1
|
Nolin SJ, Taylor RL, Edens FW, Siegel PB, Ashwell CM. Combining supervised machine learning with statistics reveals differential gene expression patterns related to energy metabolism in the jejuna of chickens divergently selected for antibody response to sheep red blood cells. Poult Sci 2023; 102:102751. [PMID: 37244088 DOI: 10.1016/j.psj.2023.102751] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/19/2023] [Accepted: 04/23/2023] [Indexed: 05/29/2023] Open
Abstract
Since the 1970s, 2 lines of White Leghorn chickens, HAS and LAS, have been continuously divergently selected for 5-day postinjection antibody titer to injection with sheep red blood cells (SRBC). Antibody response is a complex genetic trait and characterizing differences in gene expression could facilitate better understanding of physiological changes due to selection and antigen exposure. At 41 d of age, randomly selected HAS and LAS chickens, which had been coraised from hatch, were either injected with SRBC (HASI and LASI) or kept as the noninjected cohort (HASN and LASN). Five days later, all were euthanized, and samples collected from the jejunum for RNA isolation and sequencing. Resulting gene expression data were analyzed combining traditional statistics with machine learning to obtain signature gene lists for functional analysis. Differences in ATP production and cellular processes were observed in the jejunum between lines and following SRBC injection. HASN vs. LASN exhibited upregulation of ATP production, immune cell motility, and inflammation. LASI exhibits upregulation of ATP production and protein synthesis vs. LASN, reflective of what was observed in HASN vs. LASN. In contrast, no corresponding upregulation of ATP production was observed in HASI vs. HASN, and most other cellular processes appear inhibited. Without exposure to SRBC, gene expression in the jejunum indicates HAS generates more ATP than LAS, suggesting HAS maintains a "primed" system; and gene expression of HASI vs. HASN further suggests this basal ATP production is sufficient for robust antibody responses. Conversely, LASI vs. LASN jejunal gene expression implies a physiological need for increased ATP production with only minimal correlating antibody production. The results of this experiment provide insight into energetic resource needs and allocations in the jejunum in response to genetic selection and antigen exposure in HAS and LAS which may help explain phenotypic differences observed in antibody response.
Collapse
Affiliation(s)
- Shelly J Nolin
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA.
| | - Robert L Taylor
- Davis College of Agriculture, Natural Resources, and Design, West Virginia University, Morgantown West, VA 26506-6108, USA
| | - Frank W Edens
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Paul B Siegel
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Christopher M Ashwell
- Davis College of Agriculture, Natural Resources, and Design, West Virginia University, Morgantown West, VA 26506-6108, USA
| |
Collapse
|
2
|
Grolez GP, Chinigò G, Barras A, Hammadi M, Noyer L, Kondratska K, Bulk E, Oullier T, Marionneau-Lambot S, Le Mée M, Rétif S, Lerondel S, Bongiovanni A, Genova T, Roger S, Boukherroub R, Schwab A, Fiorio Pla A, Gkika D. TRPM8 as an Anti-Tumoral Target in Prostate Cancer Growth and Metastasis Dissemination. Int J Mol Sci 2022; 23:ijms23126672. [PMID: 35743115 PMCID: PMC9224463 DOI: 10.3390/ijms23126672] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/05/2022] [Accepted: 06/12/2022] [Indexed: 02/04/2023] Open
Abstract
In the fight against prostate cancer (PCa), TRPM8 is one of the most promising clinical targets. Indeed, several studies have highlighted that TRPM8 involvement is key in PCa progression because of its impact on cell proliferation, viability, and migration. However, data from the literature are somewhat contradictory regarding the precise role of TRPM8 in prostatic carcinogenesis and are mostly based on in vitro studies. The purpose of this study was to clarify the role played by TRPM8 in PCa progression. We used a prostate orthotopic xenograft mouse model to show that TRPM8 overexpression dramatically limited tumor growth and metastasis dissemination in vivo. Mechanistically, our in vitro data revealed that TRPM8 inhibited tumor growth by affecting the cell proliferation and clonogenic properties of PCa cells. Moreover, TRPM8 impacted metastatic dissemination mainly by impairing cytoskeleton dynamics and focal adhesion formation through the inhibition of the Cdc42, Rac1, ERK, and FAK pathways. Lastly, we proved the in vivo efficiency of a new tool based on lipid nanocapsules containing WS12 in limiting the TRPM8-positive cells' dissemination at metastatic sites. Our work strongly supports the protective role of TRPM8 on PCa progression, providing new insights into the potential application of TRPM8 as a therapeutic target in PCa treatment.
Collapse
Affiliation(s)
- Guillaume P. Grolez
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
| | - Giorgia Chinigò
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
- Department of Life Science and Systems Biology, University of Turin, 10123 Turin, Italy;
| | - Alexandre Barras
- CNRS, Centrale Lille, Univ. Lille, Univ. Polytechnique Hauts-de-France, UMR 8520—IEMN, 59000 Lille, France; (A.B.); (M.H.); (R.B.)
| | - Mehdi Hammadi
- CNRS, Centrale Lille, Univ. Lille, Univ. Polytechnique Hauts-de-France, UMR 8520—IEMN, 59000 Lille, France; (A.B.); (M.H.); (R.B.)
| | - Lucile Noyer
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
| | - Kateryna Kondratska
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
| | - Etmar Bulk
- Institute of Physiology II, University of Münster, 48149 Münster, Germany; (E.B.); (A.S.)
| | - Thibauld Oullier
- Cancéropôle du Grand Ouest, Plateforme In Vivo, 44000 Nantes, France; (T.O.); (S.M.-L.)
| | | | - Marilyne Le Mée
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Stéphanie Rétif
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Stéphanie Lerondel
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Antonino Bongiovanni
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UMS 2014—PLBS, University of Lille, 59000 Lille, France;
| | - Tullio Genova
- Department of Life Science and Systems Biology, University of Turin, 10123 Turin, Italy;
- Nanostructured Interfaces and Surfaces Centre of Excellence (NIS), University of Turin, 10123 Turin, Italy
| | - Sébastien Roger
- Transplantation, Immunologie et Inflammation T2I-EA 4245, Université de Tours, 37044 Tours, France;
| | - Rabah Boukherroub
- CNRS, Centrale Lille, Univ. Lille, Univ. Polytechnique Hauts-de-France, UMR 8520—IEMN, 59000 Lille, France; (A.B.); (M.H.); (R.B.)
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, 48149 Münster, Germany; (E.B.); (A.S.)
| | - Alessandra Fiorio Pla
- Laboratoire de Physiologie Cellulaire, INSERM U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille, 59000 Villeneuve d’Ascq, France; (G.P.G.); (G.C.); (L.N.); (K.K.); (A.F.P.)
- Department of Life Science and Systems Biology, University of Turin, 10123 Turin, Italy;
- CNRS UAR44, PHENOMIN-TAAM, 45071 Orléans, France; (M.L.M.); (S.R.); (S.L.)
| | - Dimitra Gkika
- CNRS, INSERM, CHU Lille, Centre Oscar Lambret, UMR 9020-UMR 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, University Lille, 59000 Villeneuve d’Ascq, France
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Institut Universitaire de France (IUF), 75231 Paris, France
- Correspondence:
| |
Collapse
|
3
|
Esposito D, Pant I, Shen Y, Qiao RF, Yang X, Bai Y, Jin J, Poulikakos PI, Aaronson SA. ROCK1 mechano-signaling dependency of human malignancies driven by TEAD/YAP activation. Nat Commun 2022; 13:703. [PMID: 35121738 PMCID: PMC8817028 DOI: 10.1038/s41467-022-28319-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Rho family mechano-signaling through the actin cytoskeleton positively regulates physiological TEAD/YAP transcription, while the evolutionarily conserved Hippo tumor suppressor pathway antagonizes this transcription through YAP cytoplasmic localization/degradation. The mechanisms responsible for oncogenic dysregulation of these pathways, their prevalence in tumors, as well as how such dysregulation can be therapeutically targeted are not resolved. We demonstrate that p53 DNA contact mutants in human tumors, indirectly hyperactivate RhoA/ROCK1/actomyosin signaling, which is both necessary and sufficient to drive oncogenic TEAD/YAP transcription. Moreover, we demonstrate that recurrent lesions in the Hippo pathway depend on physiological levels of ROCK1/actomyosin signaling for oncogenic TEAD/YAP transcription. Finally, we show that ROCK inhibitors selectively antagonize proliferation and motility of human tumors with either mechanism. Thus, we identify a cancer driver paradigm and a precision medicine approach for selective targeting of human malignancies driven by TEAD/YAP transcription through mechanisms that either upregulate or depend on homeostatic RhoA mechano-signaling.
Collapse
Affiliation(s)
- Davide Esposito
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ila Pant
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yao Shen
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rui F Qiao
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xiaobao Yang
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yiyang Bai
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Poulikos I Poulikakos
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Dermatology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stuart A Aaronson
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Li W, Li F, Zhang X, Lin HK, Xu C. Insights into the post-translational modification and its emerging role in shaping the tumor microenvironment. Signal Transduct Target Ther 2021; 6:422. [PMID: 34924561 PMCID: PMC8685280 DOI: 10.1038/s41392-021-00825-8] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/11/2022] Open
Abstract
More and more in-depth studies have revealed that the occurrence and development of tumors depend on gene mutation and tumor heterogeneity. The most important manifestation of tumor heterogeneity is the dynamic change of tumor microenvironment (TME) heterogeneity. This depends not only on the tumor cells themselves in the microenvironment where the infiltrating immune cells and matrix together forming an antitumor and/or pro-tumor network. TME has resulted in novel therapeutic interventions as a place beyond tumor beds. The malignant cancer cells, tumor infiltrate immune cells, angiogenic vascular cells, lymphatic endothelial cells, cancer-associated fibroblastic cells, and the released factors including intracellular metabolites, hormonal signals and inflammatory mediators all contribute actively to cancer progression. Protein post-translational modification (PTM) is often regarded as a degradative mechanism in protein destruction or turnover to maintain physiological homeostasis. Advances in quantitative transcriptomics, proteomics, and nuclease-based gene editing are now paving the global ways for exploring PTMs. In this review, we focus on recent developments in the PTM area and speculate on their importance as a critical functional readout for the regulation of TME. A wealth of information has been emerging to prove useful in the search for conventional therapies and the development of global therapeutic strategies.
Collapse
Affiliation(s)
- Wen Li
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China
| | - Feifei Li
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Chuan Xu
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China.
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA.
| |
Collapse
|
5
|
Lee H, Bonin K, Guthold M. Human mammary epithelial cells in a mature, stratified epithelial layer flatten and stiffen compared to single and confluent cells. Biochim Biophys Acta Gen Subj 2021; 1865:129891. [PMID: 33689830 DOI: 10.1016/j.bbagen.2021.129891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND The epithelium forms a protective barrier against external biological, chemical and physical insults. So far, AFM-based, micro-mechanical measurements have only been performed on single cells and confluent cells, but not yet on cells in mature layers. METHODS Using a combination of atomic force, fluorescence and confocal microscopy, we determined the changes in stiffness, morphology and actin distribution of human mammary epithelial cells (HMECs) as they transition from single cells to confluency to a mature layer. RESULTS Single HMECs have a tall, round (planoconvex) morphology, have actin stress fibers at the base, have diffuse cortical actin, and have a stiffness of 1 kPa. Confluent HMECs start to become flatter, basal actin stress fibers start to disappear, and actin accumulates laterally where cells abut. Overall stiffness is still 1 kPa with two-fold higher stiffness in the abutting regions. As HMECs mature and form multilayered structures, cells on apical surfaces become flatter (apically more level), wider, and seven times stiffer (mean, 7 kPa) than single and confluent cells. The main drivers of these changes are actin filaments, as cells show strong actin accumulation in the regions where cells adjoin, and in the apical regions. CONCLUSIONS HMECs stiffen, flatten and redistribute actin upon transiting from single cells to mature, confluent layers. GENERAL SIGNIFICANCE Our findings advance the understanding of breast ductal morphogenesis and mechanical homeostasis.
