1
|
Li S, Chen A, Gui J, Zhou H, Zhu L, Mi Y. TLN1: an oncogene associated with tumorigenesis and progression. Discov Oncol 2024; 15:716. [PMID: 39589610 PMCID: PMC11599537 DOI: 10.1007/s12672-024-01593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Talin-1 (TLN1), encoded by the TLN1 gene, is a focal adhesion-related protein capable of binding various proteins in the cytoskeleton. It is also expressed at high levels in many cancers wherein it influences cellular adhesion and the activation of integrins. TLN1 is also capable of promoting tumor cell invasivity, proliferation, and metastatic progression, in addition to being a relevant biomarker and therapeutic target in certain cancers. The present review offers a comprehensive overview of current knowledge regarding TLN1 with respect to its structural properties, functions, and role in tumor development.
Collapse
Affiliation(s)
- Sixin Li
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Anjie Chen
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
2
|
Katoh K. Signal Transduction Mechanisms of Focal Adhesions: Src and FAK-Mediated Cell Response. FRONT BIOSCI-LANDMRK 2024; 29:392. [PMID: 39614431 DOI: 10.31083/j.fbl2911392] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 12/01/2024]
Abstract
Cell-to-substrate adhesion sites, also known as focal adhesion sites (FAs), are complexes of different proteins on the cell surface. FAs play important roles in communication between cells and the extracellular matrix (ECM), leading to signal transduction involving different proteins that ultimately produce the cell response. This cell response involves cell adhesion, migration, motility, cell survival, and cell proliferation. The most important component of FAs are integrins. Integrins are transmembrane proteins that receive signals from the ECM and communicate them to the cytoplasm, thus activating several downstream proteins in a signaling cascade. Cellular Proto-oncogene tyrosine-protein kinase Src (c-Src) and focal adhesion kinase (FAK) are non-receptor tyrosine kinases that functionally interact to promote crucial roles in FAs. c-Src is a tyrosine kinase, activated by autophosphorylation and, in turn, activates another important protein, FAK. Activated FAK directly interacts with the cytoplasmic domain of integrin and activates other FA proteins by attaching to them. These proteins activated by FAK then activate other downstream pathways such as mitogen-activated protein kinase (MAPK) and Akt pathways involved in cell proliferation, migration, and cell survival. Src can induce detachment of FAK from the integrin to increase the focal adhesion turnover. As a result, the Src-FAK complex in FAs is critical for cell adhesion and survival mechanisms. Overexpression of FA proteins has been linked to a variety of pathological disorders, including cancers, growth retardation, and bone deformities. FAK and Src are overexpressed in various cancers. This review, which focuses on the roles of two important signaling proteins, c-Src and FAK, attempts to provide a thorough and up-to-date examination of the signal transduction mechanisms mediated by focal adhesions. The author also described that FAK and Src may serve as potential targets for future therapies against diseases associated with their overexpression, such as certain types of cancer.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, 305-8521 Tsukuba, Japan
| |
Collapse
|
3
|
Chanduri M, Kumar A, Weiss D, Emuna N, Barsukov I, Shi M, Tanaka K, Wang X, Datye A, Kanyo J, Collin F, Lam T, Schwarz UD, Bai S, Nottoli T, Goult BT, Humphrey JD, Schwartz MA. Cellular stiffness sensing through talin 1 in tissue mechanical homeostasis. SCIENCE ADVANCES 2024; 10:eadi6286. [PMID: 39167642 PMCID: PMC11338229 DOI: 10.1126/sciadv.adi6286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Tissue mechanical properties are determined mainly by the extracellular matrix (ECM) and actively maintained by resident cells. Despite its broad importance to biology and medicine, tissue mechanical homeostasis remains poorly understood. To explore cell-mediated control of tissue stiffness, we developed mutations in the mechanosensitive protein talin 1 to alter cellular sensing of ECM. Mutation of a mechanosensitive site between talin 1 rod-domain helix bundles R1 and R2 increased cell spreading and tension exertion on compliant substrates. These mutations promote binding of the ARP2/3 complex subunit ARPC5L, which mediates the change in substrate stiffness sensing. Ascending aortas from mice bearing these mutations showed less fibrillar collagen, reduced axial stiffness, and lower rupture pressure. Together, these results demonstrate that cellular stiffness sensing contributes to ECM mechanics, directly supporting the mechanical homeostasis hypothesis and identifying a mechanosensitive interaction within talin that contributes to this mechanism.
