1
|
Micovic K, Canuel A, Remtulla A, Chuyen A, Byrsan M, McGarry DJ, Olson MF. Mical1 deletion in tyrosinase expressing cells affects mouse running gaits. GENES, BRAIN, AND BEHAVIOR 2024; 23:e70004. [PMID: 39344934 PMCID: PMC11440367 DOI: 10.1111/gbb.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
Neuronal development is a highly regulated process that is dependent on the correct coordination of cellular responses to extracellular cues. In response to semaphorin axon guidance proteins, the MICAL1 protein is stimulated to produce reactive oxygen species that oxidize actin on specific methionine residues, leading to filamentous actin depolymerization and consequent changes in neuronal growth cone dynamics. Crossing genetically modified mice homozygous for floxed Mical1 (Mical1fl/fl) alleles with transgenic mice expressing Cre recombinase under the control of a tyrosinase gene enhancer/promoter (Tyr::Cre) enabled conditional Mical1 deletion. Immunohistochemical analysis showed Mical1 expression in the cerebellum, which plays a prominent role in the coordination of motor movements, with reduced Mical1 expression in Mical1fl/fl mice co-expressing Tyr::Cre. Analysis of the gaits of mice running on a treadmill showed that both male and female Mical1fl/fl, Tyr::Cre mutant mice had significant alterations to their striding patterns relative to wild-type mice, although the specific aspects of their altered gaits differed between the sexes. Additional motor tests that involved movement on a rotating rod, descending a vertical pole, or crossing a balance beam did not show significant differences between the genotypes, suggesting that the effect of the Mical1fl/fl, Tyr::Cre genetic modifications was only manifested during specific highly coordinated movements that contribute to running. These findings indicate that there is a behavioral consequence in Mical1fl/fl, Tyr::Cre mutant mice that affects motor control as manifested by alterations in their gait.
Collapse
Affiliation(s)
- Katarina Micovic
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Alicia Canuel
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Aasiya Remtulla
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
| | - Alexandre Chuyen
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Margarita Byrsan
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| | - David J. McGarry
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Michael F. Olson
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
- Department of Pharmacology and ToxicologyUniversity of TorontoTorontoOntarioCanada
- Biomedical Engineering ProgramToronto Metropolitan UniversityTorontoOntarioCanada
| |
Collapse
|
2
|
Ishii M, Matsumoto Y, Yamada T, Uga H, Katada T, Ohata S. Targeting dermatophyte Cdc42 and Rac GTPase signaling to hinder hyphal elongation and virulence. iScience 2024; 27:110139. [PMID: 38952678 PMCID: PMC11215307 DOI: 10.1016/j.isci.2024.110139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/18/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024] Open
Abstract
The development of antifungal drugs requires novel molecular targets due to limited treatment options and drug resistance. Through chemical screening and establishment of a novel genetic technique to repress gene expression in Trichophyton rubrum, the primary causal fungus of dermatophytosis, we demonstrated that fungal Cdc42 and Rac GTPases are promising antifungal drug targets. Chemical inhibitors of these GTPases impair hyphal formation, which is crucial for growth and virulence in T. rubrum. Conditional repression of Cdc24, a guanine nucleotide exchange factor for Cdc42 and Rac, led to hyphal growth defects, abnormal cell morphology, and cell death. EHop-016 inhibited the promotion of the guanine nucleotide exchange reaction in Cdc42 and Rac by Cdc24 as well as germination and growth on the nail fragments of T. rubrum and improved animal survival in an invertebrate infection model of T. rubrum. Our results provide a novel antifungal therapeutic target and a potential lead compound.
Collapse
Affiliation(s)
- Masaki Ishii
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Yasuhiko Matsumoto
- Department of Microbiology, Meiji Pharmaceutical University, 2–522–1 Noshio, Kiyose, Tokyo 204–8588, Japan
| | - Tsuyoshi Yamada
- Teikyo University Institute of Medical Mycology, Teikyo University, Hachioji, Tokyo 192-0395, Japan
- Asia International Institute of Infectious Disease Control, Teikyo University, Tokyo 173-0003, Japan
| | - Hideko Uga
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Toshiaki Katada
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Shinya Ohata
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| |
Collapse
|
3
|
Dent LG, Curry N, Sparks H, Bousgouni V, Maioli V, Kumar S, Munro I, Butera F, Jones I, Arias-Garcia M, Rowe-Brown L, Dunsby C, Bakal C. Environmentally dependent and independent control of 3D cell shape. Cell Rep 2024; 43:114016. [PMID: 38636520 DOI: 10.1016/j.celrep.2024.114016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
How cancer cells determine their shape in response to three-dimensional (3D) geometric and mechanical cues is unclear. We develop an approach to quantify the 3D cell shape of over 60,000 melanoma cells in collagen hydrogels using high-throughput stage-scanning oblique plane microscopy (ssOPM). We identify stereotypic and environmentally dependent changes in shape and protrusivity depending on whether a cell is proximal to a flat and rigid surface or is embedded in a soft environment. Environmental sensitivity metrics calculated for small molecules and gene knockdowns identify interactions between the environment and cellular factors that are important for morphogenesis. We show that the Rho guanine nucleotide exchange factor (RhoGEF) TIAM2 contributes to shape determination in environmentally independent ways but that non-muscle myosin II, microtubules, and the RhoGEF FARP1 regulate shape in ways dependent on the microenvironment. Thus, changes in cancer cell shape in response to 3D geometric and mechanical cues are modulated in both an environmentally dependent and independent fashion.
Collapse
Affiliation(s)
- Lucas G Dent
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Nathan Curry
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Hugh Sparks
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Vicky Bousgouni
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Vincent Maioli
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Sunil Kumar
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Ian Munro
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Francesca Butera
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Ian Jones
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Mar Arias-Garcia
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Leo Rowe-Brown
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK
| | - Chris Dunsby
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK.
| | - Chris Bakal
- Dynamical Cell Systems Group, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| |
Collapse
|
4
|
Fox S, Gaudreau-LaPierre A, Reshke R, Podinic I, Gibbings DJ, Trinkle-Mulcahy L, Copeland JW. Identification of an FMNL2 Interactome by Quantitative Mass Spectrometry. Int J Mol Sci 2024; 25:5686. [PMID: 38891874 PMCID: PMC11171801 DOI: 10.3390/ijms25115686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Formin Homology Proteins (Formins) are a highly conserved family of cytoskeletal regulatory proteins that participate in a diverse range of cellular processes. FMNL2 is a member of the Diaphanous-Related Formin sub-group, and previous reports suggest FMNL2's role in filopodia assembly, force generation at lamellipodia, subcellular trafficking, cell-cell junction assembly, and focal adhesion formation. How FMNL2 is recruited to these sites of action is not well understood. To shed light on how FMNL2 activity is partitioned between subcellular locations, we used biotin proximity labeling and proteomic analysis to identify an FMNL2 interactome. The interactome identified known and new FMNL2 interacting proteins with functions related to previously described FMNL2 activities. In addition, our interactome predicts a novel connection between FMNL2 and extracellular vesicle assembly. We show directly that FMNL2 protein is present in exosomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - John W. Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (S.F.)
| |
Collapse
|
5
|
Ji C, Huang Y. Durotaxis and negative durotaxis: where should cells go? Commun Biol 2023; 6:1169. [PMID: 37973823 PMCID: PMC10654570 DOI: 10.1038/s42003-023-05554-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
Durotaxis and negative durotaxis are processes in which cell migration is directed by extracellular stiffness. Durotaxis is the tendency of cells to migrate toward stiffer areas, while negative durotaxis occurs when cells migrate toward regions with lower stiffness. The mechanisms of both processes are not yet fully understood. Additionally, the connection between durotaxis and negative durotaxis remains unclear. In this review, we compare the mechanisms underlying durotaxis and negative durotaxis, summarize the basic principles of both, discuss the possible reasons why some cell types exhibit durotaxis while others exhibit negative durotaxis, propose mechanisms of switching between these processes, and emphasize the challenges in the investigation of durotaxis and negative durotaxis.