Collapse
Affiliation(s)
- Hyunsu Lee
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Keith Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Martin Guthold
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA.
| |
Collapse
|
6
|
Barcelona‐Estaje E, Dalby MJ, Cantini M, Salmeron‐Sanchez M. You Talking to Me? Cadherin and Integrin Crosstalk in Biomaterial Design. Adv Healthc Mater 2021; 10:e2002048. [PMID: 33586353 DOI: 10.1002/adhm.202002048] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/20/2021] [Indexed: 12/21/2022]
Abstract
While much work has been done in the design of biomaterials to control integrin-mediated adhesion, less emphasis has been put on functionalization of materials with cadherin ligands. Yet, cell-cell contacts in combination with cell-matrix interactions are key in driving embryonic development, collective cell migration, epithelial to mesenchymal transition, and cancer metastatic processes, among others. This review focuses on the incorporation of both cadherin and integrin ligands in biomaterial design, to promote what is called the "adhesive crosstalk." First, the structure and function of cadherins and their role in eliciting mechanotransductive processes, by themselves or in combination with integrin mechanosensing, are introduced. Then, biomaterials that mimic cell-cell interactions, and recent applications to get insights in fundamental biology and tissue engineering, are critically discussed.
Collapse
Affiliation(s)
- Eva Barcelona‐Estaje
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | - Matthew J. Dalby
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | - Marco Cantini
- Centre for the Cellular Microenvironment University of Glasgow Glasgow G12 8QQ UK
| | | |
Collapse
|
7
|
Díaz-Díaz C, Baonza G, Martín-Belmonte F. The vertebrate epithelial apical junctional complex: Dynamic interplay between Rho GTPase activity and cell polarization processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183398. [DOI: 10.1016/j.bbamem.2020.183398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
|
8
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
9
|
Jahn SK, Hennicke T, Kassack MU, Drews L, Reichert AS, Fritz G. Distinct influence of the anthracycline derivative doxorubicin on the differentiation efficacy of mESC-derived endothelial progenitor cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118711. [PMID: 32224192 DOI: 10.1016/j.bbamcr.2020.118711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/13/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
Cardiotoxicity is a highly relevant, because often life-threatening, adverse effect of doxorubicin (Doxo)-based anticancer therapy. Here, we investigated the Doxo-response of cardiovascular stem/progenitor cells employing a mouse embryonic stem cell (mESC)-based in vitro differentiation model. Endothelial progenitor cells revealed a pronounced Doxo sensitivity as compared to mESC, differentiated endothelial-like (EC) and cardiomyocyte-like cells (CM) and CM progenitors, which rests on the activation of senescence. Doxo treatment of EC progenitors altered protein expression of individual endothelial markers, actin cytoskeleton morphology, mRNA expression of genes related to mitochondrial functions, autophagy, apoptosis, and DNA repair as well as mitochondrial DNA content, respiration and ATP production in the surviving differentiated EC progeny. By contrast, LDL uptake, ATP-stimulated Ca2+ release, and cytokine-stimulated ICAM-1 expression remained unaffected by the anthracycline treatment. Thus, exposure of EC progenitors to Doxo elicits isolated and persistent dysfunctions in the surviving EC progeny. In conclusion, we suggest that Doxo-induced injury of EC progenitors adds to anthracycline-induced cardiotoxicity, making this cell-type a preferential target for pharmacoprotective and regenerative strategies.
Collapse
Affiliation(s)
- Sarah K Jahn
- Institute of Toxicology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Tatiana Hennicke
- Institute of Toxicology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Matthias U Kassack
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Duesseldorf, Universitätsstr. 1, 40225 Duesseldorf, Germany
| | - Leonie Drews
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitätsstr. 1, 40225 Duesseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitätsstr. 1, 40225 Duesseldorf, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| |
Collapse
|
10
|
Qiao W, Li D, Shi Q, Wang H, Wang H, Guo J. miR-224-5p protects dental pulp stem cells from apoptosis by targeting Rac1. Exp Ther Med 2019; 19:9-18. [PMID: 31897093 PMCID: PMC6923752 DOI: 10.3892/etm.2019.8213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 07/12/2019] [Indexed: 12/27/2022] Open
Abstract
Dental pulp stem cells (DPSCs) are reported to be enriched in stem/progenitor cells, however to the best of our knowledge they have yet to be well documented and characterized. In the present study, in order to characterize DPSCs and the effect of microRNAs (miRs/miRNAs) on DPSC properties, a miRNA array was performed between dental periodontal ligament cells (DPLCs) and DPSCs. The results revealed that miR-224-5p (miR-224) was highly expressed in the DPSCs compared with that in the DPLCs. The transfection of DPSCs with an miR-224 inhibitor impaired cell viability. In addition, miR-224 inhibition significantly promoted cell apoptosis in DPSCscompared with the NC group. In silico analysis and a dual-luciferase reporter assay demonstrated that miR-224 targets the 3′-untranslated region of the Rac family small GTPase 1 (Rac1) gene. miR-224 downregulation resulted in the increased expression of Rac1 in DPSCs compared with DPLCs. Furthermore, miR-224 inhibition caused augmented mitogen-activated protein kinase 8, caspase-3, caspase-9 and Fas ligand expression in DPSC, which may be recovered by Rac1 silencing with transfection with short hairpin RNA-Rac1. Furthermore, Annexin V-fluorescein isothiocyanate/propidium iodide flow cytometry indicated that the silencing of Rac1 restored the pro-apoptotic DPSC cell number with miR-224 transfection. Therefore, the results of the present study suggested miR-224 in DPSC serves an important function in protecting cells against apoptosis by downregulating Rac1 expression, and also identified miR-224 as a novel miRNA in regulating the features of DPSC.
Collapse
Affiliation(s)
- Wenlan Qiao
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Stomatology, Qilu Hospital, and Institute of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dong Li
- Department of Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qing Shi
- Department of Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Huanhuan Wang
- Department of Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hao Wang
- Department of Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jing Guo
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
11
|
Garcia De Las Bayonas A, Philippe JM, Lellouch AC, Lecuit T. Distinct RhoGEFs Activate Apical and Junctional Contractility under Control of G Proteins during Epithelial Morphogenesis. Curr Biol 2019; 29:3370-3385.e7. [PMID: 31522942 PMCID: PMC6839405 DOI: 10.1016/j.cub.2019.08.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/15/2019] [Accepted: 08/07/2019] [Indexed: 01/08/2023]
Abstract
Small RhoGTPases direct cell shape changes and movements during tissue morphogenesis. Their activities are tightly regulated in space and time to specify the desired pattern of actomyosin contractility that supports tissue morphogenesis. This is expected to stem from polarized surface stimuli and from polarized signaling processing inside cells. We examined this general problem in the context of cell intercalation that drives extension of the Drosophila ectoderm. In the ectoderm, G protein-coupled receptors (GPCRs) and their downstream heterotrimeric G proteins (Gα and Gβγ) activate Rho1 both medial-apically, where it exhibits pulsed dynamics, and at junctions, where its activity is planar polarized. However, the mechanisms responsible for polarizing Rho1 activity are unclear. We report that distinct guanine exchange factors (GEFs) activate Rho1 in these two cellular compartments. RhoGEF2 acts uniquely to activate medial-apical Rho1 but is recruited both medial-apically and at junctions by Gα12/13-GTP, also called Concertina (Cta) in Drosophila. On the other hand, Dp114RhoGEF (Dp114), a newly characterized RhoGEF, is required for cell intercalation in the extending ectoderm, where it activates Rho1 specifically at junctions. Its localization is restricted to adherens junctions and is under Gβ13F/Gγ1 control. Furthermore, Gβ13F/Gγ1 activates junctional Rho1 and exerts quantitative control over planar polarization of Rho1. Finally, we found that Dp114RhoGEF is absent in the mesoderm, arguing for a tissue-specific control over junctional Rho1 activity. These results clarify the mechanisms of polarization of Rho1 activity in different cellular compartments and reveal that distinct GEFs are sensitive tuning parameters of cell contractility in remodeling epithelia. Dp114RhoGEF activates junctional Rho1 and is involved in cell intercalation Gα/Cta and Gβγ subunits tune, respectively, RhoGEF2 and Dp114RhoGEF membrane levels Gβγ subunits control planar polarity of junctional Rho1 signaling via Dp114RhoGEF Tissue-specific RhoGEFs could diversify morphogenesis in different tissues
Collapse
Affiliation(s)
| | - Jean-Marc Philippe
- Aix Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| | - Annemarie C Lellouch
- Aix Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| | - Thomas Lecuit
- Aix Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France; Collège de France, 11 Place Marcelin Berthelot, 75005 Paris, France.
| |
Collapse
|
12
|
Shigetomi K, Ikenouchi J. Cell Adhesion Structures in Epithelial Cells Are Formed in Dynamic and Cooperative Ways. Bioessays 2019; 41:e1800227. [PMID: 31187900 DOI: 10.1002/bies.201800227] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/16/2019] [Indexed: 01/13/2023]
Abstract
There are many morphologically distinct membrane structures with different functions at the surface of epithelial cells. Among these, adherens junctions (AJ) and tight junctions (TJ) are responsible for the mechanical linkage of epithelial cells and epithelial barrier function, respectively. In the process of new cell-cell adhesion formation between two epithelial cells, such as after wounding, AJ form first and then TJ form on the apical side of AJ. This process is very complicated because AJ formation triggers drastic changes in the organization of actin cytoskeleton, the activity of Rho family of small GTPases, and the lipid composition of the plasma membrane, all of which are required for subsequent TJ formation. In this review, the authors focus on the relationship between AJ and TJ as a representative example of specialization of plasma membrane regions and introduce recent findings on how AJ formation promotes the subsequent formation of TJ.
Collapse
Affiliation(s)
- Kenta Shigetomi
- Department of Biology, Faculty of Sciences, Kyushu University, 774 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Junichi Ikenouchi
- Department of Biology, Faculty of Sciences, Kyushu University, 774 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.,Japan Science and Technology Agency, Saitama, 332-0012, Japan.,AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| |
Collapse
|
13
|
Coordination between Rac1 and Rab Proteins: Functional Implications in Health and Disease. Cells 2019; 8:cells8050396. [PMID: 31035701 PMCID: PMC6562727 DOI: 10.3390/cells8050396] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023] Open
Abstract
The small GTPases of the Rho family regulate many aspects of actin dynamics, but are functionally connected to many other cellular processes. Rac1, a member of this family, besides its known function in the regulation of actin cytoskeleton, plays a key role in the production of reactive oxygen species, in gene transcription, in DNA repair, and also has been proven to have specific roles in neurons. This review focuses on the cooperation between Rac1 and Rab proteins, analyzing how the coordination between these GTPases impact on cells and how alterations of their functions lead to disease.
Collapse
|
14
|
Braga V. Signaling by Small GTPases at Cell-Cell Junctions: Protein Interactions Building Control and Networks. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028746. [PMID: 28893858 DOI: 10.1101/cshperspect.a028746] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A number of interesting reports highlight the intricate network of signaling proteins that coordinate formation and maintenance of cell-cell contacts. We have much yet to learn about how the in vitro binding data is translated into protein association inside the cells and whether such interaction modulates the signaling properties of the protein. What emerges from recent studies is the importance to carefully consider small GTPase activation in the context of where its activation occurs, which upstream regulators are involved in the activation/inactivation cycle and the GTPase interacting partners that determine the intracellular niche and extent of signaling. Data discussed here unravel unparalleled cooperation and coordination of functions among GTPases and their regulators in supporting strong adhesion between cells.