Collapse
Affiliation(s)
- Manasa Chanduri
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Abhishek Kumar
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Dar Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Nir Emuna
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Igor Barsukov
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - Miusi Shi
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Keiichiro Tanaka
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Xinzhe Wang
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511, USA
| | - Amit Datye
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511, USA
| | - Jean Kanyo
- Keck MS & Proteomics Resource, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Florine Collin
- Keck MS & Proteomics Resource, Yale University School of Medicine, New Haven, CT 06510, USA
| | - TuKiet Lam
- Keck MS & Proteomics Resource, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Udo D. Schwarz
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511, USA
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT 06510, USA
| | - Suxia Bai
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Timothy Nottoli
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Benjamin T Goult
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
- School of Biosciences, University of Kent, Canterbury, UK
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
4
|
Chanduri MVL, Kumar A, Weiss D, Emuna N, Barsukov I, Shi M, Tanaka K, Wang X, Datye A, Kanyo J, Collin F, Lam T, Schwarz UD, Bai S, Nottoli T, Goult BT, Humphrey JD, Schwartz MA. Mechanosensing through talin 1 contributes to tissue mechanical homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.03.556084. [PMID: 38328095 PMCID: PMC10849504 DOI: 10.1101/2023.09.03.556084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
It is widely believed that tissue mechanical properties, determined mainly by the extracellular matrix (ECM), are actively maintained. However, despite its broad importance to biology and medicine, tissue mechanical homeostasis is poorly understood. To explore this hypothesis, we developed mutations in the mechanosensitive protein talin1 that alter cellular sensing of ECM stiffness. Mutation of a novel mechanosensitive site between talin1 rod domain helix bundles 1 and 2 (R1 and R2) shifted cellular stiffness sensing curves, enabling cells to spread and exert tension on compliant substrates. Opening of the R1-R2 interface promotes binding of the ARP2/3 complex subunit ARPC5L, which mediates the altered stiffness sensing. Ascending aortas from mice bearing these mutations show increased compliance, less fibrillar collagen, and rupture at lower pressure. Together, these results demonstrate that cellular stiffness sensing regulates ECM mechanical properties. These data thus directly support the mechanical homeostasis hypothesis and identify a novel mechanosensitive interaction within talin that contributes to this mechanism.
Collapse
|
5
|
Wang Y, Huang H, Weng H, Jia C, Liao B, Long Y, Yu F, Nie Y. Talin mechanotransduction in disease. Int J Biochem Cell Biol 2024; 166:106490. [PMID: 37914021 DOI: 10.1016/j.biocel.2023.106490] [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: 07/29/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
Talin protein (Talin 1/2) is a mechanosensitive cytoskeleton protein. The unique structure of the Talin plays a vital role in transmitting mechanical forces. Talin proteins connect the extracellular matrix to the cytoskeleton by linking to integrins and actin, thereby mediating the conversion of mechanical signals into biochemical signals and influencing disease progression as potential diagnostic indicators, therapeutic targets, and prognostic indicators of various diseases. Most studies in recent years have confirmed that mechanical forces also have a crucial role in the development of disease, and Talin has been found to play a role in several diseases. Still, more studies need to be done on how Talin is involved in mechanical signaling in disease. This review focuses on the mechanical signaling of Talin in disease, aiming to summarize the mechanisms by which Talin plays a role in disease and to provide references for further studies.