Collapse
Affiliation(s)
- Congcong Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuxing Huang
- Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| |
Collapse
|
6
|
Haage A, Tanentzapf G. Analysis of Integrin-Dependent Melanoblast Migration During Development. Methods Mol Biol 2023; 2608:207-221. [PMID: 36653710 DOI: 10.1007/978-1-0716-2887-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The neural crest is a transient embryonic structure that gives rise to a number of important cell types and tissues, including most of the peripheral and enteric nervous systems, pigment-producing skin cells known as melanocytes, and many craniofacial structures. Melanoblasts, the precursors of melanocytes, are derived from the so-called trunk neural crest cells. These cells delaminate and migrate along a dorsolateral pathway to colonize their final destination in the skin, and consequently, defects in melanoblast migration result in pigmentation defects. Studying melanocyte migration is a topic of great interest due to the involvement of melanocytes in highly metastatic skin cancer. A role for integrin-mediated adhesion is well established in neural crest migration, and our recent work has provided direct evidence for a key role for integrin-based adhesion in melanocyte migration. Imaging of melanoblast migration in the context of intact skin has proven to be a particularly powerful tool to study integrin-based adhesion during melanoblast migration. Here, we describe the use of skin explants combined with genetically encoded markers for melanocytes and high-resolution live imaging as a powerful and informative approach to analyze melanoblast migration in an ex vivo context.
Collapse
Affiliation(s)
- Amanda Haage
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA.
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
van den Bosch QCC, Nguyen JQN, Brands T, van den Bosch TPP, Verdijk RM, Paridaens D, Naus NC, de Klein A, Kiliç E, Brosens E. FOXD1 Is a Transcription Factor Important for Uveal Melanocyte Development and Associated with High-Risk Uveal Melanoma. Cancers (Basel) 2022; 14:cancers14153668. [PMID: 35954332 PMCID: PMC9367502 DOI: 10.3390/cancers14153668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/12/2022] [Accepted: 07/22/2022] [Indexed: 11/27/2022] Open
Abstract
Simple Summary Despite successful treatment of primary uveal melanoma (UM), metastases still occur in approximately 50% of the patients. Unfortunately, little is known about the mechanism behind metastasized UM. By reanalyzing publicly available single-cell RNA sequencing data of embryonic zebrafish larvae and validating the results with UM data, we have identified five transcription regulators of interest: ELL2, KDM5B, REXO4, RBFOX2 and FOXD1. The most significant finding is FOXD1, which is nearly exclusively expressed in high-risk UM and is associated with poor survival. FOXD1 is a novel gene which could be involved in the metastatic capability of UM. Elucidating its function and role in metastatic UM could help to understand and develop treatment for UM. Abstract Uveal melanoma (UM) is a deadly ocular malignancy, originating from uveal melanocytes. Although much is known regarding prognostication in UM, the exact mechanism of metastasis is mostly unknown. Metastatic tumor cells are known to express a more stem-like RNA profile which is seen often in cell-specific embryonic development to induce tumor progression. Here, we identified novel transcription regulators by reanalyzing publicly available single cell RNA sequencing experiments. We identified five transcription regulators of interest: ELL2, KDM5B, REXO4, RBFOX2 and FOXD1. Our most significant finding is FOXD1, as this gene is nearly exclusively expressed in high-risk UM and its expression is associated with a poor prognosis. Even within the BAP1-mutated UM, the expression of FOXD1 is correlated with poor survival. FOXD1 is a novel factor which could potentially be involved in the metastatic capacity of high-risk UM. Elucidating the function of FOXD1 in UM could provide insight into the malignant transformation of uveal melanocytes, especially in high-risk UM.
Collapse
Affiliation(s)
- Quincy C. C. van den Bosch
- Department of Ophthalmology, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Q.C.C.v.d.B.); (J.Q.N.N.); (T.B.); (N.C.N.)
- Department of Clinical Genetics, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands;
| | - Josephine Q. N. Nguyen
- Department of Ophthalmology, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Q.C.C.v.d.B.); (J.Q.N.N.); (T.B.); (N.C.N.)
- Department of Clinical Genetics, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands;
| | - Tom Brands
- Department of Ophthalmology, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Q.C.C.v.d.B.); (J.Q.N.N.); (T.B.); (N.C.N.)
- Department of Clinical Genetics, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands;
| | - Thierry P. P. van den Bosch
- Department of Pathology, Section Ophthalmic Pathology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (T.P.P.v.d.B.); (R.M.V.)
| | - Robert M. Verdijk
- Department of Pathology, Section Ophthalmic Pathology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (T.P.P.v.d.B.); (R.M.V.)
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Dion Paridaens
- The Rotterdam Eye Hospital, 3011 BH Rotterdam, The Netherlands;
| | - Nicole C. Naus
- Department of Ophthalmology, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Q.C.C.v.d.B.); (J.Q.N.N.); (T.B.); (N.C.N.)
| | - Annelies de Klein
- Department of Clinical Genetics, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands;
| | - Emine Kiliç
- Department of Ophthalmology, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Q.C.C.v.d.B.); (J.Q.N.N.); (T.B.); (N.C.N.)
- Correspondence: (E.K.); (E.B.); Tel.: +31-107030683 (E.B.)
| | - Erwin Brosens
- Department of Clinical Genetics, Erasmus MC Cancer Center, Erasmus MC University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands;
- Correspondence: (E.K.); (E.B.); Tel.: +31-107030683 (E.B.)
| |
Collapse
|
8
|
Wang XM, Liu WL, Chen Y, Pang XX, Wang YH, Wu M, Shi B, Li CH. Lithium-induced overexpression of β-catenin delays murine palatal shelf elevation by Cdc-42 mediated F-actin remodeling in mesenchymal cells. Birth Defects Res 2020; 113:427-438. [PMID: 33300673 DOI: 10.1002/bdr2.1853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/11/2020] [Accepted: 11/19/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Lithium chloride (LiCl) is widely used for the treatment of manic and other psychotic disorders, but the administration of lithium can result in several congenital defects in the fetus, including cleft palate (Meng, Wang, Torensma, Jw & Bian, 2015) (Szabo, 1970). However, the mechanism of Lithium's action as a developmental toxicant in palatogenesis is not well known. METHODS In this study, hematoxylin-eosin and immunofluorescence staining were employed to evaluate the phenotypes and the expression of related markers in the LiCl-treated mice model. The palatal mesenchymal cells were cultured in vitro, and stimulated with LiCl or SKL2000, and co-treated with CASIN. β-catenin protein and other cytoskeleton associated markers were evaluated by Western blotting. RESULTS We found that Lithium disrupted palate elevation by increasing the expression of β-catenin in C57BL/6J mice with the high incidence of cleft palate (62.5%). LiCl disturbed the F-actin responsible for cytoskeletal remodeling in mesenchymal cells, which proved to be essential in generating the elevating force during palatal elevation. Additionally, our Western blotting analysis revealed that the overexpression of β-catenin resulted in up-regulation of Cdc42, which mediated the downstream F-actin synthesis. CONCLUSIONS We concluded the LiCl-induced β-catenin overexpression delayed murine palatal shelf elevation by disturbing Cdc42 mediated F-actin cytoskeleton synthesis in the palatal mesenchyme.
Collapse
Affiliation(s)
- Xiao-Ming Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei-Long Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiao-Xiao Pang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Hong Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Min Wu
- Department of Biomedical Sciences School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Bing Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Cheng-Hao Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Papalazarou V, Swaminathan K, Jaber-Hijazi F, Spence H, Lahmann I, Nixon C, Salmeron-Sanchez M, Arnold HH, Rottner K, Machesky LM. The Arp2/3 complex is crucial for colonisation of the mouse skin by melanoblasts. Development 2020; 147:dev194555. [PMID: 33028610 PMCID: PMC7687863 DOI: 10.1242/dev.194555] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/22/2020] [Indexed: 01/10/2023]
Abstract
The Arp2/3 complex is essential for the assembly of branched filamentous actin, but its role in physiology and development is surprisingly little understood. Melanoblasts deriving from the neural crest migrate along the developing embryo and traverse the dermis to reach the epidermis, colonising the skin and eventually homing within the hair follicles. We have previously established that Rac1 and Cdc42 direct melanoblast migration in vivo We hypothesised that the Arp2/3 complex might be the main downstream effector of these small GTPases. Arp3 depletion in the melanocyte lineage results in severe pigmentation defects in dorsal and ventral regions of the mouse skin. Arp3 null melanoblasts demonstrate proliferation and migration defects and fail to elongate as their wild-type counterparts. Conditional deletion of Arp3 in primary melanocytes causes improper proliferation, spreading, migration and adhesion to extracellular matrix. Collectively, our results suggest that the Arp2/3 complex is absolutely indispensable in the melanocyte lineage in mouse development, and indicate a significant role in developmental processes that require tight regulation of actin-mediated motility.
