient to confer early pro-tumorigenic changes within days, acting through Rap-1-dependent signalling to alter cell-cell contacts and ECM disruption.
Collapse
Affiliation(s)
- Anu Prakash
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Shishir Paunikar
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Mark Webber
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Emma McDermott
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Sri H. Vellanki
- Department of Surgery, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin D09 DK19, Ireland
| | - Kerry Thompson
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Peter Dockery
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Hanne Jahns
- Pathobiology Section, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - James A. L. Brown
- Department of Biological Sciences, University of Limerick, Limerick V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC) and Health Research Institute, University of Limerick, Limerick V94T9PX, Ireland
| | - Ann M. Hopkins
- Department of Surgery, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin D09 DK19, Ireland
| | - Emer Bourke
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| |
Collapse
|
11
|
Wang D, Wang Y, Di X, Wang F, Wanninayaka A, Carnell M, Hardeman EC, Jin D, Gunning PW. Cortical tension drug screen links mitotic spindle integrity to Rho pathway. Curr Biol 2023; 33:4458-4469.e4. [PMID: 37875071 DOI: 10.1016/j.cub.2023.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 07/24/2023] [Accepted: 09/11/2023] [Indexed: 10/26/2023]
Abstract
Mechanical force generation plays an essential role in many cellular functions, including mitosis. Actomyosin contractile forces mediate changes in cell shape in mitosis and are implicated in mitotic spindle integrity via cortical tension. An unbiased screen of 150 small molecules that impact actin organization and 32 anti-mitotic drugs identified two molecular targets, Rho kinase (ROCK) and tropomyosin 3.1/2 (Tpm3.1/2), whose inhibition has the greatest impact on mitotic cortical tension. The converse was found for compounds that depolymerize microtubules. Tpm3.1/2 forms a co-polymer with mitotic cortical actin filaments, and its inhibition prevents rescue of multipolar spindles induced by anti-microtubule chemotherapeutics. We examined the role of mitotic cortical tension in this rescue mechanism. Inhibition of ROCK and Tpm3.1/2 and knockdown (KD) of cortical nonmuscle myosin 2A (NM2A), all of which reduce cortical tension, inhibited rescue of multipolar mitotic spindles, further implicating cortical tension in the rescue mechanism. GEF-H1 released from microtubules by depolymerization increased cortical tension through the RhoA pathway, and its KD also inhibited rescue of multipolar mitotic spindles. We conclude that microtubule depolymerization by anti-cancer drugs induces cortical-tension-based rescue to ensure integrity of the mitotic bipolar spindle mediated via the RhoA pathway. Central to this mechanism is the dependence of NM2A on Tpm3.1/2 to produce the functional engagement of actin filaments responsible for cortical tension.
Collapse
Affiliation(s)
- Dejiang Wang
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Yao Wang
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Xiangjun Di
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Fan Wang
- School of Electrical and Data Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW 2007, Australia; School of Physics, Beihang University, Beijing 100191, P.R. China
| | - Amanda Wanninayaka
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Michael Carnell
- Katharina Gaus Light Microscope Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Edna C Hardeman
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Dayong Jin
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; UTS-SUStech Joint Research Centre for Biomedical Materials & Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, P.R. China
| | - Peter W Gunning
- School of Biomedical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
12
|
Abraham A, Virdi S, Herrero N, Bryant I, Nwakama C, Jacob M, Khaparde G, Jordan D, McCuddin M, McKinley S, Taylor A, Peeples C, Ekpenyong A. Microfluidic Microcirculation Mimetic for Exploring Biophysical Mechanisms of Chemotherapy-Induced Metastasis. MICROMACHINES 2023; 14:1653. [PMID: 37763816 PMCID: PMC10536821 DOI: 10.3390/mi14091653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023]
Abstract
There is rapidly emerging evidence from pre-clinical studies, patient samples and patient subpopulations that certain chemotherapeutics inadvertently produce prometastatic effects. Prior to this, we showed that doxorubicin and daunorubicin stiffen cells before causing cell death, predisposing the cells to clogging and extravasation, the latter being a step in metastasis. Here, we investigate which other anti-cancer drugs might have similar prometastatic effects by altering the biophysical properties of cells. We treated myelogenous (K562) leukemic cancer cells with the drugs nocodazole and hydroxyurea and then measured their mechanical properties using a microfluidic microcirculation mimetic (MMM) device, which mimics aspects of blood circulation and enables the measurement of cell mechanical properties via transit times through the device. We also quantified the morphological properties of cells to explore biophysical mechanisms underlying the MMM results. Results from MMM measurements show that nocodazole- and hydroxyurea-treated K562 cells exhibit significantly altered transit times. Nocodazole caused a significant (p < 0.01) increase in transit times, implying a stiffening of cells. This work shows the feasibility of using an MMM to explore possible biophysical mechanisms that might contribute to chemotherapy-induced metastasis. Our work also suggests cell mechanics as a therapeutic target for much needed antimetastatic strategies in general.