Collapse
Affiliation(s)
- Yingzi Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Haozhong Huang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Huimin Weng
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Chunsen Jia
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China; Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yongmei Nie
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China.
| |
Collapse
|
6
|
Osuka K, Ohmichi Y, Ohmichi M, Honma S, Suzuki C, Aoyama M, Iwami K, Watanabe Y, Miyachi S. Angiogenesis in the Outer Membrane of Chronic Subdural Hematomas through Thrombin-Cleaved Osteopontin and the Integrin α9 and Integrin β1 Signaling Pathways. Biomedicines 2023; 11:biomedicines11051440. [PMID: 37239111 DOI: 10.3390/biomedicines11051440] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND A chronic subdural hematoma (CSDH) is considered to be an inflammatory and angiogenic disease. The CSDH outer membrane, which contains inflammatory cells, plays an important role in CSDH development. Osteopontin (OPN) is an extracellular matrix protein that is cleaved by thrombin, generating the N-terminal half of OPN, which is prominently involved in integrin signal transduction. We explored the expression of the N-terminal half of OPN in CSDH fluid and the expression of integrins α9 and β1 and the downstream components of the angiogenic signaling pathways in the outer membrane of CSDHs. METHODS Twenty samples of CSDH fluid and eight samples of CSDH outer membrane were collected from patients suffering from CSDHs. The concentrations of the N-terminal half of OPN in CSDH fluid samples were measured using ELISA kits. The expression levels of integrins α9 and β1, vinculin, talin-1, focal adhesion kinase (FAK), paxillin, α-actin, Src and β-actin were examined by Western blot analysis. The expression levels of integrins α9 and β1, FAK and paxillin were also examined by immunohistochemistry. We investigated whether CSDH fluid could activate FAK in cultured endothelial cells in vitro. RESULTS The concentration of the N-terminal half of OPN in CSDH fluid was significantly higher than that in the serum. Western blot analysis confirmed the presence of these molecules. In addition, integrins α9 and β1, FAK and paxillin were localized in the endothelial cells of vessels within the CSDH outer membrane. FAK was significantly phosphorylated immediately after treatment with CSDH fluid. CONCLUSION Our data suggest that the N-terminal half of OPN in CSDH fluid promotes neovascularization in endothelial cells through integrins α9 and β1. The N-terminal half of OPN, which is part of the extracellular matrix, plays a critical role in the promotion of CSDHs.
Collapse
Affiliation(s)
- Koji Osuka
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Yusuke Ohmichi
- Department of Anatomy II, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Mika Ohmichi
- Department of Anatomy II, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Satoru Honma
- Department of Anatomy II, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Chiharu Suzuki
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Masahiro Aoyama
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Kenichiro Iwami
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Yasuo Watanabe
- High Technology Research Center, Pharmacology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawa Gakuen, Machida 194-8543, Japan
| | - Shigeru Miyachi
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| |
Collapse
|
7
|
Gallego-Paez LM, Edwards WJS, Chanduri M, Guo Y, Koorman T, Lee CY, Grexa N, Derksen P, Yan J, Schwartz MA, Mauer J, Goult BT. TLN1 contains a cancer-associated cassette exon that alters talin-1 mechanosensitivity. J Cell Biol 2023; 222:213923. [PMID: 36880935 PMCID: PMC9997659 DOI: 10.1083/jcb.202209010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/08/2023] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Abstract
Talin-1 is the core mechanosensitive adapter protein linking integrins to the cytoskeleton. The TLN1 gene is comprised of 57 exons that encode the 2,541 amino acid TLN1 protein. TLN1 was previously considered to be expressed as a single isoform. However, through differential pre-mRNA splicing analysis, we discovered a cancer-enriched, non-annotated 51-nucleotide exon in TLN1 between exons 17 and 18, which we refer to as exon 17b. TLN1 is comprised of an N-terminal FERM domain, linked to 13 force-dependent switch domains, R1-R13. Inclusion of exon 17b introduces an in-frame insertion of 17 amino acids immediately after Gln665 in the region between R1 and R2 which lowers the force required to open the R1-R2 switches potentially altering downstream mechanotransduction. Biochemical analysis of this isoform revealed enhanced vinculin binding, and cells expressing this variant show altered adhesion dynamics and motility. Finally, we showed that the TGF-β/SMAD3 signaling pathway regulates this isoform switch. Future studies will need to consider the balance of these two TLN1 isoforms.