Collapse
Affiliation(s)
- Vassilis Papalazarou
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, Switchback Road, Bearsden, Glasgow G61 1QH, UK
- Centre for the Cellular Microenvironment, University of Glasgow, Glasgow G12 8LT, UK
| | - Karthic Swaminathan
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Farah Jaber-Hijazi
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Heather Spence
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Ines Lahmann
- Cell and Molecular Biology, Institute of Biochemistry and Biotechnology, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Colin Nixon
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | | | - Hans-Henning Arnold
- Cell and Molecular Biology, Institute of Biochemistry and Biotechnology, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Laura M Machesky
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| |
Collapse
|
10
|
McMenamin PG, Shields GT, Seyed-Razavi Y, Kalirai H, Insall RH, Machesky LM, Coupland SE. Melanoblasts Populate the Mouse Choroid Earlier in Development Than Previously Described. Invest Ophthalmol Vis Sci 2020; 61:33. [PMID: 32797202 PMCID: PMC7441366 DOI: 10.1167/iovs.61.10.33] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/14/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Human choroidal melanocytes become evident in the last trimester of development, but very little is known about them. To better understand normal and diseased choroidal melanocyte biology we examined their precursors, melanoblasts (MB), in mouse eyes during development, particularly their relation to the developing vasculature and immune cells. Methods Naïve B6(Cg)-Tyrc-2J/J albino mice were used between embryonic (E) day 15.5 and postnatal (P) day 8, with adult controls. Whole eyes, posterior segments, or dissected choroidal wholemounts were stained with antibodies against tyrosinase-related protein 2, ionized calcium binding adaptor molecule-1 or isolectin B4, and examined by confocal microscopy. Immunoreactive cell numbers in the choroid were quantified with Imaris. One-way ANOVA with Tukey's post hoc test assessed statistical significance. Results Small numbers of MB were present in the presumptive choroid at E15.5 and E18.5. The density significantly increased between E18.5 (381.4 ± 45.8 cells/mm2) and P0 (695.2 ± 87.1 cells/mm2; P = 0.032). In postnatal eyes MB increased in density and formed multiple layers beneath the choriocapillaris. MB in the periocular mesenchyme preceded the appearance of vascular structures at E15.5. Myeloid cells (Ionized calcium binding adaptor molecule-1-positive) were also present at high densities from this time, and attained adult-equivalent densities by P8 (556.4 ± 73.6 cells/mm2). Conclusions We demonstrate that choroidal MB and myeloid cells are both present at very early stages of mouse eye development (E15.5). Although MB and vascularization seemed to be unlinked early in choroidal development, they were closely associated at later stages. MB did not migrate into the choroid in waves, nor did they have a consistent relationship with nerves.
Collapse
Affiliation(s)
- Paul G. McMenamin
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Graham T. Shields
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Yashar Seyed-Razavi
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Robert H. Insall
- CRUK Beatson Institute, Bearsden, University of Glasgow, Glasgow, G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Laura M. Machesky
- CRUK Beatson Institute, Bearsden, University of Glasgow, Glasgow, G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sarah E. Coupland
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
11
|
Haage A, Wagner K, Deng W, Venkatesh B, Mitchell C, Goodwin K, Bogutz A, Lefebvre L, Van Raamsdonk CD, Tanentzapf G. Precise coordination of cell-ECM adhesion is essential for efficient melanoblast migration during development. Development 2020; 147:dev.184234. [PMID: 32580934 DOI: 10.1242/dev.184234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 06/08/2020] [Indexed: 01/07/2023]
Abstract
Melanoblasts disperse throughout the skin and populate hair follicles through long-range cell migration. During migration, cells undergo cycles of coordinated attachment and detachment from the extracellular matrix (ECM). Embryonic migration processes that require cell-ECM attachment are dependent on the integrin family of adhesion receptors. Precise regulation of integrin-mediated adhesion is important for many developmental migration events. However, the mechanisms that regulate integrin-mediated adhesion in vivo in melanoblasts are not well understood. Here, we show that autoinhibitory regulation of the integrin-associated adapter protein talin coordinates cell-ECM adhesion during melanoblast migration in vivo Specifically, an autoinhibition-defective talin mutant strengthens and stabilizes integrin-based adhesions in melanocytes, which impinges on their ability to migrate. Mice with defective talin autoinhibition exhibit delays in melanoblast migration and pigmentation defects. Our results show that coordinated integrin-mediated cell-ECM attachment is essential for melanoblast migration and that talin autoinhibition is an important mechanism for fine-tuning cell-ECM adhesion during cell migration in development.
Collapse
Affiliation(s)
- Amanda Haage
- Department of Biomedical Sciences, University of North Dakota, 1301 N Columbia Rd, Grand Forks, ND 58202, ND, USA
| | - Kelsey Wagner
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Wenjun Deng
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Bhavya Venkatesh
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Caitlin Mitchell
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Aaron Bogutz
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Louis Lefebvre
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Catherine D Van Raamsdonk
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Actin polymerization downstream of integrins: signaling pathways and mechanotransduction. Biochem J 2020; 477:1-21. [PMID: 31913455 DOI: 10.1042/bcj20170719] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/17/2019] [Accepted: 12/16/2019] [Indexed: 01/04/2023]
Abstract
A cell constantly adapts to its environment. Cell decisions to survive, to proliferate or to migrate are dictated not only by soluble growth factors, but also through the direct interaction of the cell with the surrounding extracellular matrix (ECM). Integrins and their connections to the actin cytoskeleton are crucial for monitoring cell attachment and the physical properties of the substratum. Cell adhesion dynamics are modulated in complex ways by the polymerization of branched and linear actin arrays, which in turn reinforce ECM-cytoskeleton connection. This review describes the major actin regulators, Ena/VASP proteins, formins and Arp2/3 complexes, in the context of signaling pathways downstream of integrins. We focus on the specific signaling pathways that transduce the rigidity of the substrate and which control durotaxis, i.e. directed migration of cells towards increased ECM rigidity. By doing so, we highlight several recent findings on mechanotransduction and put them into a broad integrative perspective that is the result of decades of intense research on the actin cytoskeleton and its regulation.
Collapse
|
13
|
Diverse roles of non-muscle myosin II contractility in 3D cell migration. Essays Biochem 2020; 63:497-508. [PMID: 31551323 DOI: 10.1042/ebc20190026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/15/2019] [Accepted: 08/27/2019] [Indexed: 01/13/2023]
Abstract
All is flux, nothing stays still. Heraclitus of Ephesus' characterization of the universe holds true for cells within animals and for proteins within cells. In this review, we examine the dynamics of actin and non-muscle myosin II within cells, and how their dynamics power the movement of cells within tissues. The 3D environment that migrating cells encounter along their path also changes over time, and cells can adopt various mechanisms of motility, depending on the topography, mechanics and chemical composition of their surroundings. We describe the differential spatio-temporal regulation of actin and myosin II-mediated contractility in mesenchymal, lobopodial, amoeboid, and swimming modes of cell migration. After briefly reviewing the biochemistry of myosin II, we discuss the role actomyosin contractility plays in the switch between modes of 3D migration that cells use to adapt to changing environments.
Collapse
|
14
|
Pfisterer K, Levitt J, Lawson CD, Marsh RJ, Heddleston JM, Wait E, Ameer-Beg SM, Cox S, Parsons M. FMNL2 regulates dynamics of fascin in filopodia. J Cell Biol 2020; 219:e201906111. [PMID: 32294157 PMCID: PMC7199847 DOI: 10.1083/jcb.201906111] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/30/2019] [Accepted: 02/20/2020] [Indexed: 12/31/2022] Open
Abstract
Filopodia are peripheral F-actin-rich structures that enable cell sensing of the microenvironment. Fascin is an F-actin-bundling protein that plays a key role in stabilizing filopodia to support efficient adhesion and migration. Fascin is also highly up-regulated in human cancers, where it increases invasive cell behavior and correlates with poor patient prognosis. Previous studies have shown that fascin phosphorylation can regulate F-actin bundling, and that this modification can contribute to subcellular fascin localization and function. However, the factors that regulate fascin dynamics within filopodia remain poorly understood. In the current study, we used advanced live-cell imaging techniques and a fascin biosensor to demonstrate that fascin phosphorylation, localization, and binding to F-actin are highly dynamic and dependent on local cytoskeletal architecture in cells in both 2D and 3D environments. Fascin dynamics within filopodia are under the control of formins, and in particular FMNL2, that binds directly to dephosphorylated fascin. Our data provide new insight into control of fascin dynamics at the nanoscale and into the mechanisms governing rapid cytoskeletal adaptation to environmental changes. This filopodia-driven exploration stage may represent an essential regulatory step in the transition from static to migrating cancer cells.