Collapse
Affiliation(s)
- Vania Braga
- Molecular Medicine, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
15
|
Frank SR, Köllmann CP, Luong P, Galli GG, Zou L, Bernards A, Getz G, Calogero RA, Frödin M, Hansen SH. p190 RhoGAP promotes contact inhibition in epithelial cells by repressing YAP activity. J Cell Biol 2018; 217:3183-3201. [PMID: 29934311 PMCID: PMC6122998 DOI: 10.1083/jcb.201710058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/06/2018] [Accepted: 05/29/2018] [Indexed: 12/14/2022] Open
Abstract
ARHGAP35 encoding p190A RhoGAP is a cancer-associated gene with a mutation spectrum suggestive of a tumor-suppressor function. In this study, we demonstrate that loss of heterozygosity for ARHGAP35 occurs in human tumors. We sought to identify tumor-suppressor capacities for p190A RhoGAP (p190A) and its paralog p190B in epithelial cells. We reveal an essential role for p190A and p190B to promote contact inhibition of cell proliferation (CIP), a function that relies on RhoGAP activity. Unbiased mRNA sequencing analyses establish that p190A and p190B modulate expression of genes associated with the Hippo pathway. Accordingly, we determine that p190A and p190B induce CIP by repressing YAP-TEAD-regulated gene transcription through activation of LATS kinases and inhibition of the Rho-ROCK pathway. Finally, we demonstrate that loss of a single p190 paralog is sufficient to elicit nuclear translocation of YAP and perturb CIP in epithelial cells cultured in Matrigel. Collectively, our data reveal a novel mechanism consistent with a tumor-suppressor function for ARHGAP35.
Collapse
Affiliation(s)
- Scott R Frank
- GI Cell Biology Research Laboratory, Boston Children's Hospital and Harvard Medical School, Boston, MA
| | - Clemens P Köllmann
- GI Cell Biology Research Laboratory, Boston Children's Hospital and Harvard Medical School, Boston, MA
| | - Phi Luong
- GI Cell Biology Research Laboratory, Boston Children's Hospital and Harvard Medical School, Boston, MA
| | - Giorgio G Galli
- Stem Cell Program, Boston Children's Hospital and Harvard Stem Cell Institute, Boston, MA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
| | - Lihua Zou
- The Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - André Bernards
- The Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
- Massachusetts General Hospital Center for Cancer Research and Harvard Medical School, Charlestown, MA
| | - Gad Getz
- The Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
- Massachusetts General Hospital Center for Cancer Research and Harvard Medical School, Charlestown, MA
| | - Raffaele A Calogero
- University of Torino, Department of Molecular Biotechnology and Health Sciences, Torino, Italy
| | - Morten Frödin
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Steen H Hansen
- GI Cell Biology Research Laboratory, Boston Children's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
Derksen PWB, van de Ven RAH. Shared mechanisms regulate spatiotemporal RhoA-dependent actomyosin contractility during adhesion and cell division. Small GTPases 2018; 11:113-121. [PMID: 29291271 DOI: 10.1080/21541248.2017.1366966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Local modulation of the actin cytoskeleton is essential for the initiation and maintenance of strong homotypic adhesive interfaces between neighboring cells. The epithelial adherens junction (AJ) fulfils a central role in this process by mediating E-cadherin interactions and functioning as a signaling scaffold to control the activity of the small GTPase RhoA and subsequent actomyosin contractility. Interestingly, a number of regulatory proteins that modulate RhoA activity at the AJ also control RhoA during cytokinesis, an actomyosin-dependent process that divides the cytoplasm to generate two daughter cells at the final stages of mitosis. Recent insights have revealed that the central player in AJ stability, p120-catenin (p120), interacts with and modulates essential regulators of actomyosin contraction during cytokinesis. In cancer, loss of this modulation is a common event during tumor progression that can induce chromosomal instability and tumor progression.In this review, we will highlight the functional differences and similarities of the different RhoA-associated factors that have been linked to both the regulation of cell-cell adhesion and cytokinesis.
Collapse
Affiliation(s)
- Patrick W B Derksen
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan CX Utrecht, the Netherlands
| | - Robert A H van de Ven
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue Boston, MA, USA
| |
Collapse
|
17
|
A new role of the Rac-GAP β2-chimaerin in cell adhesion reveals opposite functions in breast cancer initiation and tumor progression. Oncotarget 2017; 7:28301-19. [PMID: 27058424 PMCID: PMC5053728 DOI: 10.18632/oncotarget.8597] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/27/2016] [Indexed: 01/06/2023] Open
Abstract
β2-chimaerin is a Rac1-specific negative regulator and a candidate tumor suppressor in breast cancer but its precise function in mammary tumorigenesis in vivo is unknown. Here, we study for the first time the role of β2-chimaerin in breast cancer using a mouse model and describe an unforeseen role for this protein in epithelial cell-cell adhesion. We demonstrate that expression of β2-chimaerin in breast cancer epithelial cells reduces E-cadherin protein levels, thus loosening cell-cell contacts. In vivo, genetic ablation of β2-chimaerin in the MMTV-Neu/ErbB2 mice accelerates tumor onset, but delays tumor progression. Finally, analysis of clinical databases revealed an inverse correlation between β2-chimaerin and E-cadherin gene expressions in Her2+ breast tumors. Furthermore, breast cancer patients with low β2-chimaerin expression have reduced relapse free survival but develop metastasis at similar times. Overall, our data redefine the role of β2-chimaerin as tumor suppressor and provide the first in vivo evidence of a dual function in breast cancer, suppressing tumor initiation but favoring tumor progression.
Collapse
|
18
|
Verma D, Bajpai VK, Ye N, Maneshi MM, Jetta D, Andreadis ST, Sachs F, Hua SZ. Flow induced adherens junction remodeling driven by cytoskeletal forces. Exp Cell Res 2017; 359:327-336. [PMID: 28803065 DOI: 10.1016/j.yexcr.2017.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/18/2017] [Accepted: 08/05/2017] [Indexed: 12/30/2022]
Abstract
Adherens junctions (AJs) are a key structural component for tissue organization and function. Under fluid shear stress, AJs exhibit dynamic assembly/disassembly, but how shear stress couples to AJs is unclear. In MDCK cells we measured simultaneously the forces in cytoskeletal α-actinin and the density and length of AJs using a genetically coded optical force sensor, actinin-sstFRET, and fluorescently labeled E-cadherin (E-cad). We found that shear stress of 0.74dyn/cm2 for 3h significantly enhanced E-cad expression at cell-cell contacts and this phenomenon has two phases. The initial formation of segregated AJ plaques coincided with a decrease in cytoskeletal tension, but an increase in tension was necessary for expansion of the plaques and the formation of continuous AJs in the later phase. The changes in cytoskeletal tension and reorganization appear to be an upstream process in response to flow since it occurred in both wild type and dominant negative E-cad cells. Disruption of F-actin with a Rho-ROCK inhibitor eliminated AJ growth under flow. These results delineate the shear stress transduction paths in cultured cells, which helps to understand pathology of a range of diseases that involve dysfunction of E-cadherin.
Collapse
Affiliation(s)
- Deepika Verma
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY 14260, USA; Department of Physiology and Biophysics, University at Buffalo, Buffalo, NY 14260, USA
| | - Vivek K Bajpai
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Nannan Ye
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Mohammad M Maneshi
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Deekshitha Jetta
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Frederick Sachs
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY 14260, USA
| | - Susan Z Hua
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY 14260, USA; Department of Physiology and Biophysics, University at Buffalo, Buffalo, NY 14260, USA.
| |
Collapse
|
19
|
Carmon KS, Gong X, Yi J, Wu L, Thomas A, Moore CM, Masuho I, Timson DJ, Martemyanov KA, Liu QJ. LGR5 receptor promotes cell-cell adhesion in stem cells and colon cancer cells via the IQGAP1-Rac1 pathway. J Biol Chem 2017; 292:14989-15001. [PMID: 28739799 PMCID: PMC5592675 DOI: 10.1074/jbc.m117.786798] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/21/2017] [Indexed: 12/18/2022] Open
Abstract
Leucine-rich repeat-containing G protein–coupled receptor 5 (LGR5) is a bona fide marker of adult stem cells in several epithelial tissues, most notably in the intestinal crypts, and is highly up-regulated in many colorectal, hepatocellular, and ovarian cancers. LGR5 activation by R-spondin (RSPO) ligands potentiates Wnt/β-catenin signaling in vitro; however, deletion of LGR5 in stem cells has little or no effect on Wnt/β-catenin signaling or cell proliferation in vivo. Remarkably, modulation of LGR5 expression has a major impact on the actin cytoskeletal structure and cell adhesion in the absence of RSPO stimulation, but the molecular mechanism is unclear. Here, we show that LGR5 interacts with IQ motif-containing GTPase-activating protein 1 (IQGAP1), an effector of Rac1/CDC42 GTPases, in the regulation of actin cytoskeleton dynamics and cell–cell adhesion. Specifically, LGR5 decreased levels of IQGAP1 phosphorylation at Ser-1441/1443, leading to increased binding of Rac1 to IQGAP1 and thus higher levels of cortical F-actin and enhanced cell–cell adhesion. LGR5 ablation in colon cancer cells and crypt stem cells resulted in loss of cortical F-actin, reduced cell–cell adhesion, and disrupted localization of adhesion-associated proteins. No evidence of LGR5 coupling to any of the four major subtypes of heterotrimeric G proteins was found. These findings suggest that LGR5 primarily functions via the IQGAP1–Rac1 pathway to strengthen cell–cell adhesion in normal adult crypt stem cells and colon cancer cells.
Collapse
Affiliation(s)
- Kendra S Carmon
- From the Brown Foundation Institute of Molecular Medicine and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030
| | - Xing Gong
- From the Brown Foundation Institute of Molecular Medicine and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030
| | - Jing Yi
- From the Brown Foundation Institute of Molecular Medicine and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030.,Department of Cancer Biology, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030
| | - Ling Wu
- From the Brown Foundation Institute of Molecular Medicine and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030
| | - Anthony Thomas
- From the Brown Foundation Institute of Molecular Medicine and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030
| | - Catherine M Moore
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, Ireland, United Kingdom
| | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, and
| | - David J Timson
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, Ireland, United Kingdom.,School of Pharmacy and Biomolecular Sciences, University of Brighton, Huxley Building, Lewes Road, Brighton BN2 4GJ, United Kingdom
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, and
| | - Qingyun J Liu
- From the Brown Foundation Institute of Molecular Medicine and Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas 77030,
| |
Collapse
|
20
|
Bachir AI, Horwitz AR, Nelson WJ, Bianchini JM. Actin-Based Adhesion Modules Mediate Cell Interactions with the Extracellular Matrix and Neighboring Cells. Cold Spring Harb Perspect Biol 2017; 9:9/7/a023234. [PMID: 28679638 DOI: 10.1101/cshperspect.a023234] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell adhesions link cells to the extracellular matrix (ECM) and to each other and depend on interactions with the actin cytoskeleton. Both cell-ECM and cell-cell adhesion sites contain discrete, yet overlapping, functional modules. These modules establish physical associations with the actin cytoskeleton, locally modulate actin organization and dynamics, and trigger intracellular signaling pathways. Interplay between these modules generates distinct actin architectures that underlie different stages, types, and functions of cell-ECM and cell-cell adhesions. Actomyosin contractility is required to generate mature, stable adhesions, as well as to sense and translate the mechanical properties of the cellular environment into changes in cell organization and behavior. Here, we review the organization and function of different adhesion modules and how they interact with the actin cytoskeleton. We highlight the molecular mechanisms of mechanotransduction in adhesions and how adhesion molecules mediate cross talk between cell-ECM and cell-cell adhesion sites.