Collapse
Affiliation(s)
| | | | - Manasa Chanduri
- Departments of Internal Medicine (Cardiology) and Yale Cardiovascular Research Center , New Haven, CT, USA
| | - Yanyu Guo
- Mechanobiology Institute, National University of Singapore , Singapore, Singapore
| | - Thijs Koorman
- Department of Pathology, University Medical Center Utrecht , Utrecht, Netherlands
| | | | - Nina Grexa
- Biomed X Institute (GmbH) , Heidelberg, Germany
| | - Patrick Derksen
- Department of Pathology, University Medical Center Utrecht , Utrecht, Netherlands
| | - Jie Yan
- Mechanobiology Institute, National University of Singapore , Singapore, Singapore.,Department of Physics, National University of Singapore , Singapore, Singapore
| | - Martin A Schwartz
- Departments of Internal Medicine (Cardiology) and Yale Cardiovascular Research Center , New Haven, CT, USA.,Departments of Cell Biology and Biomedical Engineering, Yale School of Medicine , New Haven, CT, USA
| | - Jan Mauer
- Biomed X Institute (GmbH) , Heidelberg, Germany.,Department of Immunology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | |
Collapse
|
8
|
Zhang S, Chong LH, Woon JYX, Chua TX, Cheruba E, Yip AK, Li HY, Chiam KH, Koh CG. Zyxin regulates embryonic stem cell fate by modulating mechanical and biochemical signaling interface. Commun Biol 2023; 6:62. [PMID: 36653484 PMCID: PMC9849324 DOI: 10.1038/s42003-023-04421-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Biochemical signaling and mechano-transduction are both critical in regulating stem cell fate. How crosstalk between mechanical and biochemical cues influences embryonic development, however, is not extensively investigated. Using a comparative study of focal adhesion constituents between mouse embryonic stem cell (mESC) and their differentiated counterparts, we find while zyxin is lowly expressed in mESCs, its levels increase dramatically during early differentiation. Interestingly, overexpression of zyxin in mESCs suppresses Oct4 and Nanog. Using an integrative biochemical and biophysical approach, we demonstrate involvement of zyxin in regulating pluripotency through actin stress fibres and focal adhesions which are known to modulate cellular traction stress and facilitate substrate rigidity-sensing. YAP signaling is identified as an important biochemical effector of zyxin-induced mechanotransduction. These results provide insights into the role of zyxin in the integration of mechanical and biochemical cues for the regulation of embryonic stem cell fate.
Collapse
Affiliation(s)
- Songjing Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lor Huai Chong
- Bioinformatics Institute A*STAR, Singapore, Singapore.,School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jessie Yong Xing Woon
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Theng Xuan Chua
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Ai Kia Yip
- Bioinformatics Institute A*STAR, Singapore, Singapore
| | - Hoi-Yeung Li
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
9
|
Qin R, Melamed S, Yang B, Saxena M, Sheetz MP, Wolfenson H. Tumor Suppressor DAPK1 Catalyzes Adhesion Assembly on Rigid but Anoikis on Soft Matrices. Front Cell Dev Biol 2022; 10:959521. [PMID: 35927990 PMCID: PMC9343699 DOI: 10.3389/fcell.2022.959521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 01/05/2023] Open
Abstract
Cancer cells normally grow on soft surfaces due to impaired mechanosensing of the extracellular matrix rigidity. Upon restoration of proper mechanosensing, cancer cells undergo apoptosis on soft surfaces (anoikis) like most normal cells. However, the link between mechanosensing and activation of anoikis is not clear. Here we show that death associated protein kinase 1 (DAPK1), a tumor suppressor that activates cell death, is directly linked to anoikis activation through rigidity sensing. We find that when rigidity sensing is decreased through inhibition of DAPK1 activity, cells are transformed for growth on soft matrices. Further, DAPK1 catalyzes matrix adhesion assembly and is part of adhesions on rigid surfaces. This pathway involves DAPK1 phosphorylation of tropomyosin1.1, the talin1 head domain, and tyrosine phosphorylation of DAPK1 by Src. On soft surfaces, DAPK1 rapidly dissociates from the adhesion complexes and activates apoptosis as catalyzed by PTPN12 activity and talin1 head. Thus, DAPK1 is important for adhesion assembly on rigid surfaces and the activation of anoikis on soft surfaces through its binding to rigidity-sensing modules.