Collapse
Affiliation(s)
- Karin Pfisterer
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - James Levitt
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
- Microscopy Innovation Centre, King's College London, Guy's Campus, London, UK
| | - Campbell D. Lawson
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Richard J. Marsh
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - John M. Heddleston
- Advanced Imaging Centre, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | - Eric Wait
- Advanced Imaging Centre, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | - Simon Morris Ameer-Beg
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, London, UK
| | - Susan Cox
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| |
Collapse
|
15
|
Dai J, Wang Y, Gong J, Yao Y. Biointerface anisotropy modulates migration of breast cancer cell. Colloids Surf B Biointerfaces 2020; 190:110973. [PMID: 32199258 DOI: 10.1016/j.colsurfb.2020.110973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/24/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022]
Abstract
Migration of cancer cell is a cyclic process, which involves dynamic interaction between extracellular biointerface and cellular responds. In tumors, collagen as extracellular matrix reorganizes biointerface from curl and isotropic fibers to straightened and anisotropic fibers during tumorigenesis, yet how cell migration respond to topography of biointerface is unknown. In this research, we introduced a facile fabrication method on nanofibers of varying topography, which was mimicking the alignment of extracellular nanofibers, to examine the change of cytoskeleton during cell migration. We took advantage of breast carcinoma cell line (MDA-MB-231) for time-lapse imaging analysis. We found that biointerface anisotropy modulated morphology of cell and mediated the pattern of migration. Morphologically, cells on anisotropic nanofiber showed extending spindle shape. The trajectories of migration templated the topographic pattern on biointerface. Besides, aligned nanofiber induced caterpillar-like model of migration through protrusion - retraction cycle, which was indicated by periodical variation of aspect ratio and velocity of cells. The biointerface anisotropy triggered vimentin filaments and microtubule networks preferentially oriented along the alignment of nanofibers. And the velocity of cell mobility by vimentin, β-catenin or CDC42 knockdown was significantly enhanced on aligned nanofibers. Thus, we implied that biointerface anisotropy modulated migration of breast cancer cell and it associated with reorganization of cytoskeleton filaments.
Collapse
Affiliation(s)
- Jing Dai
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China; Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Changning, Shanghai, 200050, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Shijingshan, Beijing, 100049, China
| | - Yiqun Wang
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China; Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China
| | - Jinkang Gong
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yuan Yao
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China.
| |
Collapse
|
16
|
Xie W, Chen C, Han Z, Huang J, Liu X, Chen H, Zhang T, Chen S, Chen C, Lu M, Shen X, Xue X. CD2AP inhibits metastasis in gastric cancer by promoting cellular adhesion and cytoskeleton assembly. Mol Carcinog 2020; 59:339-352. [PMID: 31989722 PMCID: PMC7078920 DOI: 10.1002/mc.23158] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/29/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022]
Abstract
Diffuse gastric cancer (DGC) is a lethal malignancy lacking effective systemic therapy. Among the most provocative recent results in DGC has been that the alter of the cellular cytoskeleton and intercellular adhesion. CD2‐associated protein (CD2AP) is one of the critical proteins regulating cytoskeleton assembly and intercellular adhesion. However, no study has investigated the expression and biological significance of CD2AP in gastric cancer (GC) to date. Therefore, the aim of our study was to explore if the expression of CD2AP is associated with any clinical features of GC and to elucidate the underlying mechanism. Immunohistochemistry of 620 patient tissue samples indicated that the expression of CD2AP is downregulated in DGC. Moreover, a low CD2AP level was indicative of poor patient prognosis. In vitro, forced expression of CD2AP caused a significant decrease in the migration and invasion of GC cells, whereas depletion of CD2AP had the opposite effect. Immunofluorescence analysis indicated that CD2AP promoted cellular adhesion and influenced cell cytoskeleton assembly via interaction with the F‐actin capping protein CAPZA1. Overall, the upregulation of CD2AP could attenuate GC metastasis, suggesting CD2AP as a novel biomarker for the prognosis and treatment of patients with GC.
Collapse
Affiliation(s)
- Wangkai Xie
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Chao Chen
- Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China.,Department of Oncology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Zhejiang Chinese Medical University, Wenzhou, China
| | - Zheng Han
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Huang
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Liu
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hongjun Chen
- Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Teming Zhang
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Sian Chen
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenbin Chen
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| | - Mingdong Lu
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xian Shen
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, School of Basic Medical Science, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Modeling and analysis of melanoblast motion. J Math Biol 2019; 79:2111-2132. [PMID: 31515603 DOI: 10.1007/s00285-019-01422-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 07/05/2019] [Indexed: 10/26/2022]
Abstract
Melanoblast migration is important for embryogenesis and is a key feature of melanoma metastasis. Many studies have characterized melanoblast movement, focusing on statistical properties and have highlighted basic mechanisms of melanoblast motility. We took a slightly different and complementary approach: we previously developed a mathematical model of melanoblast motion that enables the testing of biological assumptions about the displacement of melanoblasts and we created tests to analyze the geometric features of cell trajectories and the specific issue of trajectory interactions. Within this model, we performed simulations and compared the results with experimental data using geometric tests. In this paper, we developed the associated mathematical model and the main focus is to study the crossings between trajectories with new theoretical results about the variation of number of intersection points with respect to the crossing times. Using these results it is possible to study the random nature of displacements and the interactions between trajectories. This analysis has raised new questions, leading to the generation of strong arguments in favor of a trail left behind each moving melanoblast.
Collapse
|
18
|
Crawford M, Leclerc V, Barr K, Dagnino L. Essential Role for Integrin-Linked Kinase in Melanoblast Colonization of the Skin. J Invest Dermatol 2019; 140:425-434.e10. [PMID: 31330146 DOI: 10.1016/j.jid.2019.07.681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 01/16/2023]
Abstract
Melanocytes are pigment-producing cells found in the skin and other tissues. Alterations in the melanocyte lineage give rise to a plethora of human diseases, from neurocristopathies and pigmentation disorders to melanoma. During embryogenesis, neural crest cell subsets give rise to two waves of melanoblasts, which migrate dorsolaterally, hone to the skin, and differentiate into melanocytes. However, the mechanisms that govern colonization of the skin by the first wave of melanoblasts are poorly understood. Here we report that targeted inactivation of the integrin-linked kinase gene in first wave melanoblasts causes defects in the ability of these cells to form long pseudopods, to migrate, and to proliferate in vivo. As a result, integrin-linked kinase-deficient melanoblasts fail to populate normally the developing epidermis and hair follicles. We also show that defects in motility and dendricity occur upon integrin-linked kinase gene inactivation in mature melanocytes, causing abnormalities in cell responses to the extracellular matrix substrates collagen I and laminin 332. Significantly, the ability to form long protrusions in mutant cells in response to collagen is restored in the presence of constitutively active Rac1, suggesting that an integrin-linked kinase-Rac1 nexus is likely implicated in melanocytic cell establishment, dendricity, and functions in the skin.