Collapse
Affiliation(s)
- Alexia I Bachir
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - Alan Rick Horwitz
- Protein and Cell Analysis, Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon 97402
| | - W James Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| | - Julie M Bianchini
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
21
|
van Dijk IA, Ferrando ML, van der Wijk AE, Hoebe RA, Nazmi K, de Jonge WJ, Krawczyk PM, Bolscher JGM, Veerman ECI, Stap J. Human salivary peptide histatin-1 stimulates epithelial and endothelial cell adhesion and barrier function. FASEB J 2017; 31:3922-3933. [PMID: 28522595 DOI: 10.1096/fj.201700180r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/24/2017] [Indexed: 12/17/2022]
Abstract
Histatins are multifunctional histidine-rich peptides secreted by the salivary glands and exclusively present in the saliva of higher primates, where they play a fundamental role in the protection of the oral cavity. Our previously published results demonstrated that histatin-1 (Hst1) promotes cell-substrate adhesion in various cell types and hinted that it could also be involved in cell-cell adhesion, a process of fundamental importance to epithelial and endothelial barriers. Here we explore the effects of Hst1 on cellular barrier function. We show that Hst1 improved endothelial barrier integrity, decreased its permeability for large molecules, and prevented translocation of bacteria across epithelial cell layers. These effects are mediated by the adherens junction protein E-cadherin (E-cad) and by the tight junction protein zonula occludens 1, as Hst1 increases the levels of zonula occludens 1 and of active E-cad. Hst1 may also promote epithelial differentiation as Hst1 induced transcription of the epithelial cell differentiation marker apolipoprotein A-IV (a downstream E-cad target). In addition, Hst1 counteracted the effects of epithelial-mesenchymal transition inducers on the outgrowth of oral cancer cell spheroids, suggesting that Hst1 affects processes that are implicated in cancer progression.-Van Dijk, I. A., Ferrando, M. L., van der Wijk, A.-E., Hoebe, R. A., Nazmi, K., de Jonge, W. J., Krawczyk, P. M., Bolscher, J. G. M., Veerman, E. C. I., Stap, J. Human salivary peptide histatin-1 stimulates epithelial and endothelial cell adhesion and barrier function.
Collapse
Affiliation(s)
- Irene A van Dijk
- Department of Medical Biology and Core Facility Cellular Imaging, Van Leeuwenhoek Centre for Advanced Microscopy-Academic Medical Center (LCAM-AMC), University of Amsterdam, Amsterdam, The Netherlands; .,Department of Oral Biochemistry, University of Amsterdam and Vrije Universiteit (VU) Amsterdam, Amsterdam, The Netherlands
| | - Maria Laura Ferrando
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Eva van der Wijk
- Department of Ophthalmology, Ocular Angiogenesis Group, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Medical Biology, Ocular Angiogenesis Group, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ron A Hoebe
- Department of Medical Biology and Core Facility Cellular Imaging, Van Leeuwenhoek Centre for Advanced Microscopy-Academic Medical Center (LCAM-AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Kamran Nazmi
- Department of Oral Biochemistry, University of Amsterdam and Vrije Universiteit (VU) Amsterdam, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Przemek M Krawczyk
- Department of Medical Biology and Core Facility Cellular Imaging, Van Leeuwenhoek Centre for Advanced Microscopy-Academic Medical Center (LCAM-AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Jan G M Bolscher
- Department of Oral Biochemistry, University of Amsterdam and Vrije Universiteit (VU) Amsterdam, Amsterdam, The Netherlands
| | - Enno C I Veerman
- Department of Oral Biochemistry, University of Amsterdam and Vrije Universiteit (VU) Amsterdam, Amsterdam, The Netherlands
| | - Jan Stap
- Department of Medical Biology and Core Facility Cellular Imaging, Van Leeuwenhoek Centre for Advanced Microscopy-Academic Medical Center (LCAM-AMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Arnold TR, Stephenson RE, Miller AL. Rho GTPases and actomyosin: Partners in regulating epithelial cell-cell junction structure and function. Exp Cell Res 2017; 358:20-30. [PMID: 28363828 DOI: 10.1016/j.yexcr.2017.03.053] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 01/11/2023]
Abstract
Epithelial tissues are defined by polarized epithelial cells that are integrated into tissues and exhibit barrier function in order to regulate what is allowed to pass between cells. Cell-cell junctions must be stable enough to promote barrier function and tissue integrity, yet plastic enough to remodel when necessary. This remarkable ability to dynamically sense and respond to changes in cell shape and tissue tension allows cell-cell junctions to remain functional during events that disrupt epithelial homeostasis including morphogenesis, wound healing, and cell division. In order to achieve this plasticity, both tight junctions and adherens junctions are coupled to the underlying actomyosin cytoskeleton. Here, we discuss the importance of the junctional linkage to actomyosin and how a localized zone of active RhoA along with other Rho GTPases work together to orchestrate junctional actomyosin dynamics. We focus on how scaffold proteins help coordinate Rho GTPases, their upstream regulators, and their downstream effectors for efficient, localized Rho GTPase signaling output. Additionally, we highlight important roles junctional actin-binding proteins play in addition to their traditional roles in organizing actin. Together, Rho GTPases, their regulators, and effectors form compartmentalized signaling modules that regulate actomyosin structure and contractility to achieve proper cell-cell adhesion and tissue barriers.
Collapse
Affiliation(s)
- Torey R Arnold
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Rachel E Stephenson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
23
|
Sluysmans S, Vasileva E, Spadaro D, Shah J, Rouaud F, Citi S. The role of apical cell-cell junctions and associated cytoskeleton in mechanotransduction. Biol Cell 2017; 109:139-161. [PMID: 28220498 DOI: 10.1111/boc.201600075] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 01/13/2023]
Abstract
Tissues of multicellular organisms are characterised by several types of specialised cell-cell junctions. In vertebrate epithelia and endothelia, tight and adherens junctions (AJ) play critical roles in barrier and adhesion functions, and are connected to the actin and microtubule cytoskeletons. The interaction between junctions and the cytoskeleton is crucial for tissue development and physiology, and is involved in the molecular mechanisms governing cell shape, motility, growth and signalling. The machineries which functionally connect tight and AJ to the cytoskeleton comprise proteins which either bind directly to cytoskeletal filaments, or function as adaptors for regulators of the assembly and function of the cytoskeleton. In the last two decades, specific cytoskeleton-associated junctional molecules have been implicated in mechanotransduction, revealing the existence of multimolecular complexes that can sense mechanical cues and translate them into adaptation to tensile forces and biochemical signals. Here, we summarise the current knowledge about the machineries that link tight and AJ to actin filaments and microtubules, and the molecular basis for mechanotransduction at epithelial and endothelial AJ.
Collapse
Affiliation(s)
- Sophie Sluysmans
- Department of Cell Biology, Institute of Genomics and Genetics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Ekaterina Vasileva
- Department of Cell Biology, Institute of Genomics and Genetics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Domenica Spadaro
- Department of Cell Biology, Institute of Genomics and Genetics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Jimit Shah
- Department of Cell Biology, Institute of Genomics and Genetics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Florian Rouaud
- Department of Cell Biology, Institute of Genomics and Genetics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| | - Sandra Citi
- Department of Cell Biology, Institute of Genomics and Genetics of Geneva (iGE3), University of Geneva, Geneva, Switzerland
| |
Collapse
|
24
|
Kaminota T, Yano H, Shiota K, Nomura N, Yaguchi H, Kirino Y, Ohara K, Tetsumura I, Sanada T, Ugumori T, Tanaka J, Hato N. Elevated Na +/H + exchanger-1 expression enhances the metastatic collective migration of head and neck squamous cell carcinoma cells. Biochem Biophys Res Commun 2017; 486:101-107. [PMID: 28268168 DOI: 10.1016/j.bbrc.2017.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/03/2017] [Indexed: 01/13/2023]
Abstract
Cancer cells can migrate as collectives during invasion and/or metastasis; however, the precise molecular mechanisms of this form of migration are less clear compared with single cell migration following epithelial-mesenchymal transition. Elevated Na+/H+ exchanger1 (NHE1) expression has been suggested to have malignant roles in a number of cancer cell lines and in vivo tumor models. Furthermore, a metastatic human head and neck squamous cell carcinoma (HNSCC) cell line (SASL1m) that was isolated based on its increased metastatic potential also exhibited higher NHE1 expression than its parental line SAS. Time-lapse video recordings indicated that both cell lines migrate as collectives, although with different features, e.g., SASL1m was much more active and changed the direction of migration more frequently than SAS cells, whereas locomotive activities were comparable. SASL1m cells also exhibited higher invasive activity than SAS in Matrigel invasion assays. shRNA-mediated NHE1 knockdown in SASL1m led to reduced locomotive and invasive activities, suggesting a critical role for NHE1 in the collective migration of SASL1m cells. SASL1m cells also exhibited a higher metastatic rate than SAS cells in a mouse lymph node metastasis model, while NHE1 knockdown suppressed in vivo SASL1m metastasis. Finally, elevated NHE1 expression was observed in human HNSCC tissue, and Cariporide, a specific NHE1 inhibitor, reduced the invasive activity of SASL1m cells, implying NHE1 could be a target for anti-invasion/metastasis therapy.
Collapse
Affiliation(s)
- Teppei Kaminota
- Department of Otorhinolaryngology, Head and Neck Surgery, Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Kohei Shiota
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan
| | - Noriko Nomura
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Haruna Yaguchi
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Yui Kirino
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Kentaro Ohara
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Issei Tetsumura
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Tomoyoshi Sanada
- Department of Otorhinolaryngology, Head and Neck Surgery, Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Toru Ugumori
- Department of Otorhinolaryngology, Head and Neck Surgery, Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| | - Naohito Hato
- Department of Otorhinolaryngology, Head and Neck Surgery, Ehime University Medical School, 454, Shitsukawa, To-on, Ehime, 791-0295, Japan.
| |
Collapse
|
25
|
Mascia A, Gentile F, Izzo A, Mollo N, De Luca M, Bucci C, Nitsch L, Calì G. Rab7 Regulates CDH1 Endocytosis, Circular Dorsal Ruffles Genesis, and Thyroglobulin Internalization in a Thyroid Cell Line. J Cell Physiol 2015; 231:1695-708. [PMID: 26599499 DOI: 10.1002/jcp.25267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/23/2015] [Indexed: 01/02/2023]
Abstract
Rab7 regulates the biogenesis of late endosomes, lysosomes, and autophagosomes. It has been proposed that a functional and physical interaction exists between Rab7 and Rac1 GTPases in CDH1 endocytosis and ruffled border formation. In FRT cells over-expressing Rab7, increased expression and activity of Rac1 was observed, whereas a reduction of Rab7 expression by RNAi resulted in reduced Rac1 activity, as measured by PAK1 phosphorylation. We found that CDH1 endocytosis was extremely reduced only in Rab7 over-expressing cells but was unchanged in Rab7 silenced cells. In Rab7 under or over-expressing cells, Rab7 and LC3B-II co-localized and co-localization in large circular structures occurred only in Rab7 over-expressing cells. These large circular structures occurred in about 10% of the cell population; some of them (61%) showed co-localization of Rab7 with cortactin and f-actin and were identified as circular dorsal ruffles (CDRs), the others as mature autophagosomes. We propose that the over-expression of Rab7 is sufficient to induce CDRs. Furthermore, in FRT cells, we found that the expression of the insoluble/active form of Rab7, rather than Rab5, or Rab8, was inducible by cAMP and that cAMP-stimulated FRT cells showed increased PAK1 phosphorylation and were no longer able to endocytose CDH1. Finally, we demonstrated that Rab7 over-expressing cells are able to endocytose exogenous thyroglobulin via pinocytosis/CDRs more efficiently than control cells. We propose that the major thyroglobulin endocytosis described in thyroid autonomous adenomas due to Rab7 increased expression, occurs via CDRs. J. Cell. Physiol. 231: 1695-1708, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anna Mascia
- IEOS Istituto di Endocrinologia e Oncologia Sperimentale "G. Salvatore", National Council of Research, Napoli, Italy
| | - Flaviana Gentile
- IEOS Istituto di Endocrinologia e Oncologia Sperimentale "G. Salvatore", National Council of Research, Napoli, Italy
| | - Antonella Izzo
- Department of Molecular Medicine and Medical Biotechnologies, University "Federico II", Napoli, Italy
| | - Nunzia Mollo
- Department of Molecular Medicine and Medical Biotechnologies, University "Federico II", Napoli, Italy
| | - Maria De Luca
- Department of Biological and Environmental Sciences and Technologies, University of Salento (DiSTeBA), Lecce, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, University of Salento (DiSTeBA), Lecce, Italy
| | - Lucio Nitsch
- Department of Molecular Medicine and Medical Biotechnologies, University "Federico II", Napoli, Italy
| | - Gaetano Calì
- IEOS Istituto di Endocrinologia e Oncologia Sperimentale "G. Salvatore", National Council of Research, Napoli, Italy
| |
Collapse
|
26
|
Abstract
The establishment and maintenance of epithelial cell-cell junctions is crucially important to regulate adhesion, apico-basal polarity and motility of epithelial cells, and ultimately controls the architecture and physiology of epithelial organs. Junctions are supported, shaped and regulated by cytoskeletal filaments, whose dynamic organization and contractility are finely tuned by GTPases of the Rho family, primarily RhoA, Rac1 and Cdc42. Recent research has identified new molecular mechanisms underlying the cross-talk between these GTPases and epithelial junctions. Here we briefly summarize the current knowledge about the organization, molecular evolution and cytoskeletal anchoring of cell-cell junctions, and we comment on the most recent advances in the characterization of the interactions between Rho GTPases and junctional proteins, and their consequences with regards to junction assembly and regulation of cell behavior in vertebrate model systems. The concept of “zonular signalosome” is proposed, which highlights the close functional relationship between proteins of zonular junctions (zonulae occludentes and adhaerentes) and the control of cytoskeletal organization and signaling through Rho GTPases, transcription factors, and their effectors.