Collapse
Affiliation(s)
- Ashley Abraham
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Sukhman Virdi
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Nick Herrero
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Israel Bryant
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Chisom Nwakama
- Chemistry Department, Creighton University, Omaha, NE 68178, USA;
| | - Megha Jacob
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Gargee Khaparde
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Destiny Jordan
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Mackenzie McCuddin
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Spencer McKinley
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Adam Taylor
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Conner Peeples
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Andrew Ekpenyong
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| |
Collapse
|
13
|
Guru A, Saravanan S, Sharma D, Narasimha M. The microtubule end-binding proteins EB1 and Patronin modulate the spatiotemporal dynamics of myosin and pattern pulsed apical constriction. Development 2022; 149:284823. [PMID: 36440630 DOI: 10.1242/dev.199759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 10/31/2022] [Indexed: 11/29/2022]
Abstract
Apical constriction powers amnioserosa contraction during Drosophila dorsal closure. The nucleation, movement and dispersal of apicomedial actomyosin complexes generates pulsed apical constrictions during early closure. Persistent apicomedial and circumapical actomyosin complexes drive unpulsed constrictions that follow. Here, we show that the microtubule end-binding proteins EB1 and Patronin pattern constriction dynamics and contraction kinetics by coordinating the balance of actomyosin forces in the apical plane. We find that microtubule growth from moving Patronin platforms governs the spatiotemporal dynamics of apicomedial myosin through the regulation of RhoGTPase signaling by transient EB1-RhoGEF2 interactions. We uncover the dynamic reorganization of a subset of short non-centrosomally nucleated apical microtubules that surround the coalescing apicomedial myosin complex, trail behind it as it moves and disperse as the complex dissolves. We demonstrate that apical microtubule reorganization is sensitive to Patronin levels. Microtubule depolymerization compromised apical myosin enrichment and altered constriction dynamics. Together, our findings uncover the importance of reorganization of an intact apical microtubule meshwork, by moving Patronin platforms and growing microtubule ends, in enabling the spatiotemporal modulation of actomyosin contractility and, through it, apical constriction.
Collapse
Affiliation(s)
- Anwesha Guru
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai 400005, India
| | - Surat Saravanan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai 400005, India
| | - Deepanshu Sharma
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai 400005, India
| | - Maithreyi Narasimha
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai 400005, India
| |
Collapse
|
14
|
Takeichi M. Cell sorting in vitro and in vivo: How are cadherins involved? Semin Cell Dev Biol 2022; 147:2-11. [PMID: 36376196 DOI: 10.1016/j.semcdb.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Animal tissues are composed of heterogenous cells, and their sorting into different compartments of the tissue is a pivotal process for organogenesis. Cells accomplish sorting by themselves-it is well known that singly dispersed cells can self-organize into tissue-like structures in vitro. Cell sorting is regulated by both biochemical and physical mechanisms. Adhesive proteins connect cells together, selecting particular partners through their specific binding properties, while physical forces, such as cell-cortical tension, control the cohesiveness between cells and in turn cell assembly patterns in mechanical ways. These processes cooperate in determining the overall cell sorting behavior. This article focuses on the 'cadherin' family of adhesion molecules as a biochemical component of cell-cell interactions, addressing how they regulate cell sorting by themselves or by cooperating with other factors. New ideas beyond the classical models of cell sorting are also discussed.
Collapse
|
15
|
Lu ZY, Feng L, Jiang WD, Wu P, Liu Y, Jin XW, Ren HM, Kuang SY, Li SW, Tang L, Zhang L, Mi HF, Zhou XQ. An Antioxidant Supplement Function Exploration: Rescue of Intestinal Structure Injury by Mannan Oligosaccharides after Aeromonas hydrophila Infection in Grass Carp ( Ctenopharyngodon idella). Antioxidants (Basel) 2022; 11:antiox11050806. [PMID: 35624670 PMCID: PMC9137958 DOI: 10.3390/antiox11050806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/26/2022] Open
Abstract
Mannan oligosaccharides (MOS) are a type of functional oligosaccharide which have received increased attention because of their beneficial effects on fish intestinal health. However, intestinal structural integrity is a necessary prerequisite for intestinal health. This study focused on exploring the protective effects of dietary MOS supplementation on the grass carp’s (Ctenopharyngodon idella) intestinal structural integrity (including tight junction (TJ) and adherent junction (AJ)) and its related signalling molecule mechanism. A total of 540 grass carp (215.85 ± 0.30 g) were fed six diets containing graded levels of dietary MOS supplementation (0, 200, 400, 600, 800 and 1000 mg/kg) for 60 days. Subsequently, a challenge test was conducted by injection of Aeromonas hydrophila for 14 days. We used ELISA, spectrophotometry, transmission electron microscope, immunohistochemistry, qRT-PCR and Western blotting to determine the effect of dietary MOS supplementation on intestinal structural integrity and antioxidant capacity. The results revealed that dietary MOS supplementation protected the microvillus of the intestine; reduced serum diamine oxidase and d-lactate levels (p < 0.05); enhanced intestinal total antioxidant capacity (p < 0.01); up-regulated most intestinal TJ and AJ mRNA levels; and decreased GTP-RhoA protein levels (p < 0.01). In addition, we also found several interesting results suggesting that MOS supplementation has no effects on ZO-2 and Claudin-15b. Overall, these findings suggested that dietary MOS supplementation could protect intestinal ultrastructure, reduce intestinal mucosal permeability and maintain intestinal structural integrity via inhibiting MLCK and RhoA/ROCK signalling pathways.