Collapse
Affiliation(s)
- Melissa Crawford
- Department of Physiology and Pharmacology, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Valerie Leclerc
- Department of Physiology and Pharmacology, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Kevin Barr
- Department of Physiology and Pharmacology, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada; Department of Oncology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
19
|
Liu S, Liu Y, Jiang L, Li Z, Lee S, Liu C, Wang J, Zhang J. Recombinant human BMP-2 accelerates the migration of bone marrow mesenchymal stem cells via the CDC42/PAK1/LIMK1 pathway in vitro and in vivo. Biomater Sci 2019; 7:362-372. [PMID: 30484785 DOI: 10.1039/c8bm00846a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Biomaterials are widely used for bone regeneration and fracture repair. The migration of bone marrow mesenchymal stem cells (BMSCs) into bone defect sites or material implantation sites, and their differentiation into osteoblasts, is central to the fracture healing process, and the directional migration of BMSCs depends on cytokines or chemokines at the defect site. BMP-2 can stimulate the migration of a variety of cells, but it remains unclear whether BMSC migration can be induced. To provide evidence for BMP-2-induced BMSC migration, we tested the cytoskeletal changes and migration ability of BMSCs after treatment with recombinant human BMP-2 (rhBMP-2). We also explored the recruitment of BMSCs from the circulatory system using a collagen sponge incorporating rhBMP-2 that was implanted in vivo. Furthermore, to understand the mechanism underlying this migration, we investigated the effect of rhBMP-2 on migration-related signal pathways. Here, we found that, rhBMP-2 treatment significantly increased the migration of BMSCs in vitro via activation of the CDC42/PAK1/LIMK1 pathway, and that this migration could be blocked by silencing CDC42. In vivo, collagen sponge material loaded with rhBMP-2 could recruit BMSCs injected into the circulatory system. Moreover, inhibition using the small interfering RNA for CDC42 led to a significant decrease in the number of BMSCs within the material. In conclusion, our data prove that rhBMP-2 can accelerate BMSC migration via the CDC42/PAK1/LIMK1 pathway both in vivo and in vitro, and therefore provides a foundation for further understanding and application of rhBMP-2-incorporated materials by enhancing BMSC recruitment.
Collapse
Affiliation(s)
- Shuhao Liu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030 People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Bachg AC, Horsthemke M, Skryabin BV, Klasen T, Nagelmann N, Faber C, Woodham E, Machesky LM, Bachg S, Stange R, Jeong HW, Adams RH, Bähler M, Hanley PJ. Phenotypic analysis of Myo10 knockout (Myo10 tm2/tm2) mice lacking full-length (motorized) but not brain-specific headless myosin X. Sci Rep 2019; 9:597. [PMID: 30679680 PMCID: PMC6345916 DOI: 10.1038/s41598-018-37160-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/04/2018] [Indexed: 01/04/2023] Open
Abstract
We investigated the physiological functions of Myo10 (myosin X) using Myo10 reporter knockout (Myo10tm2) mice. Full-length (motorized) Myo10 protein was deleted, but the brain-specific headless (Hdl) isoform (Hdl-Myo10) was still expressed in homozygous mutants. In vitro, we confirmed that Hdl-Myo10 does not induce filopodia, but it strongly localized to the plasma membrane independent of the MyTH4-FERM domain. Filopodia-inducing Myo10 is implicated in axon guidance and mice lacking the Myo10 cargo protein DCC (deleted in colorectal cancer) have severe commissural defects, whereas MRI (magnetic resonance imaging) of isolated brains revealed intact commissures in Myo10tm2/tm2 mice. However, reminiscent of Waardenburg syndrome, a neural crest disorder, Myo10tm2/tm2 mice exhibited pigmentation defects (white belly spots) and simple syndactyly with high penetrance (>95%), and 24% of mutant embryos developed exencephalus, a neural tube closure defect. Furthermore, Myo10tm2/tm2 mice consistently displayed bilateral persistence of the hyaloid vasculature, revealed by MRI and retinal whole-mount preparations. In principle, impaired tissue clearance could contribute to persistence of hyaloid vasculature and syndactyly. However, Myo10-deficient macrophages exhibited no defects in the phagocytosis of apoptotic or IgG-opsonized cells. RNA sequence analysis showed that Myo10 was the most strongly expressed unconventional myosin in retinal vascular endothelial cells and expression levels increased 4-fold between P6 and P15, when vertical sprouting angiogenesis gives rise to deeper layers. Nevertheless, imaging of isolated adult mutant retinas did not reveal vascularization defects. In summary, Myo10 is important for both prenatal (neural tube closure and digit formation) and postnatal development (hyaloid regression, but not retinal vascularization).
Collapse
Affiliation(s)
- Anne C Bachg
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Markus Horsthemke
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Boris V Skryabin
- Department of Medicine, Transgenic Animal and Genetic Engineering Models (TRAM), Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Tim Klasen
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Nina Nagelmann
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Emma Woodham
- Cancer Research UK Beatson Institute, Glasgow University College of Medical, Veterinary and Life Sciences Garscube Estate, Glasgow, G61 1BD, United Kingdom
| | - Laura M Machesky
- Cancer Research UK Beatson Institute, Glasgow University College of Medical, Veterinary and Life Sciences Garscube Estate, Glasgow, G61 1BD, United Kingdom
| | - Sandra Bachg
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149, Münster, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine (IMM), University Hospital Münster, 48149, Münster, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, 48149, Münster, Germany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, 48149, Münster, Germany
| | - Martin Bähler
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Peter J Hanley
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany.
| |
Collapse
|
21
|
Abstract
Formin homology proteins (formins) are a highly conserved family of cytoskeletal remodeling proteins that are involved in a diverse array of cellular functions. Formins are best known for their ability to regulate actin dynamics, but the same functional domains also govern stability and organization of microtubules. It is thought that this dual activity allows them to coordinate the activity of these two major cytoskeletal networks and thereby influence cellular architecture. Golgi ribbon assembly is dependent upon cooperative interactions between actin filaments and cytoplasmic microtubules originating both at the Golgi itself and from the centrosome. Similarly, centrosome assembly, centriole duplication, and centrosome positioning are also reliant on a dialogue between both cytoskeletal networks. As presented in this chapter, a growing body of evidence suggests that multiple formin proteins play essential roles in these central cellular processes.
Collapse
Affiliation(s)
- John Copeland
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
22
|
Wang X, Tang P, Guo F, Zhang M, Yan Y, Huang M, Chen Y, Zhang L, Zhang L. mDia1 and Cdc42 Regulate Activin B-Induced Migration of Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells 2019; 37:150-162. [PMID: 30358011 PMCID: PMC7379979 DOI: 10.1002/stem.2924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022]
Abstract
In a previous study, we have shown that Activin B is a potent chemoattractant for bone marrow-derived mesenchymal stromal cells (BMSCs). As such, the combination of Activin B and BMSCs significantly accelerated rat skin wound healing. In another study, we showed that RhoA activation plays a key role in Activin B-induced BMSC migration. However, the role of the immediate downstream effectors of RhoA in this process is unclear. Here, we demonstrated that mammalian homolog of Drosophila diaphanous-1 (mDia1), a downstream effector of RhoA, exerts a crucial function in Activin B-induced BMSC migration by promoting membrane ruffling, microtubule morphology, and adhesion signaling dynamics. Furthermore, we showed that Activin B does not change Rac1 activity but increases Cdc42 activity in BMSCs. Inactivation of Cdc42 inhibited Activin B-stimulated Golgi reorientation and the cell migration of BMSCs. Furthermore, knockdown of mDia1 affected Activin B-induced BMSC-mediated wound healing in vivo. In conclusion, this study demonstrated that the RhoA-mDia1 and Cdc42 pathways regulate Activin B-induced BMSC migration. This study may help to optimize clinical MSC-based transplantation strategies to promote skin wound healing. Stem Cells 2019;37:150-162.