Collapse
Key Words
- AJ, adherens junction
- AMOT, angiomotin
- AMPK, Adenosine Monophosphate-Activated Protein Kinase
- APC, adenomatous poliposis coli
- CD2AP, CD2-associated protein
- CGN, cingulin
- CGNL1, paracingulin
- Cdc42
- Cdc42, cell division cycle 42
- DLC, deleted in liver cancer
- Dbl, diffuse B-cell lymphoma
- EPLIN, epithelial protein lost in neoplasm
- ERK, extracellular regulated kinase
- FERM, four.point.one, ezrin, radixin, moesin
- FGD5, FYVE, RhoGEF and PH domain containing 5
- GAP, GTPase activating protein
- GEF, guanine nucleotide exchange factor
- GST, glutathione -S- transferase; JAM = junctional adhesion molecule
- MCF-7, Michigan Cancer Foundation - 7
- MDCK, Madin Darby Canine Kidney
- MKLP1, mitotic kinesin-like protein-1
- MRCK, myotonic dystrophy-related Cdc42-binding kinase
- MgcRacGAP, male germ cell racGAP
- PA, puncta adhaerentia
- PAK, p21-activated kinase; PATJ, Pals1 associated tight junction protein
- PCNA, proliferating cell nuclear antigen
- PDZ, Post synaptic density protein (PSD95), Drosophila, disc large tumour suppressor (DlgA), and zonula occludens-1
- PLEKHA7, pleckstrin homology domain containing, family A member 7
- RICH-1, RhoGAP interacting with CIP4 homologues
- ROCK, Rho-associated protein kinase
- Rac
- Rho
- SH3BP1, (SH3 domain 490 binding protein-1)
- TJ, tight junction
- Tbx-3, T-box-3
- Tiam, Tumor invasion and metastasis
- WASP, Wiskott-Aldrich Syndrome Protein
- WAVE, WASP family Verprolin-homologous protein
- ZA, zonula adhaerens
- ZO, zonula occludens
- ZONAB, (ZO-1)–associated nucleic acid binding protein.
- cytoseleton
- epithelium
- junctions
Collapse
Affiliation(s)
- Sandra Citi
- a Department of Cell Biology ; University of Geneva ; Geneva , Switzerland
| | | | | | | |
Collapse
|
27
|
Abstract
Signaling via the Rho GTPases provides crucial regulation of numerous cell polarization events, including apicobasal (AB) polarity, polarized cell migration, polarized cell division and neuronal polarity. Here we review the relationships between the Rho family GTPases and epithelial AB polarization events, focusing on the 3 best-characterized members: Rho, Rac and Cdc42. We discuss a multitude of processes that are important for AB polarization, including lumen formation, apical membrane specification, cell-cell junction assembly and maintenance, as well as tissue polarity. Our discussions aim to highlight the immensely complex regulatory mechanisms that encompass Rho GTPase signaling during AB polarization. More specifically, in this review we discuss several emerging common themes, that include: 1) the need for Rho GTPase activities to be carefully balanced in both a spatial and temporal manner through a multitude of mechanisms; 2) the existence of signaling feedback loops and crosstalk to create robust cellular responses; and 3) the frequent multifunctionality that exists among AB polarity regulators. Regarding this latter theme, we provide further discussion of the potential plasticity of the cell polarity machinery and as a result the possible implications for human disease.
Collapse
Key Words
- AB, Apicobasal
- AJ, Adherens junction
- Amot, Angiomotin
- Arp2/3, Actin-related protein-2/3
- Baz, Bazooka
- C. elegans, Caenorhabditis elegans
- CA, Constitutively-active
- CD2AP, CD2-associated protein
- Caco2, Human colon carcinoma
- Cdc42
- Cora, Coracle
- Crb, Crumbs
- DN, Dominant-negative
- Dia1, Diaphanous-related formin 1
- Dlg, Discs large
- Drosophila, Drosophila melanogaster
- Dys-β, Dystrobrevin-β
- ECM, Extracellular matrix
- Ect2, Epithelial cell transforming sequence 2 oncogene
- Eya1, Eyes absent 1
- F-actin, Filamentous actin
- FRET, Fluorescence resonance energy transfer
- GAP, GTPase-activating protein
- GDI, Guanine nucleotide dissociation inhibitor
- GEF, Guanine nucleotide exchange factor
- GTPases
- JACOP, Junction-associated coiled-coiled protein
- JAM, Junctional adhesion molecule
- LKB1, Liver kinase B1
- Lgl, Lethal giant larvae
- MDCK, Madin-Darby canine kidney
- MTOC, Microtubule-organizing center
- NrxIV, Neurexin IV
- Pals1, Protein associated with Lin-7 1
- Par, Partitioning-defective
- Patj, Pals1-associated TJ protein
- ROCK, Rho-associated kinase
- Rac
- Rho
- Rich1, RhoGAP interacting with CIP4 homologues
- S. cerevisiae, Saccharomyces cerevisiae
- S. pombe, Schizosaccharomyces pombe
- SH3BP1, SH3-domain binding protein 1
- Scrib, Scribble
- Std, Stardust
- TEM4, Tumor endothelial marker 4
- TJ, Tight junction
- Tiam1, T-cell lymphoma invasion and metastasis-inducing protein 1
- WASp, Wiskott-aldrich syndrome protein
- Yrt, Yurt
- ZA, zonula adherens
- ZO, Zonula occludens
- aPKC, Atypical Protein Kinase C
- apicobasal
- epithelia
- junction
- par
- polarity
- α-cat, Alpha-catenin
- β-cat, Beta-Catenin
- β2-syn, Beta-2-syntrophin
Collapse
Affiliation(s)
- Natalie Ann Mack
- a School of Life Sciences; Queens Medical Center ; University of Nottingham ; Nottingham , UK
| | | |
Collapse
|
28
|
Reviriego-Mendoza MM, Santy LC. The cytohesin guanosine exchange factors (GEFs) are required to promote HGF-mediated renal recovery after acute kidney injury (AKI) in mice. Physiol Rep 2015; 3:3/6/e12442. [PMID: 26116550 PMCID: PMC4522160 DOI: 10.14814/phy2.12442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The lack of current treatment and preventable measures for acute kidney injury (AKI) in hospitalized patients results in an increased mortality rate of up to 80% and elevated health costs. Additionally, if not properly repaired, those who survive AKI may develop fibrosis and long-term kidney damage. The molecular aspects of kidney injury and repair are still uncertain. Hepatocyte growth factor (HGF) promotes recovery of the injured kidney by inducing survival and migration of tubular epithelial cells to repopulate bare tubule areas. HGF-stimulated kidney epithelial cell migration requires the activation of ADP-ribosylation factor 6 (Arf6) and Rac1 via the cytohesin family of Arf-guanine-nucleotide exchange factors (GEFs), in vitro. We used an ischemia and reperfusion injury (IRI) mouse model to analyze the effects of modulating this signaling pathway on kidney recovery. We treated IRI mice with either HGF, the cytohesin inhibitor SecinH3, or a combination of both. As previously reported, HGF treatment promoted rapid improvement of kidney function as evidenced by creatinine (Cre) and blood urea nitrogen (BUN) levels. In contrast, simultaneous treatment with SecinH3 and HGF blocks the ability of HGF to promote kidney recovery. Immunohistochemistry showed that HGF treatment promoted recovery of tubule structure, and had enhanced levels of active, GTP-bound Arf6 and GTP-Rac1. SecinH3 treatment, however, caused a dramatic decrease in GTP-Arf6 and GTP-Rac1 levels when compared to kidney sections from HGF-treated IRI mice. Additionally, SecinH3 counteracted the renal reparative effects of HGF. Our results support the conclusion that cytohesin function is required for HGF-stimulated renal IRI repair.
Collapse
Affiliation(s)
- Marta M Reviriego-Mendoza
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania
| | - Lorraine C Santy
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
29
|
Selamat W, Tay PLF, Baskaran Y, Manser E. The Cdc42 Effector Kinase PAK4 Localizes to Cell-Cell Junctions and Contributes to Establishing Cell Polarity. PLoS One 2015; 10:e0129634. [PMID: 26068882 PMCID: PMC4466050 DOI: 10.1371/journal.pone.0129634] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 05/11/2015] [Indexed: 01/22/2023] Open
Abstract
The serine/threonine kinase PAK4 is a Cdc42 effector whose role is not well understood; overexpression of PAK4 has been associated with some cancers, and there are reports that correlate kinase level with increased cell migration in vitro. Here we report that PAK4 is primarily associated with cell-cell junctions in all the cell lines we tested, and fails to accumulate at focal adhesions or at the leading edge of migrating cells. In U2OS osteosarcoma and MCF-7 breast cancer cell lines, PAK4 depletion did not affect collective cell migration, but affected cell polarization. By contrast, Cdc42 depletion (as reported by many studies) caused a strong defect in junctional assembly in multiple cells lines. We also report that the depletion of PAK4 protein or treatment of cells with the PAK4 inhibitor PF-3758309 can lead to defects in centrosome reorientation (polarization) after cell monolayer wounding. These experiments are consistent with PAK4 forming part of a conserved cell-cell junctional polarity Cdc42 complex. We also confirm β-catenin as a target for PAK4 in these cells. Treatment of cells with PF-3758309 caused inhibition of β-catenin Ser-675 phosphorylation, which is located predominantly at cell-cell junctions.
Collapse
Affiliation(s)
- Widyawilis Selamat
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei-Ling Felicia Tay
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yohendran Baskaran
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ed Manser
- small G-protein Signaling and Kinases (sGSK) Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
30
|
Zihni C, Balda MS, Matter K. Signalling at tight junctions during epithelial differentiation and microbial pathogenesis. J Cell Sci 2015; 127:3401-13. [PMID: 25125573 DOI: 10.1242/jcs.145029] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tight junctions are a component of the epithelial junctional complex, and they form the paracellular diffusion barrier that enables epithelial cells to create cellular sheets that separate compartments with different compositions. The assembly and function of tight junctions are intimately linked to the actomyosin cytoskeleton and, hence, are under the control of signalling mechanisms that regulate cytoskeletal dynamics. Tight junctions not only receive signals that guide their assembly and function, but transmit information to the cell interior to regulate cell proliferation, migration and survival. As a crucial component of the epithelial barrier, they are often targeted by pathogenic viruses and bacteria, aiding infection and the development of disease. In this Commentary, we review recent progress in the understanding of the molecular signalling mechanisms that drive junction assembly and function, and the signalling processes by which tight junctions regulate cell behaviour and survival. We also discuss the way in which junctional components are exploited by pathogenic viruses and bacteria, and how this might affect junctional signalling mechanisms.