Collapse
Affiliation(s)
- Ruifang Qin
- Department of Biological Sciences, Columbia University, New York City, NY, United States
| | - Shay Melamed
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Bo Yang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Mayur Saxena
- Department of Biomedical Engineering, Columbia University, New York City, NY, United States
| | - Michael P. Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
- *Correspondence: Haguy Wolfenson, ; Michael P. Sheetz,
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
- *Correspondence: Haguy Wolfenson, ; Michael P. Sheetz,
| |
Collapse
|
10
|
Benk LT, Benk AS, Lira RB, Cavalcanti-Adam EA, Dimova R, Lipowsky R, Geiger B, Spatz JP. Integrin α
IIb
β
3
Activation and Clustering in Minimal Synthetic Cells. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202100094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Lucia T. Benk
- Department of Cellular Biophysics Max Planck Institute for Medical Research Jahnstr. 29 69120 Heidelberg Germany
| | - Amelie S. Benk
- Department of Cellular Biophysics Max Planck Institute for Medical Research Jahnstr. 29 69120 Heidelberg Germany
| | - Rafael B. Lira
- Theory & Bio-Systems Max Planck Institute of Colloids and Interfaces 14424 Potsdam Germany
- Faculty of Science and Engineering Molecular Biophysics Zernike Institute for Advanced Materials 9747 AG Groningen The Netherlands
| | - Elisabetta Ada Cavalcanti-Adam
- Department of Cellular Biophysics Max Planck Institute for Medical Research Jahnstr. 29 69120 Heidelberg Germany
- Max Planck School Matter to Life Jahnstr. 29 69120 Heidelberg Germany
| | - Rumiana Dimova
- Theory & Bio-Systems Max Planck Institute of Colloids and Interfaces 14424 Potsdam Germany
| | - Reinhard Lipowsky
- Theory & Bio-Systems Max Planck Institute of Colloids and Interfaces 14424 Potsdam Germany
- Max Planck School Matter to Life Jahnstr. 29 69120 Heidelberg Germany
| | - Benjamin Geiger
- Department of Molecular Cell Biology Weizmann Institute of Science Rehovot 76100 Israel
| | - Joachim P. Spatz
- Department of Cellular Biophysics Max Planck Institute for Medical Research Jahnstr. 29 69120 Heidelberg Germany
- Max Planck School Matter to Life Jahnstr. 29 69120 Heidelberg Germany
- Institute for Molecular Systems Engineering (IMSE) Heidelberg University 69120 Heidelberg Germany
| |
Collapse
|
11
|
Prostak SM, Robinson KA, Titus MA, Fritz-Laylin LK. The actin networks of chytrid fungi reveal evolutionary loss of cytoskeletal complexity in the fungal kingdom. Curr Biol 2021; 31:1192-1205.e6. [DOI: 10.1016/j.cub.2021.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/05/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
|
12
|
Manipulation of Focal Adhesion Signaling by Pathogenic Microbes. Int J Mol Sci 2021; 22:ijms22031358. [PMID: 33572997 PMCID: PMC7866387 DOI: 10.3390/ijms22031358] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/22/2022] Open
Abstract
Focal adhesions (FAs) serve as dynamic signaling hubs within the cell. They connect intracellular actin to the extracellular matrix (ECM) and respond to environmental cues. In doing so, these structures facilitate important processes such as cell-ECM adhesion and migration. Pathogenic microbes often modify the host cell actin cytoskeleton in their pursuit of an ideal replicative niche or during invasion to facilitate uptake. As actin-interfacing structures, FA dynamics are also intimately tied to actin cytoskeletal organization. Indeed, exploitation of FAs is another avenue by which pathogenic microbes ensure their uptake, survival and dissemination. This is often achieved through the secretion of effector proteins which target specific protein components within the FA. Molecular mimicry of the leucine-aspartic acid (LD) motif or vinculin-binding domains (VBDs) commonly found within FA proteins is a common microbial strategy. Other effectors may induce post-translational modifications to FA proteins through the regulation of phosphorylation sites or proteolytic cleavage. In this review, we present an overview of the regulatory mechanisms governing host cell FAs, and provide examples of how pathogenic microbes have evolved to co-opt them to their own advantage. Recent technological advances pose exciting opportunities for delving deeper into the mechanistic details by which pathogenic microbes modify FAs.