Collapse
Affiliation(s)
- Xueer Wang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Pei Tang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer BiologyChildren's Hospital Research FoundationCincinnatiOhioUSA
| | - Min Zhang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Yuan Yan
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Mianbo Huang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Yinghua Chen
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Lin Zhang
- Guangdong Provincial Key Laboratory of Tissue Construction and Detection, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
23
|
Velle KB, Campellone KG. Enteropathogenic E. coli relies on collaboration between the formin mDia1 and the Arp2/3 complex for actin pedestal biogenesis and maintenance. PLoS Pathog 2018; 14:e1007485. [PMID: 30550556 PMCID: PMC6310289 DOI: 10.1371/journal.ppat.1007485] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/28/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022] Open
Abstract
Enteropathogenic and enterohemorrhagic E. coli (EPEC and EHEC) are closely related extracellular pathogens that reorganize host cell actin into “pedestals” beneath the tightly adherent bacteria. This pedestal-forming activity is both a critical step in pathogenesis, and it makes EPEC and EHEC useful models for studying the actin rearrangements that underlie membrane protrusions. To generate pedestals, EPEC relies on the tyrosine phosphorylated bacterial effector protein Tir to bind host adaptor proteins that recruit N-WASP, a nucleation-promoting factor that activates the Arp2/3 complex to drive actin polymerization. In contrast, EHEC depends on the effector EspFU to multimerize N-WASP and promote Arp2/3 activation. Although these core pathways of pedestal assembly are well-characterized, the contributions of additional actin nucleation factors are unknown. We investigated potential cooperation between the Arp2/3 complex and other classes of nucleators using chemical inhibitors, siRNAs, and knockout cell lines. We found that inhibition of formins impairs actin pedestal assembly, motility, and cellular colonization for bacteria using the EPEC, but not the EHEC, pathway of actin polymerization. We also identified mDia1 as the formin contributing to EPEC pedestal assembly, as its expression level positively correlates with the efficiency of pedestal formation, and it localizes to the base of pedestals both during their initiation and once they have reached steady state. Collectively, our data suggest that mDia1 enhances EPEC pedestal biogenesis and maintenance by generating seed filaments to be used by the N-WASP-Arp2/3-dependent actin nucleation machinery and by sustaining Src-mediated phosphorylation of Tir. Microbial pathogens that rearrange the host actin cytoskeleton have made valuable contributions to our understanding of cell signaling and movement. The assembly and organization of the actin cytoskeleton is driven by proteins called nucleators, which can be manipulated by bacteria including enteropathogenic Escherichia coli (EPEC), a frequent cause of pediatric diarrhea in developing countries. After ingestion, EPEC adhere tightly to cells of the intestine and hijack the underlying cytoskeleton to create protrusions called actin pedestals. While mechanisms of pedestal assembly involving a nucleator called the Arp2/3 complex have been defined for EPEC, the contribution of additional host nucleators has not been determined. We assessed the roles of several actin nucleators in EPEC pedestals and found that in addition to Arp2/3 complex-mediated nucleation, the formin mDia1 is a key contributor to actin assembly. These findings highlight the importance of nucleator collaboration in pathogenesis, and also advance our understanding of the molecular and cellular basis of EPEC infection, which is ultimately important for the discovery of new drug targets.
Collapse
Affiliation(s)
- Katrina B. Velle
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Kenneth G. Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
24
|
Wen X, Xu X, Sun W, Chen K, Pan M, Wang JM, Bolland SM, Jin T. G-protein-coupled formyl peptide receptors play a dual role in neutrophil chemotaxis and bacterial phagocytosis. Mol Biol Cell 2018; 30:346-356. [PMID: 30540534 PMCID: PMC6589574 DOI: 10.1091/mbc.e18-06-0358] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A dogma of innate immunity is that neutrophils use G-protein–coupled receptors (GPCRs) for chemoattractant to chase bacteria through chemotaxis and then use phagocytic receptors coupled with tyrosine kinases to destroy opsonized bacteria via phagocytosis. Our current work showed that G-protein–coupled formyl peptide receptors (FPRs) directly mediate neutrophil phagocytosis. Mouse neutrophils lacking formyl peptide receptors (Fpr1/2–/–) are defective in the phagocytosis of Escherichia coli and the chemoattractant N-formyl-Met-Leu-Phe (fMLP)-coated beads. fMLP immobilized onto the surface of a bead interacts with FPRs, which trigger a Ca2+ response and induce actin polymerization to form a phagocytic cup for engulfment of the bead. This chemoattractant GPCR/Gi signaling works independently of phagocytic receptor/tyrosine kinase signaling to promote phagocytosis. Thus, in addition to phagocytic receptor-mediated phagocytosis, neutrophils also utilize the chemoattractant GPCR/Gi signaling to mediate phagocytosis to fight against invading bacteria.
Collapse
Affiliation(s)
- Xi Wen
- Chemotaxis Signal Section, National Institutes of Health, Bethesda, MD 20852
| | - Xuehua Xu
- Chemotaxis Signal Section, National Institutes of Health, Bethesda, MD 20852
| | - Wenxiang Sun
- Autoimmunity and Functional Genomics Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852
| | - Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Research Institute at Frederick, Frederick, MD 21702-1201
| | - Miao Pan
- Chemotaxis Signal Section, National Institutes of Health, Bethesda, MD 20852
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Research Institute at Frederick, Frederick, MD 21702-1201
| | - Silvia M Bolland
- Autoimmunity and Functional Genomics Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852
| | - Tian Jin
- Chemotaxis Signal Section, National Institutes of Health, Bethesda, MD 20852
| |
Collapse
|
25
|
Assembling actin filaments for protrusion. Curr Opin Cell Biol 2018; 56:53-63. [PMID: 30278304 DOI: 10.1016/j.ceb.2018.09.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/31/2022]
Abstract
Cell migration entails a plethora of activities combining the productive exertion of protrusive and contractile forces to allow cells to push and squeeze themselves through cell clumps, interstitial tissues or tissue borders. All these activities require the generation and turnover of actin filaments that arrange into specific, subcellular structures. The most prominent structures mediating the protrusion at the leading edges of cells include lamellipodia and filopodia as well as plasma membrane blebs. Moreover, in cells migrating on planar substratum, mechanical support is being provided by an additional, more proximally located structure termed the lamella. Here, we systematically dissect the literature concerning the mechanisms driving actin filament nucleation and elongation in the best-studied protrusive structure, the lamellipodium. Recent work has shed light on open questions in lamellipodium protrusion, including the relative contributions of nucleation versus elongation to the assembly of both individual filaments and the lamellipodial network as a whole. However, much remains to be learned concerning the specificity and relevance of individual factors, their cooperation and their site-specific functions relative to the importance of global actin monomer and filament homeostasis.
Collapse
|
26
|
Molinar-Inglis O, Oliver SL, Rudich P, Kunttas E, McCartney BM. APC2 associates with the actin cortex through a multipart mechanism to regulate cortical actin organization and dynamics in the Drosophila ovary. Cytoskeleton (Hoboken) 2018; 75:323-335. [PMID: 30019417 DOI: 10.1002/cm.21471] [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] [Received: 02/18/2018] [Revised: 05/24/2018] [Accepted: 06/19/2018] [Indexed: 01/20/2023]
Abstract
The actin cortex that lines the plasma membrane of most eukaryotic cells resists external mechanical forces and plays critical roles in a variety of cellular processes including morphogenesis, cytokinesis, and cell migration. Despite its ubiquity and significance, we understand relatively little about the composition, dynamics, and structure of the actin cortex. Adenomatous polyposis coli (APC) proteins regulate the actin and microtubule cytoskeletons through a variety of mechanisms, and in some contexts, APC proteins are cortically enriched. Here we show that APC2 regulates cortical actin dynamics in the follicular epithelium and the nurse cells of the Drosophila ovary and in addition affects the distribution of cortical actin at the apical side of the follicular epithelium. To understand how APC2 influences these properties of the actin cortex, we investigated the mechanisms controlling the cortical localization of APC2 in S2 cultured cells. We previously showed that the N-terminal half of APC2 containing the Armadillo repeats and the C-terminal 30 amino acids (C30) are together necessary and sufficient for APC2's cortical localization. Our work presented here supports a model that cortical localization of APC2 is governed in part by self-association through the N-terminal APC Self-Association Domain (ASAD) and a highly conserved coiled-coil within the C30 domain.
Collapse
Affiliation(s)
- Olivia Molinar-Inglis
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Stacie L Oliver
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Paige Rudich
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Ezgi Kunttas
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Brooke M McCartney
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
27
|
Abstract
DNA sequencing analysis typically involves mapping reads to just one reference genome. Mapping against multiple genomes is necessary, however, when the genome of origin requires confirmation. Mapping against multiple genomes is also advisable for detecting contamination or for identifying sample swaps which, if left undetected, may lead to incorrect experimental conclusions. Consequently, we present FastQ Screen, a tool to validate the origin of DNA samples by quantifying the proportion of reads that map to a panel of reference genomes. FastQ Screen is intended to be used routinely as a quality control measure and for analysing samples in which the origin of the DNA is uncertain or has multiple sources.
Collapse
Affiliation(s)
| | - Simon Andrews
- Bioinformatics, Babraham Institute, Cambridge, CB22 3AT, UK
| |
Collapse
|
28
|
Abstract
DNA sequencing analysis typically involves mapping reads to just one reference genome. Mapping against multiple genomes is necessary, however, when the genome of origin requires confirmation. Mapping against multiple genomes is also advisable for detecting contamination or for identifying sample swaps which, if left undetected, may lead to incorrect experimental conclusions. Consequently, we present FastQ Screen, a tool to validate the origin of DNA samples by quantifying the proportion of reads that map to a panel of reference genomes. FastQ Screen is intended to be used routinely as a quality control measure and for analysing samples in which the origin of the DNA is uncertain or has multiple sources.