Collapse
Affiliation(s)
- Ceniz Zihni
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Maria S Balda
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | - Karl Matter
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| |
Collapse
|
31
|
Mui KL, Bae YH, Gao L, Liu SL, Xu T, Radice GL, Chen CS, Assoian RK. N-Cadherin Induction by ECM Stiffness and FAK Overrides the Spreading Requirement for Proliferation of Vascular Smooth Muscle Cells. Cell Rep 2015; 10:1477-1486. [PMID: 25753414 PMCID: PMC4560684 DOI: 10.1016/j.celrep.2015.02.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 11/19/2014] [Accepted: 02/04/2015] [Indexed: 10/28/2022] Open
Abstract
In contrast to the accepted pro-proliferative effect of cell-matrix adhesion, the proliferative effect of cadherin-mediated cell-cell adhesion remains unresolved. Here, we studied the effect of N-cadherin on cell proliferation in the vasculature. We show that N-cadherin is induced in smooth muscle cells (SMCs) in response to vascular injury, an in vivo model of tissue stiffening and proliferation. Complementary experiments performed with deformable substrata demonstrated that stiffness-mediated activation of a focal adhesion kinase (FAK)-p130Cas-Rac signaling pathway induces N-cadherin. Additionally, by culturing paired and unpaired SMCs on microfabricated adhesive islands of different areas, we found that N-cadherin relaxes the spreading requirement for SMC proliferation. In vivo SMC deletion of N-cadherin strongly reduced injury-induced cycling. Finally, SMC-specific deletion of FAK inhibited proliferation after vascular injury, and this was accompanied by reduced induction of N-cadherin. Thus, a stiffness- and FAK-dependent induction of N-cadherin connects cell-matrix to cell-cell adhesion and regulates the degree of cell spreading needed for cycling.
Collapse
Affiliation(s)
- Keeley L Mui
- Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yong Ho Bae
- Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Gao
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shu-Lin Liu
- Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tina Xu
- Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Glenn L Radice
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Christopher S Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Richard K Assoian
- Program in Translational Biomechanics, Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Bopp A, Wartlick F, Henninger C, Schwarz M, Kaina B, Fritz G. Rac1 promotes diethylnitrosamine (DEN)-induced formation of liver tumors. Carcinogenesis 2015; 36:378-89. [PMID: 25556150 DOI: 10.1093/carcin/bgu323] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
To elucidate the function of the Ras-homologous GTPase Rac1 in hepatocarcinogenesis induced by diethylnitrosamine (DEN), mice lacking hepatic Rac1 expression were treated with DEN and compared to the wild-type (WT). Rac1 knock-out (KO) mice were found to have a lower tumor yield as compared to Rac1 proficient mice. The small-sized tumors formed in the absence of Rac1 lack an activated Ras/Raf/mitogen-activated protein kinase pathway, as indicated by the absence of p-ERK expression. Apparently, Rac1 is required for Ras-driven oncogenic pathways. Moreover, tumors in Rac1 deficient mice were glutamine synthase (GS) negative. They displayed a high number of p-H3-positive and cyclinB1 expressing cells, pointing to a defect in mitotic progression. To elucidate the influence of Rac1 on mechanisms of tumor initiation, acute DEN-induced hepatic stress responses were monitored. Rac1 deficiency caused fairly complex, partially time-dependent, alterations in both basal and/or DEN-induced messenger RNA (mRNA) and protein levels of susceptibility-related genes. Basal protein expression of DNA repair factors Brca1 and DNA repair protein RAD51 homolog (Rad51) and the cell cycle regulatory factor p27 was enhanced in the absence of Rac1. Following DEN treatment, p21 mRNA and protein expression was stimulated independent of the Rac1 status. Lack of Rac1 increased mechanisms of the DNA damage response (DDR), as shown by elevated protein levels of p-ATR, p-p53 and γH2AX 24h after DEN treatment. The data show that Rac1 is essential for DEN-stimulated hepatocarcinogenesis. We hypothesize that it promotes tumor initiation by counteracting the elimination of initiated cells and, moreover, alleviates the outgrowth of transformed cells. Hence, pharmacological targeting of Rac1 could be suitable for chemoprevention.
Collapse
Affiliation(s)
- Anita Bopp
- Institute of Toxicology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Friedrich Wartlick
- Institute of Toxicology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Christian Henninger
- Institute of Toxicology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Michael Schwarz
- Institute of Pharmacology and Toxicology, University Tübingen, Wilhelmstrasse 76, D-72074 Tübingen, Germany
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany and
| | - Gerhard Fritz
- Institute of Toxicology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany,
| |
Collapse
|
33
|
|
34
|
Takahashi K, Matafonov A, Sumarriva K, Ito H, Lauhan C, Zemel D, Tsuboi N, Chen J, Reynolds A, Takahashi T. CD148 tyrosine phosphatase promotes cadherin cell adhesion. PLoS One 2014; 9:e112753. [PMID: 25386896 PMCID: PMC4227875 DOI: 10.1371/journal.pone.0112753] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 10/14/2014] [Indexed: 01/06/2023] Open
Abstract
CD148 is a transmembrane tyrosine phosphatase that is expressed at cell junctions. Recent studies have shown that CD148 associates with the cadherin/catenin complex and p120 catenin (p120) may serve as a substrate. However, the role of CD148 in cadherin cell-cell adhesion remains unknown. Therefore, here we addressed this issue using a series of stable cells and cell-based assays. Wild-type (WT) and catalytically inactive (CS) CD148 were introduced to A431D (lacking classical cadherins), A431D/E-cadherin WT (expressing wild-type E-cadherin), and A431D/E-cadherin 764AAA (expressing p120-uncoupled E-cadherin mutant) cells. The effects of CD148 in cadherin adhesion were assessed by Ca2+ switch and cell aggregation assays. Phosphorylation of E-cadherin/catenin complex and Rho family GTPase activities were also examined. Although CD148 introduction did not alter the expression levels and complex formation of E-cadherin, p120, and β-catenin, CD148 WT, but not CS, promoted cadherin contacts and strengthened cell-cell adhesion in A431D/E-cadherin WT cells. This effect was accompanied by an increase in Rac1, but not RhoA and Cdc42, activity and largely diminished by Rac1 inhibition. Further, we demonstrate that CD148 reduces the tyrosine phosphorylation of p120 and β-catenin; causes the dephosphorylation of Y529 suppressive tyrosine residue in Src, a well-known CD148 site, increasing Src activity and enhancing the phosphorylation of Y228 (a Src kinase site) in p120, in E-cadherin contacts. Consistent with these findings, CD148 dephosphorylated both p120 and β-catenin in vitro. The shRNA-mediated CD148 knockdown in A431 cells showed opposite effects. CD148 showed no effects in A431D and A431D/E-cadherin 764AAA cells. In aggregate, these findings provide the first evidence that CD148 promotes E-cadherin adhesion by regulating Rac1 activity concomitant with modulation of p120, β-catenin, and Src tyrosine phosphorylation. This effect requires E-cadherin and p120 association.
Collapse
Affiliation(s)
- Keiko Takahashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Anton Matafonov
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Katherine Sumarriva
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Hideyuki Ito
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Colette Lauhan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Dana Zemel
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Nobuo Tsuboi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Jin Chen
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Albert Reynolds
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Takamune Takahashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
35
|
Chang TI, Kang HY, Kim KS, Lee SH, Nam BY, Paeng J, Kim S, Park JT, Yoo TH, Kang SW, Han SH. The effect of statin on epithelial-mesenchymal transition in peritoneal mesothelial cells. PLoS One 2014; 9:e109628. [PMID: 25275561 PMCID: PMC4183618 DOI: 10.1371/journal.pone.0109628] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/03/2014] [Indexed: 01/05/2023] Open
Abstract
Background Statins have recently been highlighted for their pleiotropic actions distinct from cholesterol-lowering effects. Despite this interest, it is currently unknown whether statin therapy inhibits peritoneal dialysis (PD)-related epithelial-mesenchymal transition (EMT). Methods In vitro, human peritoneal mesothelial cells (HPMCs) were exposed to 5.6 mM glucose (NG) or 100 mM glucose (HG) with or without simvastatin (1 µM). In vivo, PD catheters were inserted into 32 Sprague-Dawley rats, and saline (C, n = 16) or 4.25% peritoneal dialysis fluid (PDF) (PD, n = 16) was infused for 4 weeks. Eight rats from each group were treated with 5 mg/kg/day of simvastatin intraperitoneally. Changes in the protein expression of EMT markers such as E-cadherin, α-SMA, Snail, and fibronectin in HPMCs and the peritoneum were evaluated by Western blot analysis and immunofluorescence or immunohistochemical staining. We also explored whether activation of the mevalonate pathway and its downstream small GTPases were involved in dialysis-related peritoneal EMT and could be inhibited by statin treatment. Results Compared to NG cells, E-cadherin expression was significantly decreased, while α-SMA, Snail, and fibronectin expression were significantly increased in HPMCs exposed to HG, and these changes were abrogated by simvastatin (p<0.05). In addition, the cobblestone-like appearance of normal HPMCs was converted into a fibroblast-like morphology after HG treatment, which was reversed by simvastatin. These EMT-like changes were also observed in HPMCs treated with geranyl-geranyl pyrophosphate (5 µM). HG significantly increased the protein expression of RhoA and Rac1 in the membrane fractions, and these increases were ameliorated by simvastatin (p<0.05). In PD rats, E-cadherin in the peritoneum was significantly decreased, whereas α-SMA, Snail, and fibronectin expression were significantly increased (p<0.05) compared to C rats. The thickness of the mesothelial layer in the peritoneum were also significantly greater in PD rats than in C rats (p<0.05). These changes of the peritoneum in PD rats were significantly attenuated by simvastatin. Conclusion This study demonstrated that PD-related EMT was mediated via the mevalonate pathway, and statin treatment inhibited the EMT changes in HG-treated HPMCs and PDF-stimulated PD rats. These findings suggest that statins may be a promising therapeutic strategy for preservation of peritoneal membrane integrity in long-term PD patients.
Collapse
Affiliation(s)
- Tae Ik Chang
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
- Department of Internal Medicine, NHIS Medical Center, Ilsan Hospital, Goyang-shi, Gyeonggi-do, Korea
| | - Hye-Young Kang
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Kyung Sik Kim
- Department of Surgery, College of Medicine, Brain Korea 21 Project for Medical Sciences, Yonsei University, Seoul, Korea
| | - Sun Ha Lee
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Bo Young Nam
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Jisun Paeng
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Seonghun Kim
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
36
|
Asad M, Wong MK, Tan TZ, Choolani M, Low J, Mori S, Virshup D, Thiery JP, Huang RYJ. FZD7 drives in vitro aggressiveness in Stem-A subtype of ovarian cancer via regulation of non-canonical Wnt/PCP pathway. Cell Death Dis 2014; 5:e1346. [PMID: 25032869 PMCID: PMC4123093 DOI: 10.1038/cddis.2014.302] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/27/2014] [Accepted: 06/09/2014] [Indexed: 01/02/2023]
Abstract
Ovarian cancer (OC) can be classified into five biologically distinct molecular subgroups: epithelial-A (Epi-A), Epi-B, mesenchymal (Mes), Stem-A and Stem-B. Among them, Stem-A expresses genes relating to stemness and is correlated with poor clinical prognosis. In this study, we show that frizzled family receptor 7 (FZD7), a receptor for Wnt signalling, is overexpressed in the Stem-A subgroup. To elucidate the functional roles of FZD7, we used an RNA interference gene knockdown approach in three Stem-A cell lines: CH1, PA1 and OV-17R. Si-FZD7 OC cells showed reduced cell proliferation with an increase in the G0/G1 sub-population, with no effect on apoptosis. The cells also displayed a distinctive morphologic change by colony compaction to become more epithelial-like and polarised with smaller internuclear distances and increased z-axis height. Immunofluorescence (IF) staining patterns of pan-cadherin and β-catenin suggested an increase in cadherin-based cell–cell adhesion in si-FZD7 cells. We also observed a significant rearrangement in the actin cytoskeleton and an increase in tensile contractility in si-FZD7 OC cells, as evident by the loss of stress fibres and the redistribution of phospho-myosin light chain (pMLC) from the sites of cell–cell contacts to the periphery of cell colonies. Furthermore, there was reciprocal regulation of RhoA (Ras homolog family member A) and Rac1 (Ras-related C3 botulinum toxin substrate 1 (Rho family, small GTP-binding protein Rac1)) activities upon FZD7 knockdown, with a significant reduction in RhoA activity and a concomitant upregulation in Rac1 activity. These changes in pMLC and RhoA, as well as the increased TopFlash reporter activities in si-FZD7 cells, suggested involvement of the non-canonical Wnt/planar cell polarity (PCP) pathway. Selected PCP pathway genes (cadherin EGF LAG seven-pass G-type receptor 3 (CELSR3), prickle homolog 4 (Drosophila) (PRICKLE4), dishevelled-associated activator of morphogenesis 1 (DAAM1), profilin 2 (PFN2), protocadherin 9 (PCDH9), protocadherin α1 (PCDHA1), protocadherin β17 pseudogene (PCDHB17), protocadherin β3 (PCDHB3), sprouty homolog 1 (SPRY1) and protein tyrosine kinase 7 (PTK7)) were found to be more highly expressed in Stem-A than non Stem-A subgroup of OC. Taken together, our results suggest that FZD7 might drive aggressiveness in Stem-A OC by regulating cell proliferation, cell cycle progression, maintenance of the Mes phenotype and cell migration via casein kinase 1ɛ-mediated non-canonical Wnt/PCP pathway.