Collapse
|
13
|
Tipton CD, Wolcott RD, Sanford NE, Miller C, Pathak G, Silzer TK, Sun J, Fleming D, Rumbaugh KP, Little TD, Phillips N, Phillips CD. Patient genetics is linked to chronic wound microbiome composition and healing. PLoS Pathog 2020; 16:e1008511. [PMID: 32555671 PMCID: PMC7302439 DOI: 10.1371/journal.ppat.1008511] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
The clinical importance of microbiomes to the chronicity of wounds is widely appreciated, yet little is understood about patient-specific processes shaping wound microbiome composition. Here, a two-cohort microbiome-genome wide association study is presented through which patient genomic loci associated with chronic wound microbiome diversity were identified. Further investigation revealed that alternative TLN2 and ZNF521 genotypes explained significant inter-patient variation in relative abundance of two key pathogens, Pseudomonas aeruginosa and Staphylococcus epidermidis. Wound diversity was lowest in Pseudomonas aeruginosa infected wounds, and decreasing wound diversity had a significant negative linear relationship with healing rate. In addition to microbiome characteristics, age, diabetic status, and genetic ancestry all significantly influenced healing. Using structural equation modeling to identify common variance among SNPs, six loci were sufficient to explain 53% of variation in wound microbiome diversity, which was a 10% increase over traditional multiple regression. Focusing on TLN2, genotype at rs8031916 explained expression differences of alternative transcripts that differ in inclusion of important focal adhesion binding domains. Such differences are hypothesized to relate to wound microbiomes and healing through effects on bacterial exploitation of focal adhesions and/or cellular migration. Related, other associated loci were functionally enriched, often with roles in cytoskeletal dynamics. This study, being the first to identify patient genetic determinants for wound microbiomes and healing, implicates genetic variation determining cellular adhesion phenotypes as important drivers of infection type. The identification of predictive biomarkers for chronic wound microbiomes may serve as risk factors and guide treatment by informing patient-specific tendencies of infection. Chronic, or non-healing, wounds represent a costly burden to patients, and bacterial infection of wounds is an important driver of chronicity. A variety of bacterial species often occur in chronic wounds, but it is unknown why certain species are observed in some wound infections and not others. In this study, genetic variation of wound clinic patients was compared to the bacteria observed in their infected wounds. Through these comparisons, genetic variation in the TLN2 and ZNF521 genes was found to be associated with both the number of bacteria observed in wounds and the abundance of common pathogens (primarily Pseudomonas aeruginosa and Staphylococcus epidermidis). Moreover, Pseudomonas infected wounds were found to have fewer species present and wounds with fewer species were slower to heal. Furthermore, patient genes associated with microbiomes commonly encode proteins known to be important for cellular structures important to healing and to which bacteria directly interact. Experimental investigation of one such gene, TLN2, identified genotype-dependent differences in the expression of functionally different versions of TLN2 that is hypothesized to shape differences in cellular adhesion structures. Finally, a new statistical approach is presented in which patient biomarkers are used to predict the number of species observed during infection. Overall, our results describe how patient genetic variation influence the types of bacteria likely to infect an individual as well as influence healing.