Collapse
Affiliation(s)
| | - Simon Andrews
- Bioinformatics, Babraham Institute, Cambridge, CB22 3AT, UK
| |
Collapse
|
29
|
Tokuo H, Bhawan J, Coluccio LM. Myosin X is required for efficient melanoblast migration and melanoma initiation and metastasis. Sci Rep 2018; 8:10449. [PMID: 29993000 PMCID: PMC6041326 DOI: 10.1038/s41598-018-28717-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/15/2018] [Indexed: 12/20/2022] Open
Abstract
Myosin X (Myo10), an actin-associated molecular motor, has a clear role in filopodia induction and cell migration in vitro, but its role in vivo in mammals is not well understood. Here, we investigate the role of Myo10 in melanocyte lineage and melanoma induction. We found that Myo10 knockout (Myo10KO) mice exhibit a white spot on their belly caused by reduced melanoblast migration. Myo10KO mice crossed with available mice that conditionally express in melanocytes the BRAFV600E mutation combined with Pten silencing exhibited reduced melanoma development and metastasis, which extended medial survival time. Knockdown of Myo10 (Myo10kd) in B16F1 mouse melanoma cell lines decreased lung colonization after tail-vein injection. Myo10kd also inhibited long protrusion (LP) formation by reducing the transportation of its cargo molecule vasodilator-stimulated phosphoprotein (VASP) to the leading edge of migrating cells. These findings provide the first genetic evidence for the involvement of Myo10 not only in melanoblast migration, but also in melanoma development and metastasis.
Collapse
Affiliation(s)
- Hiroshi Tokuo
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Lynne M Coluccio
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA
| |
Collapse
|
30
|
Høye AM, Tolstrup SD, Horton ER, Nicolau M, Frost H, Woo JH, Mauldin JP, Frankel AE, Cox TR, Erler JT. Tumor endothelial marker 8 promotes cancer progression and metastasis. Oncotarget 2018; 9:30173-30188. [PMID: 30046396 PMCID: PMC6059023 DOI: 10.18632/oncotarget.25734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/22/2018] [Indexed: 12/22/2022] Open
Abstract
Every year more than 8 million people suffer from cancer-related deaths worldwide [1]. Metastasis, the spread of cancer to distant sites, accounts for 90% of these deaths. A promising target for blocking tumor progression, without causing severe side effects [2], is Tumor Endothelial Marker 8 (TEM8), an integrin-like cell surface protein expressed predominantly in the tumor endothelium and in cancer cells [3, 4]. Here, we have investigated the role of TEM8 in cancer progression, angiogenesis and metastasis in invasive breast cancer, and validated the main findings and important results in colorectal cancer. We show that the loss of TEM8 in cancer cells results in inhibition of cancer progression, reduction in tumor angiogenesis and reduced metastatic burden in breast cancer mouse models. Furthermore, we show that TEM8 regulates cancer progression by affecting the expression levels of cell cycle-related genes. Taken together, our findings may have broad clinical and therapeutic potential for breast and colorectal primary tumor and metastasis treatment by targeting TEM8.
Collapse
Affiliation(s)
- Anette M Høye
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - Sofie D Tolstrup
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - Edward R Horton
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - Monica Nicolau
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - Helen Frost
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen (UCPH), Copenhagen, Denmark
| | - Jung H Woo
- Baylor Scott and White Health, Temple, TX, USA
| | | | - Arthur E Frankel
- University of South Alabama Mitchell Cancer Institute, Mobile, AL, USA
| | - Thomas R Cox
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen (UCPH), Copenhagen, Denmark.,The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - Janine T Erler
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen (UCPH), Copenhagen, Denmark
| |
Collapse
|
31
|
Actin-Based Cell Protrusion in a 3D Matrix. Trends Cell Biol 2018; 28:823-834. [PMID: 29970282 PMCID: PMC6158345 DOI: 10.1016/j.tcb.2018.06.003] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Cell migration controls developmental processes (gastrulation and tissue patterning), tissue homeostasis (wound repair and inflammatory responses), and the pathobiology of diseases (cancer metastasis and inflammation). Understanding how cells move in physiologically relevant environments is of major importance, and the molecular machinery behind cell movement has been well studied on 2D substrates, beginning over half a century ago. Studies over the past decade have begun to reveal the mechanisms that control cell motility within 3D microenvironments – some similar to, and some highly divergent from those found in 2D. In this review we focus on migration and invasion of cells powered by actin, including formation of actin-rich protrusions at the leading edge, and the mechanisms that control nuclear movement in cells moving in a 3D matrix. Cell migration has been well studied in 2D, but how this relates to movement in physiological 3D tissues and matrix is not clear, particularly in vertebrate interstitial matrix. In 3D matrix cells actin polymerisation directly contributes to the formation of lamellipodia to facilitate migration and invasion (mesenchymal movement), analogous to 2D migration; actomyosin contractility promotes bleb formation to indirectly promote protrusion (amoeboid movement). Mesenchymal migration can be characterised by polymerisation of actin to form filopodial protrusions, in the absence of lamellipodia. Translocation of the nucleus is emerging as a critical step due to the constrictive environment of 3D matrices, and the mechanisms that transmit force to the nucleus and allow movement are beginning to be uncovered.
Collapse
|
32
|
Laurent-Gengoux P, Petit V, Aktary Z, Gallagher S, Tweedy L, Machesky L, Larue L. Simulation of melanoblast displacements reveals new features of developmental migration. Development 2018; 145:dev160200. [PMID: 29769218 PMCID: PMC6031402 DOI: 10.1242/dev.160200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 05/09/2018] [Indexed: 01/17/2023]
Abstract
To distribute and establish the melanocyte lineage throughout the skin and other developing organs, melanoblasts undergo several rounds of proliferation, accompanied by migration through complex environments and differentiation. Melanoblast migration requires interaction with extracellular matrix of the epidermal basement membrane and with surrounding keratinocytes in the developing skin. Migration has been characterized by measuring speed, trajectory and directionality of movement, but there are many unanswered questions about what motivates and defines melanoblast migration. Here, we have established a general mathematical model to simulate the movement of melanoblasts in the epidermis based on biological data, assumptions and hypotheses. Comparisons between experimental data and computer simulations reinforce some biological assumptions, and suggest new ideas for how melanoblasts and keratinocytes might influence each other during development. For example, it appears that melanoblasts instruct each other to allow a homogeneous distribution in the tissue and that keratinocytes may attract melanoblasts until one is stably attached to them. Our model reveals new features of how melanoblasts move and, in particular, suggest that melanoblasts leave a repulsive trail behind them as they move through the skin.
Collapse
Affiliation(s)
- Pascal Laurent-Gengoux
- Laboratory Mathematics in Interaction with Computer Science (MICS), Centrale Supélec, Université Paris Saclay, Gif-sur-Yvette 91190, France
| | - Valérie Petit
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay 91405, France
- Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay 91405, France
- Equipe Labellisée Ligue Contre le Cancer, Orsay 91405, France
| | - Zackie Aktary
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay 91405, France
- Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay 91405, France
- Equipe Labellisée Ligue Contre le Cancer, Orsay 91405, France
| | - Stuart Gallagher
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay 91405, France
- Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay 91405, France
- Equipe Labellisée Ligue Contre le Cancer, Orsay 91405, France
| | - Luke Tweedy
- CRUK Beatson Institute, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Laura Machesky
- CRUK Beatson Institute, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Lionel Larue
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay 91405, France
- Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay 91405, France
- Equipe Labellisée Ligue Contre le Cancer, Orsay 91405, France
| |
Collapse
|
33
|
Abstract
DNA sequencing analysis typically involves mapping reads to just one reference genome. Mapping against multiple genomes is necessary, however, when the genome of origin requires confirmation. Mapping against multiple genomes is also advisable for detecting contamination or for identifying sample swaps which, if left undetected, may lead to incorrect experimental conclusions. Consequently, we present FastQ Screen, a tool to validate the origin of DNA samples by quantifying the proportion of reads that map to a panel of reference genomes. FastQ Screen is intended to be used routinely as a quality control measure and for analysing samples in which the origin of the DNA is uncertain or has multiple sources.