Collapse
Affiliation(s)
- M Asad
- Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore
| | - M K Wong
- Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore
| | - T Z Tan
- Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore
| | - M Choolani
- Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore, Singapore
| | - J Low
- 1] Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore, Singapore [2] National University Cancer Institute of Singapore, Singapore, Singapore
| | - S Mori
- Division of Cancer Genomics, Cancer Institute of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, Japan
| | - D Virshup
- Duke NUS Graduate Medical School, Singapore, Singapore
| | - J P Thiery
- 1] Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore [2] National University Cancer Institute of Singapore, Singapore, Singapore [3] Department of Biochemistry, National University of Singapore, Singapore, Singapore [4] Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - R Y-J Huang
- 1] Cancer Science Institute of Singapore, Centre for Translational Medicine NUS Yong Loo Lin School of Medicine, Singapore, Singapore [2] Department of Obstetrics and Gynaecology, National University Hospital of Singapore, Singapore, Singapore [3] National University Cancer Institute of Singapore, Singapore, Singapore
| |
Collapse
|
37
|
Deng Z, Huang Z, Yuan M, Yang K, Pang Y. Baculovirus induces host cell aggregation via a Rho/Rok-dependent mechanism. J Gen Virol 2014; 95:2310-2320. [PMID: 24866850 DOI: 10.1099/vir.0.066811-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Several baculoviruses can induce host cell aggregation during infection; however, the molecular basis remains unknown. The Rho family of small GTPases, including Rho1, Racs and Cdc42, plays important roles in cell migration and cell-cell contact. Activated GTPases target actin polymerization to discrete sites on the plasma membrane, thereby inducing membrane protrusions. In this study, we demonstrated that Spodoptera litura nucleopolyhedrovirus (SpltNPV) infection induced the amoeboid movement and aggregation of SpLi-221 cells in vitro. The amount of Rho1-GTP increased in the infected cells, which suggested that Rho1 was activated upon infection. RNA interference and superinfection of dominant-negative recombinants revealed that the SpltNPV-induced SpLi-221 cell aggregation was dependent on the Rho1, but not Racs or Cdc42, signalling pathway. Inhibition of Rho-associated protein kinase (Rok) activity by the inhibitor Y-27632 significantly reduced SpLi-221 cell aggregation. Silencing Rho1 expression with RNA interference decreased SpltNPV propagation by approximately 40 % in vitro, when SpLi-221 cells were infected at a low, but not high, m.o.i., suggesting that the SpltNPV-induced cell aggregation may benefit SpltNPV spread.
Collapse
Affiliation(s)
- Zihao Deng
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Zhihong Huang
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Meijin Yuan
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Kai Yang
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yi Pang
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou 510275, PR China
| |
Collapse
|
38
|
Guillemot L, Guerrera D, Spadaro D, Tapia R, Jond L, Citi S. MgcRacGAP interacts with cingulin and paracingulin to regulate Rac1 activation and development of the tight junction barrier during epithelial junction assembly. Mol Biol Cell 2014; 25:1995-2005. [PMID: 24807907 PMCID: PMC4072573 DOI: 10.1091/mbc.e13-11-0680] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The Rac1 inhibitor MgcRacGAP regulates Rac1 activation and TJ barrier development during junction assembly in epithelial cells. CGN and CGNL1 recruit MgcRacGAP to the TJ and interact with MgcRacGAP. The regulation of Rho-family GTPases is crucial to direct the formation of cell–cell junctions and tissue barriers. Cingulin (CGN) and paracingulin (CGNL1) control RhoA activation in epithelial cells by interacting with RhoA guanidine exchange factors. CGNL1 depletion also inhibits Rac1 activation during junction assembly. Here we show that, unexpectedly, Madin–Darby canine kidney epithelial cells depleted of both CGN and CGNL1 (double-KD cells) display normal Rac1 activation and tight junction (TJ) formation, despite decreased junctional recruitment of the Rac1 activator Tiam1. The expression of the Rac1 inhibitor MgcRacGAP is decreased in double-KD cells, and the barrier development and Rac1 activation phenotypes are rescued by exogenous expression of MgcRacGAP. MgcRacGAP colocalizes with CGN and CGNL1 at TJs and forms a complex and interacts directly in vitro with CGN and CGNL1. Depletion of either CGN or CGNL1 in epithelial cells results in decreased junctional localization of MgcRacGAP but not of ECT2, a centralspindlin-interacting Rho GEF. These results provide new insight into coordination of Rho-family GTPase activities at junctions, since apical accumulation of CGN and CGNL1 at TJs during junction maturation provides a mechanism to spatially restrict down-regulation of Rac1 activation through the recruitment of MgcRacGAP.
Collapse
Affiliation(s)
- Laurent Guillemot
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Diego Guerrera
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Domenica Spadaro
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Rocio Tapia
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Lionel Jond
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Sandra Citi
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, SwitzerlandDepartment of Cell Biology, University of Geneva, CH-1211 Geneva, SwitzerlandInstitute of Genetics and Genomics in Geneva, University of Geneva, CH-1211 Geneva, Switzerland
| |
Collapse
|
39
|
Amsellem V, Dryden NH, Martinelli R, Gavins F, Almagro LO, Birdsey GM, Haskard DO, Mason JC, Turowski P, Randi AM. ICAM-2 regulates vascular permeability and N-cadherin localization through ezrin-radixin-moesin (ERM) proteins and Rac-1 signalling. Cell Commun Signal 2014; 12:12. [PMID: 24593809 PMCID: PMC4015342 DOI: 10.1186/1478-811x-12-12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023] Open
Abstract
Background Endothelial junctions control functions such as permeability, angiogenesis and contact inhibition. VE-Cadherin (VECad) is essential for the maintenance of intercellular contacts. In confluent endothelial monolayers, N-Cadherin (NCad) is mostly expressed on the apical and basal membrane, but in the absence of VECad it localizes at junctions. Both cadherins are required for vascular development. The intercellular adhesion molecule (ICAM)-2, also localized at endothelial junctions, is involved in leukocyte recruitment and angiogenesis. Results In human umbilical vein endothelial cells (HUVEC), both VECad and NCad were found at nascent cell contacts of sub-confluent monolayers, but only VECad localized at the mature junctions of confluent monolayers. Inhibition of ICAM-2 expression by siRNA caused the appearance of small gaps at the junctions and a decrease in NCad junctional staining in sub-confluent monolayers. Endothelioma lines derived from WT or ICAM-2-deficient mice (IC2neg) lacked VECad and failed to form junctions, with loss of contact inhibition. Re-expression of full-length ICAM-2 (IC2 FL) in IC2neg cells restored contact inhibition through recruitment of NCad at the junctions. Mutant ICAM-2 lacking the binding site for ERM proteins (IC2 ΔERM) or the cytoplasmic tail (IC2 ΔTAIL) failed to restore junctions. ICAM-2-dependent Rac-1 activation was also decreased in these mutant cell lines. Barrier function, measured in vitro via transendothelial electrical resistance, was decreased in IC2neg cells, both in resting conditions and after thrombin stimulation. This was dependent on ICAM-2 signalling to the small GTPase Rac-1, since transendothelial electrical resistance of IC2neg cells was restored by constitutively active Rac-1. In vivo, thrombin-induced extravasation of FITC-labeled albumin measured by intravital fluorescence microscopy in the mouse cremaster muscle showed that permeability was increased in ICAM-2-deficient mice compared to controls. Conclusions These results indicate that ICAM-2 regulates endothelial barrier function and permeability through a pathway involving N-Cadherin, ERMs and Rac-1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Anna M Randi
- Imperial College for Translational and Experimental Medicine, NHLI Vascular Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12, ONN, UK.
| |
Collapse
|
40
|
Cameron AR, Frith JE, Gomez GA, Yap AS, Cooper-White JJ. The effect of time-dependent deformation of viscoelastic hydrogels on myogenic induction and Rac1 activity in mesenchymal stem cells. Biomaterials 2014; 35:1857-68. [DOI: 10.1016/j.biomaterials.2013.11.023] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/07/2013] [Indexed: 12/26/2022]
|
41
|
Tian R, Wang RL, Xie H, Jin W, Yu KL. Overexpressed miRNA-155 dysregulates intestinal epithelial apical junctional complex in severe acute pancreatitis. World J Gastroenterol 2013; 19:8282-8291. [PMID: 24363519 PMCID: PMC3857451 DOI: 10.3748/wjg.v19.i45.8282] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 09/11/2013] [Accepted: 09/29/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether miRNA-155 (miR-155) dysregulates apical junctional complex (AJC) protein expression in experimental severe acute pancreatitis (SAP).
METHODS: Twenty-four male BALB/c mice were randomly assigned to two groups: the SAP group (n = 12) receiving sequential intraperitoneal injection of 50 µg/kg caerulein and 10 mg/kg lipopolysaccharide over 6 h, and the control group (n = 12) receiving intraperitoneal injection of normal saline. Animals were sacrificed 3 h following the last injection for collection of blood samples and pancreas and distal ileal segment specimens. Routine pancreas and intestine histology was used to assess SAP pathology and intestinal epithelial barrier damage. Levels of serum amylase, diamine oxidase (DAO), and tumor necrosis factor (TNF)-α were determined using commercial kits. Total RNA samples were isolated from intestinal epithelial specimens and reversely transcribed into cDNA. miR-155 and RhoA mRNA expression profiles were determined using quantitative real-time polymerase chain reaction. Target genes for miR-155 were predicted using the miRTarBase database, RNA22 and PicTar computational methods. Western blotting was performed to quantitate the protein expression levels of the target gene RhoA, as well as zonula occludens (ZO)-1 and E-cadherin, two AJC component proteins.
RESULTS: Intraperitoneal injection of caerulein and lipopolysaccharide successfully induced experimental acute pancreatic damage (SAP vs control, 10.0 ± 2.0 vs 3.2 ± 1.2, P < 0.01) and intestinal epithelial barrier damage (3.2 ± 0.7 vs 1.4 ± 0.7, P < 0.01). Levels of serum amylase (21.6 ± 5.1 U/mL vs 14.3 ± 4.2 U/mL, P < 0.01), DAO (21.4 ± 4.1 mg/mL vs 2.6 ± 0.8 mg/mL, P < 0.01), and TNF-α (61.0 ± 15.1 ng/mL vs 42.9 ± 13.9 ng/mL, P < 0.01) increased significantly in SAP mice compared to those in control mice. miR-155 was significantly overexpressed in SAP intestinal epithelia (1.94 ± 0.50 fold vs 1.03 ± 0.23 fold, P < 0.01), and RhoA gene containing three miR-155-specific binding sites in the three prime untranslated regions was one of the target genes for miR-155. RhoA (22.7 ± 5.8 folds vs 59.6 ± 11.6 folds, P < 0.01), ZO-1 (46 ± 18 folds vs 68 ± 19 folds, P < 0.01), and E-cadherin proteins (48 ± 15 folds vs 77 ± 18 folds, P < 0.01) were underexpressed in SAP intestinal epithelia although RhoA mRNA expression was not significantly changed in SAP (0.97 ± 0.18 folds vs 1.01 ± 0.17 folds, P > 0.05).