Collapse
Affiliation(s)
- Zhi-Yuan Lu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co., Ltd., Chengdu 610066, China; (S.-Y.K.); (S.-W.L.); (L.T.)
| | - Shu-Wei Li
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co., Ltd., Chengdu 610066, China; (S.-Y.K.); (S.-W.L.); (L.T.)
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co., Ltd., Chengdu 610066, China; (S.-Y.K.); (S.-W.L.); (L.T.)
| | - Lu Zhang
- Healthy Aquaculture Key Laboratory of Sichuan Province, Tongwei Co., Ltd., Chengdu 610041, China; (L.Z.); (H.-F.M.)
| | - Hai-Feng Mi
- Healthy Aquaculture Key Laboratory of Sichuan Province, Tongwei Co., Ltd., Chengdu 610041, China; (L.Z.); (H.-F.M.)
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (Z.-Y.L.); (L.F.); (W.-D.J.); (P.W.); (Y.L.); (X.-W.J.); (H.-M.R.)
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence:
| |
Collapse
|
16
|
Loss of KAP3 decreases intercellular adhesion and impairs intracellular transport of laminin in signet ring cell carcinoma of the stomach. Sci Rep 2022; 12:5050. [PMID: 35322078 PMCID: PMC8943207 DOI: 10.1038/s41598-022-08904-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/14/2022] [Indexed: 12/14/2022] Open
Abstract
Signet-ring cell carcinoma (SRCC) is a unique subtype of gastric cancer that is impaired for cell-cell adhesion. The pathogenesis of SRCC remains unclear. Here, we show that expression of kinesin-associated protein 3 (KAP3), a cargo adaptor subunit of the kinesin superfamily protein 3 (KIF3), a motor protein, is specifically decreased in SRCC of the stomach. CRISPR/Cas9-mediated gene knockout experiments indicated that loss of KAP3 impairs the formation of circumferential actomyosin cables by inactivating RhoA, leading to the weakening of cell-cell adhesion. Furthermore, in KAP3 knockout cells, post-Golgi transport of laminin, a key component of the basement membrane, was inhibited, resulting in impaired basement membrane formation. Together, these findings uncover a potential role for KAP3 in the pathogenesis of SRCC of the stomach.
Collapse
|
17
|
Kinoshita N, Yamamoto TS, Yasue N, Takagi C, Fujimori T, Ueno N. Force-dependent remodeling of cytoplasmic ZO-1 condensates contributes to cell-cell adhesion through enhancing tight junctions. iScience 2022; 25:103846. [PMID: 35198899 PMCID: PMC8850805 DOI: 10.1016/j.isci.2022.103846] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/22/2021] [Accepted: 01/25/2022] [Indexed: 12/01/2022] Open
Abstract
The physiological importance of biomolecular condensates is widely recognized, but how it is controlled in time and space during development is largely unknown. Here, we show that a tight junction protein ZO-1 forms cytoplasmic condensates in the trophectoderm (TE) of the mouse embryo before E4.0. These disappear via dissolution, and ZO-1 accumulates at the cell junction as the blastocyst cavity grows and internal pressure on TE cells increases. In contrast, this dissolution was less evident in TE cells attached to the inner cell mass because they receive weaker tensile forces. Furthermore, analyses using MDCK cells demonstrated that the ZO-1 condensates are generated and maintained by liquid-liquid phase separation. Our study also highlights that the dynamics of these condensates depends on the physical environment via an interaction between ZO-1 and F-actin. We propose that the force-dependent regulation of ZO-1 condensation contributes to the establishment of robust cell-cell adhesion during early development. ZO-1 forms cytoplasmic droplets via liquid-liquid phase separation In hatching mouse embryos, ZO-1 droplets dissolve and it localizes to cell junctions In MDCK cells, ZO-1 forms droplets in response to mechanical environments Interaction with F-actin negatively regulates ZO-1 phase separation
Collapse
Affiliation(s)
- Noriyuki Kinoshita
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, Aichi 444-8585, Japan.,School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Takamasa S Yamamoto
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Naoko Yasue
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Chiyo Takagi
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Toshihiko Fujimori
- Division of Embryology, Department of Developmental Biology, National Institute for Basic Biology, 5-1 Higashiyama Myodaiji, Okazaki, Aichi 444-8787, Japan.,School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Naoto Ueno
- Division of Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, Aichi 444-8585, Japan.,School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan.,Unit of Quantitative and Imaging Biology, International Research Collaboration Center, National Institutes of Natural Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
18
|
Stoiber P, Scribani Rossi P, Pokharel N, Germany JL, York EA, Schaus SE, Hansen U. Factor quinolinone inhibitors alter cell morphology and motility by destabilizing interphase microtubules. Sci Rep 2021; 11:23564. [PMID: 34876605 PMCID: PMC8651680 DOI: 10.1038/s41598-021-02962-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022] Open
Abstract