Collapse
Affiliation(s)
| | - Simon Andrews
- Bioinformatics, Babraham Institute, Cambridge, CB22 3AT, UK
| |
Collapse
|
34
|
Abstract
DNA sequencing analysis typically involves mapping reads to just one reference genome. Mapping against multiple genomes is necessary, however, when the genome of origin requires confirmation. Mapping against multiple genomes is also advisable for detecting contamination or for identifying sample swaps which, if left undetected, may lead to incorrect experimental conclusions. Consequently, we present FastQ Screen, a tool to validate the origin of DNA samples by quantifying the proportion of reads that map to a panel of reference genomes. FastQ Screen is intended to be used routinely as a quality control measure and for analysing samples in which the origin of the DNA is uncertain or has multiple sources.
Collapse
Affiliation(s)
| | - Simon Andrews
- Bioinformatics, Babraham Institute, Cambridge, CB22 3AT, UK
| |
Collapse
|
35
|
Heimsath EG, Yim YI, Mustapha M, Hammer JA, Cheney RE. Myosin-X knockout is semi-lethal and demonstrates that myosin-X functions in neural tube closure, pigmentation, hyaloid vasculature regression, and filopodia formation. Sci Rep 2017; 7:17354. [PMID: 29229982 PMCID: PMC5725431 DOI: 10.1038/s41598-017-17638-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/28/2017] [Indexed: 01/07/2023] Open
Abstract
Myosin-X (Myo10) is an unconventional myosin best known for its striking localization to the tips of filopodia. Despite the broad expression of Myo10 in vertebrate tissues, its functions at the organismal level remain largely unknown. We report here the generation of KO-first (Myo10tm1a/tm1a), floxed (Myo10tm1c/tm1c), and KO mice (Myo10tm1d/tm1d). Complete knockout of Myo10 is semi-lethal, with over half of homozygous KO embryos exhibiting exencephaly, a severe defect in neural tube closure. All Myo10 KO mice that survive birth exhibit a white belly spot, all have persistent fetal vasculature in the eye, and ~50% have webbed digits. Myo10 KO mice that survive birth can breed and produce litters of KO embryos, demonstrating that Myo10 is not absolutely essential for mitosis, meiosis, adult survival, or fertility. KO-first mice and an independent spontaneous deletion (Myo10m1J/m1J) exhibit the same core phenotypes. During retinal angiogenesis, KO mice exhibit a ~50% decrease in endothelial filopodia, demonstrating that Myo10 is required to form normal numbers of filopodia in vivo. The Myo10 mice generated here demonstrate that Myo10 has important functions in mammalian development and provide key tools for defining the functions of Myo10 in vivo.
Collapse
Affiliation(s)
- Ernest G Heimsath
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yang-In Yim
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mirna Mustapha
- Department of Otolaryngology, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - John A Hammer
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard E Cheney
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
36
|
Crawford M, Leclerc V, Dagnino L. A reporter mouse model for in vivo tracing and in vitro molecular studies of melanocytic lineage cells and their diseases. Biol Open 2017. [PMID: 28642245 PMCID: PMC5576081 DOI: 10.1242/bio.025833] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alterations in melanocytic lineage cells give rise to a plethora of distinct human diseases, including neurocristopathies, cutaneous pigmentation disorders, loss of vision and hearing, and melanoma. Understanding the ontogeny and biology of melanocytic cells, as well as how they interact with their surrounding environment, are key steps in the development of therapies for diseases that involve this cell lineage. Efforts to culture and characterize primary melanocytes from normal or genetically engineered mouse models have at times yielded contrasting observations. This is due, in part, to differences in the conditions used to isolate, purify and culture these cells in individual studies. By breeding ROSAmT/mG and Tyr::CreERT2 mice, we generated animals in which melanocytic lineage cells are identified through expression of green fluorescent protein. We also used defined conditions to systematically investigate the proliferation and migration responses of primary melanocytes on various extracellular matrix (ECM) substrates. Under our culture conditions, mouse melanocytes exhibit doubling times in the range of 10 days, and retain exponential proliferative capacity for 50-60 days. In culture, these melanocytes showed distinct responses to different ECM substrates. Specifically, laminin-332 promoted cell spreading, formation of dendrites, random motility and directional migration. In contrast, low or intermediate concentrations of collagen I promoted adhesion and acquisition of a bipolar morphology, and interfered with melanocyte forward movements. Our systematic evaluation of primary melanocyte responses emphasizes the importance of clearly defining culture conditions for these cells. This, in turn, is essential for the interpretation of melanocyte responses to extracellular cues and to understand the molecular basis of disorders involving the melanocytic cell lineage.
Collapse
Affiliation(s)
- Melissa Crawford
- Dept. of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Valerie Leclerc
- Dept. of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Lina Dagnino
- Dept. of Physiology and Pharmacology, Children's Health Research Institute and Lawson Health Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| |
Collapse
|
37
|
Wang W, Yi M, Zhang R, Li J, Chen S, Cai J, Zeng Z, Li X, Xiong W, Wang L, Li G, Xiang B. Vimentin is a crucial target for anti-metastasis therapy of nasopharyngeal carcinoma. Mol Cell Biochem 2017; 438:47-57. [PMID: 28744809 DOI: 10.1007/s11010-017-3112-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/01/2017] [Indexed: 12/23/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a unique subtype of head and neck cancer, with tendency to spread to regional lymph nodes and distant organs at early stage. Vimentin, a major cytoskeletal protein constituent of the intermediate filament, plays a critical role in the epithelial-mesenchymal transition. Overexpression of vimentin is considered to be a critical prerequisite for metastasis in numerous human cancers. Therefore, targeting vimentin for cancer therapy has gained a lot of interest. In the present study, we detected vimentin expression in NPC tissues and found that overexpression of vimentin is associated with poor prognosis of NPC patients. Silencing of vimentin in NPC CNE2 cells by RNAi suppresses cells migration and invasion in vitro. However, blocking vimentin did not affect cell proliferation of CNE2 cells. In addition, the in vivo metastatic potential of CNE2 cells transfected with Vimentin shRNA was suppressed in a nude mouse model of pulmonary metastasis. Silencing of Vimentin in CNE2 cells leads to a decrease of microvessel density and VEGF, CD31, MMP2, and MMP9 expressions in pulmonary metastatic tumors. Importantly, we found that it is easier for the tumor cells from the high vimentin-expressing pulmonary metastatic tumors to reinvade the microvessel and to form stable tumor plaques attached to the endothelial cells, which resemble the resource of circulating tumor cells and are very hard to eliminate. However, depletion of vimentin inhibits the formation of vascular tumor plaques. Our findings suggest that RNAi-based vimentin silencing may be a potential and promising anti-metastatic therapeutic strategy for NPC.
Collapse
Affiliation(s)
- Wei Wang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Mei Yi
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Renya Zhang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Junjun Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Shengnan Chen
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Jing Cai
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Bo Xiang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China. .,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
38
|
Bryant D, Johnson A. Meeting report - Intercellular interactions in context: towards a mechanistic understanding of cells in organs. J Cell Sci 2017; 130:2083-2085. [PMID: 28738319 DOI: 10.1242/jcs.205740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Company of Biologists held the workshop 'Intercellular interactions in context: towards a mechanistic understanding of cells in organs' at historic Wiston House in West Sussex, UK, 5-8 February 2017. The meeting brought together around 30 scientists from disparate backgrounds - yet with a common interest of how tissue morphogenesis occurs and its dysregulation leads to pathologies - to intensively discuss their latest research, the current state of the field, as well as any challenges for the future. This report summarises the concepts and challenges that arose as key questions for the fields of cell, cancer and developmental biology. By design of the organizers - Andrew Ewald (John Hopkins University, MA), John Wallingford (University of Texas at Austin, TX) and Peter Friedl (Radboud University, Nijmegen, The Netherlands) - the attendee makeup was cross-sectional: both in terms of career stage and scientific background. This intermingling was mirrored in the workshop format; all participants - irrespective of career stage - were given equal speaking and question time, and all early-career researchers also chaired a session, which promoted an atmosphere for discussions that were open, egalitarian and supportive. This was particularly evident in the scheduled 'out-of-the-box' sessions, which provided an avenue for participants to raise ideas and concepts or to discuss specific problems they wanted feedback or clarification on. In the following, rather than act as court reporters and convey chronological accounting of presentations, we present the questions that arose from the workshop and should be posed to the field at large, by discussing the presentations as they relate to these concepts.
Collapse
Affiliation(s)
- David Bryant
- Cancer Research UK Beatson Institute and Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Aaron Johnson
- Department of Integrative Biology, University of Colorado Denver, Denver, CO 80217, USA
| |
Collapse
|