Collapse
Affiliation(s)
- Craig D Tipton
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, United States of America.,RTL Genomics, Lubbock, Texas, United States of America
| | - Randall D Wolcott
- Southwest Regional Wound Care Center, Lubbock, Texas, United States of America
| | - Nicholas E Sanford
- Southwest Regional Wound Care Center, Lubbock, Texas, United States of America
| | - Clint Miller
- Southwest Regional Wound Care Center, Lubbock, Texas, United States of America
| | - Gita Pathak
- Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Talisa K Silzer
- Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Jie Sun
- Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Derek Fleming
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Kendra P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America.,Burn Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, Texas, United States of America
| | - Todd D Little
- Department of Educational Psychology, Texas Tech University, Lubbock, Texas, United States of America.,Optentia Research Focus Area, North West University, Vanderbijlpark, South Africa
| | - Nicole Phillips
- Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Caleb D Phillips
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, United States of America.,Natural Science Research Laboratory, Texas Tech University, Lubbock, Texas, United States of America
| |
Collapse
|
14
|
Miller AE, Hu P, Barker TH. Feeling Things Out: Bidirectional Signaling of the Cell-ECM Interface, Implications in the Mechanobiology of Cell Spreading, Migration, Proliferation, and Differentiation. Adv Healthc Mater 2020; 9:e1901445. [PMID: 32037719 PMCID: PMC7274903 DOI: 10.1002/adhm.201901445] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/10/2020] [Indexed: 12/16/2022]
Abstract
Biophysical cues stemming from the extracellular environment are rapidly transduced into discernible chemical messages (mechanotransduction) that direct cellular activities-placing the extracellular matrix (ECM) as a potent regulator of cell behavior. Dynamic reciprocity between the cell and its associated matrix is essential to the maintenance of tissue homeostasis and dysregulation of both ECM mechanical signaling, via pathological ECM turnover, and internal mechanotransduction pathways contribute to disease progression. This review covers the current understandings of the key modes of signaling used by both the cell and ECM to coregulate one another. By taking an outside-in approach, the inherent complexities and regulatory processes at each level of signaling (ECM, plasma membrane, focal adhesion, and cytoplasm) are captured to give a comprehensive picture of the internal and external mechanoregulatory environment. Specific emphasis is placed on the focal adhesion complex which acts as a central hub of mechanical signaling, regulating cell spreading, migration, proliferation, and differentiation. In addition, a wealth of available knowledge on mechanotransduction is curated to generate an integrated signaling network encompassing the central components of the focal adhesion, cytoplasm and nucleus that act in concert to promote durotaxis, proliferation, and differentiation in a stiffness-dependent manner.
Collapse
Affiliation(s)
- Andrew E Miller
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Ping Hu
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| |
Collapse
|
15
|
Haage A, Goodwin K, Whitewood A, Camp D, Bogutz A, Turner CT, Granville DJ, Lefebvre L, Plotnikov S, Goult BT, Tanentzapf G. Talin Autoinhibition Regulates Cell-ECM Adhesion Dynamics and Wound Healing In Vivo. Cell Rep 2019; 25:2401-2416.e5. [PMID: 30485809 DOI: 10.1016/j.celrep.2018.10.098] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/07/2018] [Accepted: 10/25/2018] [Indexed: 01/15/2023] Open
Abstract
Cells in multicellular organisms are arranged in complex three-dimensional patterns. This requires both transient and stable adhesions with the extracellular matrix (ECM). Integrin adhesion receptors bind ECM ligands outside the cell and then, by binding the protein talin inside the cell, assemble an adhesion complex connecting to the cytoskeleton. The activity of talin is controlled by several mechanisms, but these have not been well studied in vivo. By generating mice containing the activating point mutation E1770A in talin (Tln1), which disrupts autoinhibition, we show that talin autoinhibition controls cell-ECM adhesion, cell migration, and wound healing in vivo. In particular, blocking autoinhibition gives rise to more mature, stable focal adhesions that exhibit increased integrin activation. Mutant cells also show stronger attachment to ECM and decreased traction force. Overall, these results demonstrate that modulating talin function via autoinhibition is an important mechanism for regulating multiple aspects of integrin-mediated cell-ECM adhesion in vivo.