Factor quinolinone inhibitors are promising anti-cancer compounds, initially characterized as specific inhibitors of the oncogenic transcription factor LSF (TFCP2). These compounds exert anti-proliferative activity at least in part by disrupting mitotic spindles. Herein, we report additional interphase consequences of the initial lead compound, FQI1, in two telomerase immortalized cell lines. Within minutes of FQI1 addition, the microtubule network is disrupted, resulting in a substantial, although not complete, depletion of microtubules as evidenced both by microtubule sedimentation assays and microscopy. Surprisingly, this microtubule breakdown is quickly followed by an increase in tubulin acetylation in the remaining microtubules. The sudden breakdown and partial depolymerization of the microtubule network precedes FQI1-induced morphological changes. These involve rapid reduction of cell spreading of interphase fetal hepatocytes and increase in circularity of retinal pigment epithelial cells. Microtubule depolymerization gives rise to FH-B cell compaction, as pretreatment with taxol prevents this morphological change. Finally, FQI1 decreases the rate and range of locomotion of interphase cells, supporting an impact of FQI1-induced microtubule breakdown on cell motility. Taken together, our results show that FQI1 interferes with microtubule-associated functions in interphase, specifically cell morphology and motility.
Collapse
Affiliation(s)
- Patrick Stoiber
- grid.189504.10000 0004 1936 7558MCBB Graduate Program, Boston University, Boston, MA 02215 USA ,grid.189504.10000 0004 1936 7558Department of Biology, Boston University, Boston, MA 02215 USA
| | - Pietro Scribani Rossi
- grid.189504.10000 0004 1936 7558Department of Biology, Boston University, Boston, MA 02215 USA ,grid.7841.aPresent Address: Faculty of Medicine and Dentistry, Sapienza University of Rome, 00185 Rome, Italy
| | - Niranjana Pokharel
- grid.189504.10000 0004 1936 7558Department of Chemistry, Boston University, Boston, MA 02215 USA ,grid.189504.10000 0004 1936 7558Center for Molecular Discovery, Boston University, Boston, MA 02215 USA
| | - Jean-Luc Germany
- grid.189504.10000 0004 1936 7558Department of Biology, Boston University, Boston, MA 02215 USA
| | - Emily A. York
- grid.189504.10000 0004 1936 7558Department of Chemistry, Boston University, Boston, MA 02215 USA ,grid.189504.10000 0004 1936 7558Center for Molecular Discovery, Boston University, Boston, MA 02215 USA
| | - Scott E. Schaus
- grid.189504.10000 0004 1936 7558Department of Chemistry, Boston University, Boston, MA 02215 USA ,grid.189504.10000 0004 1936 7558Center for Molecular Discovery, Boston University, Boston, MA 02215 USA
| | - Ulla Hansen
- MCBB Graduate Program, Boston University, Boston, MA, 02215, USA. .,Department of Biology, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
19
|
Shang BB, Chen J, Wang ZG, Liu H. Significant correlation between HSPA4 and prognosis and immune regulation in hepatocellular carcinoma. PeerJ 2021; 9:e12315. [PMID: 34754620 PMCID: PMC8555498 DOI: 10.7717/peerj.12315] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/24/2021] [Indexed: 12/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is an inflammation-associated tumor involved in immune tolerance and evasion in the immune microenvironment. Heat shock proteins (HSPs) are involved in the occurrence, progression, and immune regulation of tumors. Therefore, HSPs have been considered potential therapeutic targets. Here, we aimed to elucidate the value of HSP family A (Hsp70) member 4 (HSPA4) in the diagnosis and predicting prognosis of HCC, and its relationship with immune cell infiltration, immune cell biomarkers, and immune checkpoints. Gene mutation, DNA methylation, and the pathway involved in HCC were also analyzed. Methods The gene expression omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases were used to compare HSPA4 expression, and the results were confirmed by immunohistochemical staining of clinical samples. R package was used to analyze the correlation between HSPA4 and cancer stage, and to establish receiver operating characteristic (ROC) curve of diagnosis, time-dependent survival ROC curve, and a nomogram model. cBioPortal and MethSurv were used to identify genetic alterations and DNA methylation, and their effect on prognosis. The Tumor Immune Estimation Resource (TIMER) was used to analyze immune cell infiltration, immune cell biomarkers, and immune checkpoints. The STRING database was used to analyze protein-protein interaction network information. Gene Ontology (GO) analysis and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to investigate the functions of HSPA4 and its functional partner genes. Results Overexpression of HSPA4 was identified in 25 cancers. Overexpression of HSPA4 considerably correlated with cancer stage and alpha-fetoprotein (AFP) level in HCC. Patients with higher HSPA4 expression showed poorer prognosis. HSPA4 expression can accurately identify tumor from normal tissue (AUC = 0.957). The area under 1-, 3-, and 5-year survival ROCs were above 0.6. The HSPA4 genetic alteration rate was 1.3%. Among the 14 DNA methylation CpG sites, seven were related to the prognosis of HCC. HSPA4 was positively related to immune cell infiltration and immune checkpoints (PD-1 and CTLA-4) in HCC. The KEGG pathway enrichment analysis revealed HSPA4 enrichment in antigen processing and presentation together with HSPA8 and HSP90AA1. We verified the value of HSPA4 in the diagnosis and predicting prognosis of HCC. HSPA4 may not only participate in the occurrence and progression but also the immune regulation of HCC. Therefore, HSPA4 can be a potential diagnostic and prognostic biomarker and a therapeutic target for HCC.