CONCLUSION: TNF-α-regulated miR-155 overexpression inhibits AJC component protein syntheses of ZO-1, and E-cadherin by downregulating post-transcriptional RhoA expression, and disrupts intestinal epithelial barrier in experimental SAP.
Collapse
|
42
|
p120 catenin: an essential regulator of cadherin stability, adhesion-induced signaling, and cancer progression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:409-32. [PMID: 23481205 DOI: 10.1016/b978-0-12-394311-8.00018-2] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
p120 catenin is the best studied member of a subfamily of proteins that associate with the cadherin juxtamembrane domain to suppress cadherin endocytosis. p120 also recruits the minus ends of microtubules to the cadherin complex, leading to junction maturation. In addition, p120 regulates the activity of Rho family GTPases through multiple interactions with Rho GEFs, GAPs, Rho GTPases, and their effectors. Nuclear signaling is affected by the interaction of p120 with Kaiso, a transcription factor regulating Wnt-responsive genes as well as transcriptionally repressing methylated promoters. Multiple alternatively spliced p120 isoforms and complex phosphorylation events affect these p120 functions. In cancer, reduced p120 expression correlates with reduced E-cadherin function and with tumor progression. In contrast, in tumor cells that have lost E-cadherin expression, p120 promotes cell invasion and anchorage-independent growth. Furthermore, p120 is required for Src-induced oncogenic transformation and provides a potential target for future therapeutic interventions.
Collapse
|
43
|
Otsu K, Sakano M, Masuda T, Fujiwara N, Harada H. The role of Rho-kinases in ameloblast differentiation. J Oral Biosci 2013. [DOI: 10.1016/j.job.2013.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
44
|
Thumkeo D, Watanabe S, Narumiya S. Physiological roles of Rho and Rho effectors in mammals. Eur J Cell Biol 2013; 92:303-15. [PMID: 24183240 DOI: 10.1016/j.ejcb.2013.09.002] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/25/2013] [Accepted: 09/25/2013] [Indexed: 02/06/2023] Open
Abstract
Rho GTPase is a master regulator controlling cytoskeleton in multiple contexts such as cell migration, adhesion and cytokinesis. Of several Rho GTPases in mammals, the best characterized is the Rho subfamily including ubiquitously expressed RhoA and its homologs RhoB and RhoC. Upon binding GTP, Rho exerts its functions through downstream Rho effectors, such as ROCK, mDia, Citron, PKN, Rhophilin and Rhotekin. Until recently, our knowledge about functions of Rho and Rho effectors came mostly from in vitro studies utilizing cultured cells, and their physiological roles in vivo were largely unknown. However, gene-targeting studies of Rho and its effectors have now unraveled their tissue- and cell-specific roles and provide deeper insight into the physiological function of Rho signaling in vivo. In this article, we briefly describe previous studies of the function of Rho and its effectors in vitro and then review and discuss recent studies on knockout mice of Rho and its effectors.
Collapse
Affiliation(s)
- Dean Thumkeo
- Department of Pharmacology, Kyoto University Faculty of Medicine, Sakyo-ku, Kyoto 606-8501, Japan; Innovation Center for Immunoregulation, Technologies and Drugs (AK Project), Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8501, Japan.
| | | | | |
Collapse
|
45
|
N-cadherin regulates spatially polarized signals through distinct p120ctn and β-catenin-dependent signalling pathways. Nat Commun 2013; 4:1589. [PMID: 23481397 PMCID: PMC3602931 DOI: 10.1038/ncomms2560] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 01/29/2013] [Indexed: 12/18/2022] Open
Abstract
The spatial distribution of molecular signals within cells is crucial for cellular functions. Here, as a model to study the polarized spatial distribution of molecular activities, we used cells on micro-patterned strips of fibronectin with one end free and the other end contacting a neighboring cell. Phosphoinositide 3-kinase (PI3K) and the small GTPase Rac display greater activity at the free end, whereas myosin II light chain (MLC) and actin filaments are enriched near the intercellular junction. PI3K and Rac polarization depend specifically on the N-cadherin-p120ctn complex, whereas MLC and actin filament polarization depend on the N-cadherin-β-catenin complex. Integrins promote high PI3K/Rac activities at the free end, and the N-cadherin–p120ctn complex excludes integrin α5 at the junctions to suppress local PI3K and Rac activity. We hence conclude that N-cadherin couples with distinct effectors to polarize PI3K/Rac and MLC/actin filaments in migrating cells.
Collapse
|
46
|
Abstract
In mammalian testes, the blood-testis barrier (BTB), created by specialized junctions between Sertoli cells near the basement membrane of the seminiferous epithelium, provides an indispensable immune-privileged microenvironment for spermatid development. However, the BTB must experience restructuring during the epithelial cycle to facilitate the transit of preleptotene spermatocytes upon the testosterone-induced new TJ fibrils forming behind these cells, which is intimately related to the extensive dynamics of junction protein complexes between Sertoli cells. As key regulators of protein traffic, Rab GTPases participate in delivery of proteins between distinct cellular sites and cross talk with proteins that constitute tight junction and adherens junction. Using primarily cultured Sertoli cells in vitro with an established tight junction permeability barrier that mimics the BTB in vivo, RAB13 was shown to decrease during the testosterone-induced TJ integrity enhancement, accompanied with an increment in protein kinase A (PKA) activity. Furthermore, knockdown of Rab13 was found to resemble the effect of testosterone on Sertoli cell TJ permeability by reinforcing filamentous actin and occludin distribution at the cell-cell interface and promoting the direct interaction between ZO-1 and occludin. Interestingly, the effects of testosterone and Rab13 knockdown on Sertoli cell epithelium were revealed to be antagonized by PKA activity inhibition. In summary, RAB13 serves as a regulatory component in the assembly and restructuring of the TJ fibrils between adjacent Sertoli cells.
Collapse
Affiliation(s)
- Wenhui Su
- Department of Biochemistry and Molecular Biology, Basic Medical College.
| | | |
Collapse
|
47
|
Barrera M, Bahamondes V, Sepúlveda D, Quest A, Castro I, Cortés J, Aguilera S, Urzúa U, Molina C, Pérez P, Ewert P, Alliende C, Hermoso M, González S, Leyton C, González M. Sjögren's syndrome and the epithelial target: A comprehensive review. J Autoimmun 2013; 42:7-18. [DOI: 10.1016/j.jaut.2013.02.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 02/11/2013] [Indexed: 12/12/2022]
|
48
|
Jung JJ, Inamdar SM, Tiwari A, Ye D, Lin F, Choudhury A. Syntaxin 16 regulates lumen formation during epithelial morphogenesis. PLoS One 2013; 8:e61857. [PMID: 23626741 PMCID: PMC3633931 DOI: 10.1371/journal.pone.0061857] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/15/2013] [Indexed: 11/19/2022] Open
Abstract
The formation and maintenance of cell-cell junctions, both under physiological and pathological conditions, requires the targeting and trafficking of junctional proteins. Proteins of the syntaxin (Stx)-family localize to a variety of subcellular membranes and contribute to intracellular transport of cargo by regulating vesicle fusion events at these sites. Unlike plasma membrane localized Stxs, the roles of endosome- and Golgi-localized stx proteins in epithelial morphogenesis are less understood. Here we show that Stx16- an endosome- and Golgi-localized target-membrane soluble N-ethylmaleimide attachment protein receptor (t-SNARE) that plays a role in membrane trafficking between these compartments - is essential for lumen development. In cultured Madin Darby Canine Kidney (MDCK) cells, Stx16 was selectively upregulated as sparsely plated cells attained confluency. Stx16-depleted confluent monolayers consistently showed lower transepithelial resistance than control monolayers, and failed to maintain endogenous and ectopically expressed E-cadherin at the adherens junctions due to decreased recycling. We further found that whereas cysts formed by MDCK cells cultured in Matrigel have a single hollow lumen, those formed by stx16-depleted counterparts had multiple lumens, due to abnormal orientiation of the mitotic spindle. Finally, a similar role for stx16 function in vivo is indicated by our analysis of pronephric-duct development in zebrafish expressing the claudinB:lynGFP transgene; lack of stx16 function in this structure (in stx16-morphant embryos) led to the development of enlarged, torturous pronephric ducts with more than one lumen. Taken together, our in vitro and in vivo studies establish a role for Stx16 in maintaining the integrity of cell-cell junctions, and thereby in morphogenesis of the kidney epithelial lumen.
Collapse
Affiliation(s)
- Jae-Joon Jung
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Shivangi M. Inamdar
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Ajit Tiwari
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Ding Ye
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Fang Lin
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Amit Choudhury
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
- Orthopaedics and Rehabilitation, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
49
|
The Rho-kinase inhibitor fasudil restores normal motor nerve conduction velocity in diabetic rats by assuring the proper localization of adhesion-related molecules in myelinating Schwann cells. Exp Neurol 2013; 247:438-46. [PMID: 23337773 DOI: 10.1016/j.expneurol.2013.01.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 01/07/2013] [Accepted: 01/10/2013] [Indexed: 01/12/2023]
Abstract
The Rho/Rho-kinase signaling pathway has been shown to be involved in the complications of diabetes. In this study, we found that fasudil, a specific Rho-kinase inhibitor, had a beneficial effect on the motor nerve conduction velocity (MNCV), which is delayed in rats with streptozotocin (STZ)-induced diabetes. Cadherin-dependent adherens junctions (AJs) in myelinating Schwann cells, necessary for proper myelin formation and rapid propagation of action potentials, are regulated by Rho/Rho-kinase signaling. These AJ structures are maintained by E-cadherin and catenin complexes such as β-catenin and p120 catenin. To elucidate the mechanism underlying the effect of fasudil on MNCV, we examined alterations in AJ structure in the peripheral nerves of the experimental rats. Our results showed that the activities of Rho and Rho-kinase increased simultaneously in the sciatic nerves of the diabetic rats. Fasudil restored the MNCV by suppressing the up-regulation of the Rho-kinase. In the diabetic state, enhanced Rho and Rho-kinase activity reduced p120 catenin expression and altered the distribution of p120 catenin and E-cadherin, which are normally localized in the paranodal compartment of the nodes of Ranvier and Schmidt-Lanterman incisures where autotypic AJs stabilize myelin structure. Fasudil restored normal p120 catenin expression and the distribution of p120 catenin and E-cadherin in the myelin sheath. In conclusion, reduced expression and altered distribution of the adhesion molecules in the myelin sheath might contribute to the slowing of the MNCV in the diabetic rats. Fasudil, through its effect on the distribution of the adhesion-related molecules, might prevent slowing of the MNCV.
Collapse
|
50
|
Mori Y, Yagi S, Sakurai A, Matsuda M, Kiyokawa E. Insufficient ability of Rac1b to perturb cystogenesis. Small GTPases 2013; 4:9-15. [PMID: 23411476 DOI: 10.4161/sgtp.23311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Rac1b is frequently expressed in a number of human cancer cells. It is still unclear, however, whether Rac1b causes morphological abnormalities in epithelial tissues. To investigate whether Rac1b induces morphological changes in 3-dimensional epithelial structures, we utilized an auxin-dependent protein expression system, which enabled us to rapidly induce and evaluate Rac1b function in MDCK (Madin-Darby Canine Kidney) cysts, a model for polarized epithelial structure. Cells carrying the wild-type Rac1, Rac1b and constitutively active Rac1V12 gene were morphologically indistinguishable from normal, when their coding proteins were not expressed. However, upon protein induction, Rac1V12, but not the wild-type Rac1 or Rac1b, significantly induced the luminal cell accumulation. Live cell imaging with cell cycle indicators showed that expression of Rac1V12, but not the wild-type Rac1 or Rac1b, promoted cell cycle progression. From these results, we concluded that the expression of Rac1b per se cannot induce cell proliferation. Rather, it is considered that Rac1b expression may participate in progression of malignancy.
Collapse
Affiliation(s)
- Yoshifumi Mori
- Department of Pathology and Biology of Diseases; Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | |
Collapse
|