Collapse
Affiliation(s)
| | - Jun Chen
- Laboratory Animal Center, Dalian Medical University, Dalian, China
| | - Zhi-Guo Wang
- Second Hospital of Dalian Medical University, Dalian, China
| | - Hui Liu
- Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
20
|
Terada M, Ide S, Naito T, Kimura N, Matsusaki M, Kaji N. Label-Free Cancer Stem-like Cell Assay Conducted at a Single Cell Level Using Microfluidic Mechanotyping Devices. Anal Chem 2021; 93:14409-14416. [PMID: 34628861 DOI: 10.1021/acs.analchem.1c02316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mechanical phenotype of cells is an intrinsic property of individual cells. In fact, this property could serve as a label-free, non-destructive, diagnostic marker of the state of cells owing to its remarkable translational potential. A microfluidic device is a strong candidate for meeting the demand of this translational research as it can be used to diagnose a large population of cells at a single cell level in a high-throughput manner, without the need for off-line pretreatment operations. In this study, we investigated the mechanical phenotype of the human colon adenocarcinoma cell, HT29, which is known to be a heterogeneous cell line with both multipotency and self-renewal abilities. This type of cancer stem-like cell (CSC) is believed to be the unique originators of all tumor cells and may serve as the leading cause of cancer metastasis and drug resistance. By combining consecutive constrictions and microchannels with an ionic current sensing system, we found a high heterogeneity of cell deformability in the population of HT29 cells. Moreover, based on the level of aldehyde dehydrogenase (ALDH) activity and the expression level of CD44s, which are biochemical markers that suggest the multipotency of cells, the high heterogeneity of cell deformability was concluded to be a potential mechanical marker of CSCs. The development of label-free and non-destructive identification and collection techniques for CSCs has remarkable potential not only for cancer diagnosis and prognosis but also for the discovery of a new treatment for cancer.
Collapse
Affiliation(s)
- Miyu Terada
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Sachiko Ide
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Toyohiro Naito
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Niko Kimura
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Noritada Kaji
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya 464-8603, Japan
| |
Collapse
|
21
|
Joy-Immediato M, Ramirez MJ, Cerda M, Toyama Y, Ravasio A, Kanchanawong P, Bertocchi C. Junctional ER Organization Affects Mechanotransduction at Cadherin-Mediated Adhesions. Front Cell Dev Biol 2021; 9:669086. [PMID: 34222239 PMCID: PMC8247578 DOI: 10.3389/fcell.2021.669086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/23/2021] [Indexed: 11/13/2022] Open
Abstract
Cadherin-mediated adhesions (also known as adherens junctions) are adhesive complexes that connect neighboring cells in a tissue. While the role of the actin cytoskeleton in withstanding tension at these sites of contact is well documented, little is known about the involvement of microtubules and the associated endoplasmic reticulum (ER) network in cadherin mechanotransduction. Therefore, we investigated how the organization of ER extensions in close proximity of cadherin-mediated adhesions can affect such complexes, and vice versa. Here, we show that the extension of the ER to cadherin-mediated adhesions is tension dependent and appears to be cadherin-type specific. Furthermore, the different structural organization of the ER/microtubule network seems to affect the localization of ER-bound PTP1B at cadherin-mediated adhesions. This phosphatase is involved in the modulation of vinculin, a molecular clutch which enables differential engagement of the cadherin-catenin layer with the actomyosin cytoskeleton in response to tension. This suggests a link between structural organization of the ER/microtubule network around cadherin-specific adhesions, to control the mechanotransduction of adherens junctions by modulation of vinculin conformational state.
Collapse
Affiliation(s)
- Michelle Joy-Immediato
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel J Ramirez
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mauricio Cerda
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Center for Medical Informatics and Telemedicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Yusuke Toyama
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Andrea Ravasio
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
22
|
Monster JL, Donker L, Vliem MJ, Win Z, Matthews HK, Cheah JS, Yamada S, de Rooij J, Baum B, Gloerich M. An asymmetric junctional mechanoresponse coordinates mitotic rounding with epithelial integrity. J Cell Biol 2021; 220:e202001042. [PMID: 33688935 PMCID: PMC7953256 DOI: 10.1083/jcb.202001042] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 12/23/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Epithelia are continuously self-renewed, but how epithelial integrity is maintained during the morphological changes that cells undergo in mitosis is not well understood. Here, we show that as epithelial cells round up when they enter mitosis, they exert tensile forces on neighboring cells. We find that mitotic cell-cell junctions withstand these tensile forces through the mechanosensitive recruitment of the actin-binding protein vinculin to cadherin-based adhesions. Surprisingly, vinculin that is recruited to mitotic junctions originates selectively from the neighbors of mitotic cells, resulting in an asymmetric composition of cadherin junctions. Inhibition of junctional vinculin recruitment in neighbors of mitotic cells results in junctional breakage and weakened epithelial barrier. Conversely, the absence of vinculin from the cadherin complex in mitotic cells is necessary to successfully undergo mitotic rounding. Our data thus identify an asymmetric mechanoresponse at cadherin adhesions during mitosis, which is essential to maintain epithelial integrity while at the same time enable the shape changes of mitotic cells.
Collapse
Affiliation(s)
- Jooske L. Monster
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lisa Donker
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marjolein J. Vliem
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Zaw Win
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Helen K. Matthews
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Joleen S. Cheah
- Department of Biomedical Engineering, University of California, Davis, Davis, CA
| | - Soichiro Yamada
- Department of Biomedical Engineering, University of California, Davis, Davis, CA
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Buzz Baum
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Martijn Gloerich
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
23
|
Kermath BA, Vanderplow AM, Cahill ME. Dysregulated Prefrontal Cortical RhoA Signal Transduction in Bipolar Disorder with Psychosis: New Implications for Disease Pathophysiology. Cereb Cortex 2021; 30:59-71. [PMID: 31220216 DOI: 10.1093/cercor/bhz070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
While research has identified alterations in dorsolateral prefrontal cortical function as a key factor to the etiology of bipolar disorder, few studies have uncovered robust changes in protein signal transduction pathways in this disorder. Given the direct relevance of protein-based expressional alterations to cellular functions and because many of the key regulatory mechanisms for the disease pathogenesis likely include alterations in protein activity rather than changes in expression alone, the identification of alterations in discrete signal transduction pathways in bipolar disorder would have broad implications for understanding the disease pathophysiology. As prior microarray data point to a previously unrecognized involvement of the RhoA network in bipolar disorder, here we investigate the protein expression and activity of key components of a RhoA signal transduction pathway in dorsolateral prefrontal cortical homogenates from subjects with bipolar disorder. The results of this investigation implicate overactivation of prefrontal cortical RhoA signaling in specific subtypes of bipolar disorder. The specificity of these findings is demonstrated by a lack of comparable changes in schizophrenia; however, our findings do identify convergence between both disorders at the level of activity-mediated actin cytoskeletal regulation. These findings have implications for understanding the altered cortical synaptic connectivity of bipolar disorder.
Collapse
Affiliation(s)
- Bailey A Kermath
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| | - Michael E Cahill
- Department of Comparative Biosciences, University of Wisconsin at Madison, Madison, WI, USA
| |
Collapse
|
24
|
Galea GL, Maniou E, Edwards TJ, Marshall AR, Ampartzidis I, Greene NDE, Copp AJ. Cell non-autonomy amplifies disruption of neurulation by mosaic Vangl2 deletion in mice. Nat Commun 2021; 12:1159. [PMID: 33608529 PMCID: PMC7895924 DOI: 10.1038/s41467-021-21372-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/22/2021] [Indexed: 01/31/2023] Open
Abstract
Post-zygotic mutations that generate tissue mosaicism are increasingly associated with severe congenital defects, including those arising from failed neural tube closure. Here we report that neural fold elevation during mouse spinal neurulation is vulnerable to deletion of the VANGL planar cell polarity protein 2 (Vangl2) gene in as few as 16% of neuroepithelial cells. Vangl2-deleted cells are typically dispersed throughout the neuroepithelium, and each non-autonomously prevents apical constriction by an average of five Vangl2-replete neighbours. This inhibition of apical constriction involves diminished myosin-II localisation on neighbour cell borders and shortening of basally-extending microtubule tails, which are known to facilitate apical constriction. Vangl2-deleted neuroepithelial cells themselves continue to apically constrict and preferentially recruit myosin-II to their apical cell cortex rather than to apical cap localisations. Such non-autonomous effects can explain how post-zygotic mutations affecting a minority of cells can cause catastrophic failure of morphogenesis leading to clinically important birth defects.
Collapse
Affiliation(s)
- Gabriel L Galea
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK.
- Comparative Bioveterinary Sciences, Royal Veterinary College, London, UK.
| | - Eirini Maniou
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK
| | - Timothy J Edwards
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK
| | - Abigail R Marshall
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK
| | - Ioakeim Ampartzidis
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK
| | - Nicholas D E Greene
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK
| | - Andrew J Copp
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK
| |
Collapse
|
25
|
Otani T, Furuse M. Tight Junction Structure and Function Revisited. Trends Cell Biol 2020; 30:805-817. [PMID: 32891490 DOI: 10.1016/j.tcb.2020.08.004] [Citation(s) in RCA: 322] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
Abstract
Tight junctions (TJs) are intercellular junctions critical for building the epithelial barrier and maintaining epithelial polarity. The claudin family of membrane proteins play central roles in TJ structure and function. However, recent findings have uncovered claudin-independent aspects of TJ structure and function, and additional players including junctional adhesion molecules (JAMs), membrane lipids, phase separation of the zonula occludens (ZO) family of scaffolding proteins, and mechanical force have been shown to play important roles in TJ structure and function. In this review, we discuss how these new findings have the potential to transform our understanding of TJ structure and function, and how the intricate network of TJ proteins and membrane lipids dynamically interact to drive TJ assembly.
Collapse
Affiliation(s)
- Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan.
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
26
|
The Mechanical Microenvironment in Breast Cancer. Cancers (Basel) 2020; 12:cancers12061452. [PMID: 32503141 PMCID: PMC7352870 DOI: 10.3390/cancers12061452] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 01/22/2023] Open
Abstract
Mechanotransduction is the interpretation of physical cues by cells through mechanosensation mechanisms that elegantly translate mechanical stimuli into biochemical signaling pathways. While mechanical stress and their resulting cellular responses occur in normal physiologic contexts, there are a variety of cancer-associated physical cues present in the tumor microenvironment that are pathological in breast cancer. Mechanistic in vitro data and in vivo evidence currently support three mechanical stressors as mechanical modifiers in breast cancer that will be the focus of this review: stiffness, interstitial fluid pressure, and solid stress. Increases in stiffness, interstitial fluid pressure, and solid stress are thought to promote malignant phenotypes in normal breast epithelial cells, as well as exacerbate malignant phenotypes in breast cancer cells.
Collapse
|
27
|
Logan CM, Menko AS. Microtubules: Evolving roles and critical cellular interactions. Exp Biol Med (Maywood) 2019; 244:1240-1254. [PMID: 31387376 DOI: 10.1177/1535370219867296] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microtubules are cytoskeletal elements known as drivers of directed cell migration, vesicle and organelle trafficking, and mitosis. In this review, we discuss new research in the lens that has shed light into further roles for stable microtubules in the process of development and morphogenesis. In the lens, as well as other systems, distinct roles for characteristically dynamic microtubules and stabilized populations are coming to light. Understanding the mechanisms of microtubule stabilization and the associated microtubule post-translational modifications is an evolving field of study. Appropriate cellular homeostasis relies on not only one cytoskeletal element, but also rather an interaction between cytoskeletal proteins as well as other cellular regulators. Microtubules are key integrators with actin and intermediate filaments, as well as cell–cell junctional proteins and other cellular regulators including myosin and RhoGTPases to maintain this balance.Impact statementThe role of microtubules in cellular functioning is constantly expanding. In this review, we examine new and exciting fields of discovery for microtubule’s involvement in morphogenesis, highlight our evolving understanding of differential roles for stabilized versus dynamic subpopulations, and further understanding of microtubules as a cellular integrator.
Collapse
Affiliation(s)
- Caitlin M Logan
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - A Sue Menko
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
28
|
Abstract
Mechanical forces drive the remodeling of tissues during morphogenesis. This relies on the transmission of forces between cells by cadherin-based adherens junctions, which couple the force-generating actomyosin cytoskeletons of neighboring cells. Moreover, components of cadherin adhesions adopt force-dependent conformations that induce changes in the composition of adherens junctions, enabling transduction of mechanical forces into an intracellular response. Cadherin mechanotransduction can mediate reinforcement of cell–cell adhesions to withstand forces but also induce biochemical signaling to regulate cell behavior or direct remodeling of cell–cell adhesions to enable cell rearrangements. By transmission and transduction of mechanical forces, cadherin adhesions coordinate cellular behaviors underlying morphogenetic processes of collective cell migration, cell division, and cell intercalation. Here, we review recent advances in our understanding of this central role of cadherin adhesions in force-dependent regulation of morphogenesis.
Collapse
Affiliation(s)
- Willem-Jan Pannekoek
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn Gloerich
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
29
|
Aoyama G, Kurosaka H, Oka A, Nakatsugawa K, Yamamoto S, Sarper SE, Usami Y, Toyosawa S, Inubushi T, Isogai Y, Yamashiro T. Observation of Dynamic Cellular Migration of the Medial Edge Epithelium of the Palatal Shelf in vitro. Front Physiol 2019; 10:698. [PMID: 31244674 PMCID: PMC6562562 DOI: 10.3389/fphys.2019.00698] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 05/20/2019] [Indexed: 12/22/2022] Open
Abstract
Palatal fusion is a critical step during palatogenesis. In this fusing interface, the epithelial sheets need to be removed in order to achieve mesenchymal continuity. Epithelial cellular migration is one of the possible mechanisms, and live imaging of the labeled epithelium could provide direct evidence for it. However, the removal of medial edge epithelium (MEE) between the bilateral processes takes place in the middle of the dorso-ventral axis of the palatal shelf, and thus it is challenging to capture the cellular behavior directly. Here, we evaluate cellular behavior of MEE cells using a live imaging technique with a mouse model which expresses GFP under the promoter of Keratin14 (K14-GFP) and unpaired palatal shelf culture. Using this approach, we successfully obtained live images of epithelial behavior and detected epithelial cell migration on the surface of the secondary palatal shelf without touching of the opposing shelf. Additionally, the pattern of epithelial elimination resulted in oval-shaped exposed mesenchyme, which recapitulated the situation during secondary palate fusion in vivo. Detailed image processing revealed that most of the MEE migrated in an outward direction at the boundary regions as the oval shape of the exposed mesenchyme expanded. The migration was preceded by the bulging of MEE, and disappearance of GFP signals was not evident in bulging or migrating MEE at the boundary regions. Furthermore, the MEE migration and the subsequent mesenchymal exposure were disturbed by application of ROCK inhibitor. Together, these findings indicated that epithelial cell migration contributed importantly to the MEE removal and the subsequent exposure of the underlying mesenchyme. Furthermore, they indicated that the migration of epithelial cells was regulated in a time- and space-specific manner, since unpaired palatal shelf culture exhibited these cellular behaviors even in the absence of the opposing shelf. Altogether, present data indicated that this new experimental system combining live imaging with GFP-labeled epithelium mice and unpaired palatal shelf culture enabled direct visualization of cellular migration of MEE in vitro and could be a powerful tool to investigate its cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Gozo Aoyama
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Ayaka Oka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Kohei Nakatsugawa
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Sayuri Yamamoto
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Safiye Esra Sarper
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan.,Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Yu Usami
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Satoru Toyosawa
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Toshihiro Inubushi
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Yukako Isogai
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
30
|
Rafiq NBM, Nishimura Y, Plotnikov SV, Thiagarajan V, Zhang Z, Shi S, Natarajan M, Viasnoff V, Kanchanawong P, Jones GE, Bershadsky AD. A mechano-signalling network linking microtubules, myosin IIA filaments and integrin-based adhesions. NATURE MATERIALS 2019; 18:638-649. [PMID: 31114072 DOI: 10.1038/s41563-019-0371-y] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/10/2019] [Indexed: 06/09/2023]
Abstract
The interrelationship between microtubules and the actin cytoskeleton in mechanoregulation of integrin-mediated adhesions is poorly understood. Here, we show that the effects of microtubules on two major types of cell-matrix adhesion, focal adhesions and podosomes, are mediated by KANK family proteins connecting the adhesion protein talin with microtubule tips. Both total microtubule disruption and microtubule uncoupling from adhesions by manipulations with KANKs trigger a massive assembly of myosin IIA filaments, augmenting focal adhesions and disrupting podosomes. Myosin IIA filaments are indispensable effectors in the microtubule-driven regulation of integrin-mediated adhesions. Myosin IIA filament assembly depends on Rho activation by the RhoGEF GEF-H1, which is trapped by microtubules when they are connected with integrin-mediated adhesions via KANK proteins but released after their disconnection. Thus, microtubule capture by integrin-mediated adhesions modulates the GEF-H1-dependent effect of microtubules on the assembly of myosin IIA filaments. Subsequent actomyosin reorganization then remodels the focal adhesions and podosomes, closing the regulatory loop.
Collapse
Affiliation(s)
- Nisha Bte Mohd Rafiq
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Yukako Nishimura
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Sergey V Plotnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | | | - Zhen Zhang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Shidong Shi
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | | | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- CNRS UMI 3639, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Deparment of Biomedical Engineering, National University of Singapore, Singapore, Singapore.
| | - Gareth E Jones
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK.
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
31
|
Abstract
The cytoskeleton is crucially important for the assembly of cell-cell junctions and the homeostatic regulation of their functions. Junctional proteins act, in turn, as anchors for cytoskeletal filaments, and as regulators of cytoskeletal dynamics and signalling proteins. The cross-talk between junctions and the cytoskeleton is critical for the morphogenesis and physiology of epithelial and other tissues, but is not completely understood. Microtubules are implicated in the delivery of junctional proteins to cell-cell contact sites, in the differentiation and spatial organization of the cytoplasm, and in the stabilization of the barrier and adhesive functions of junctions. Here we focus on the relationships between microtubules and junctions of vertebrate epithelial cells. We highlight recent discoveries on the molecular underpinnings of microtubule-junction interactions, and report new data about the interaction of cingulin and paracingulin with microtubules. We also propose a possible new role of junctions as “molecular sinks” for microtubule-associated signalling proteins.
Collapse
Affiliation(s)
- Ekaterina Vasileva
- a Department of Cell Biology, Faculty of Sciences and Institute for Genetics and Genomics in Geneva (iGE3) , University of Geneva , Geneva , Switzerland
| | - Sandra Citi
- a Department of Cell Biology, Faculty of Sciences and Institute for Genetics and Genomics in Geneva (iGE3) , University of Geneva , Geneva , Switzerland
| |
Collapse
|