Collapse
Affiliation(s)
- Amanda Haage
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Katharine Goodwin
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Austin Whitewood
- School of Biosciences, Giles Ln, University of Kent, Canterbury CT2 7NZ, UK
| | - Darius Camp
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Aaron Bogutz
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Christopher T Turner
- Department of Pathology and Laboratory Medicine, 2211 Wesbrook Mall, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - David J Granville
- Department of Pathology and Laboratory Medicine, 2211 Wesbrook Mall, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Louis Lefebvre
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sergey Plotnikov
- Department of Cell and Systems Biology, 25 Harbord Street, University of Toronto, Toronto, ON M5S 3H7, Canada
| | - Benjamin T Goult
- School of Biosciences, Giles Ln, University of Kent, Canterbury CT2 7NZ, UK
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
16
|
Kumar A, Shutova MS, Tanaka K, Iwamoto DV, Calderwood DA, Svitkina TM, Schwartz MA. Filamin A mediates isotropic distribution of applied force across the actin network. J Cell Biol 2019; 218:2481-2491. [PMID: 31315944 PMCID: PMC6683746 DOI: 10.1083/jcb.201901086] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/03/2019] [Accepted: 06/17/2019] [Indexed: 12/02/2022] Open
Abstract
In this work, Kumar et al. use their previously developed talin tension sensor to study the immediate response of cells to uniaxial stretch. Tension measurements together with high-resolution electron microscopy reveal a novel role for the actin cross-linking protein filamin A in mediating tensional symmetry within the F-actin network. Cell sensing of externally applied mechanical strain through integrin-mediated adhesions is critical in development and physiology of muscle, lung, tendon, and arteries, among others. We examined the effects of strain on force transmission through the essential cytoskeletal linker talin. Using a fluorescence-based talin tension sensor (TS), we found that uniaxial stretch of cells on elastic substrates increased tension on talin, which was unexpectedly independent of the orientation of the focal adhesions relative to the direction of strain. High-resolution electron microscopy of the actin cytoskeleton revealed that stress fibers (SFs) are integrated into an isotropic network of cortical actin filaments in which filamin A (FlnA) localizes preferentially to points of intersection between SFs and cortical actin. Knockdown (KD) of FlnA resulted in more isolated, less integrated SFs. After FlnA KD, tension on talin was polarized in the direction of stretch, while FlnA reexpression restored tensional symmetry. These data demonstrate that a FlnA-dependent cortical actin network distributes applied forces over the entire cytoskeleton–matrix interface.
Collapse
Affiliation(s)
- Abhishek Kumar
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
| | - Maria S Shutova
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Keiichiro Tanaka
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
| | | | - David A Calderwood
- Department of Pharmacology, Yale University, New Haven, CT.,Department of Cell Biology, Yale University, New Haven, CT
| | | | - Martin A Schwartz
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT .,Department of Cell Biology, Yale University, New Haven, CT.,Department of Biomedical Engineering, Yale University, New Haven, CT
| |
Collapse
|
17
|
"Biological Adhesion" is a Significantly Regulated Molecular Process during Long-Term Primary In Vitro Culture of Oviductal Epithelial Cells (Oecs): A Transcriptomic and Proteomic Study. Int J Mol Sci 2019; 20:ijms20143387. [PMID: 31295879 PMCID: PMC6678391 DOI: 10.3390/ijms20143387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/25/2019] [Accepted: 07/08/2019] [Indexed: 01/11/2023] Open
Abstract
Oviductal epithelial cells (OECs) actively produce stimulating and protecting factors, favoring survival and viability of gametes and early embryos. The oviduct participates in the initial reproductive events, which strongly depends on adhesion. The analysis of differential gene expression in OECs, during long-term in vitro culture, enables recognition of new molecular markers regulating several processes, including "biological adhesion". Porcine oviducts were stained with hematoxylin and eosin, as well as with antibodies against epithelial markers. Then, OECs were long-term in vitro cultured and after 24 h, 7, 15, and 30 days of culture were subjected to transcriptomic and proteomic assays. Microarrays were employed to evaluate gene expression, with Matrix-assisted laser desorption/ionization-time of light (MALDI-TOF) mass spectrometry applied to determine the proteome. The results revealed proper morphology of the oviducts and typical epithelial structure of OECs during the culture. From the set of differentially expressed genes (DEGs), we have selected the 130 that encoded proteins detected by MALDI-TOF MS analysis. From this gene pool, 18 significantly enriched gene ontology biological processes (GO BP) terms were extracted. Among them we focused on genes belonging to "biological adhesion" GO BP. It is suggested that increased expression of studied genes can be attributed to the process of intensive secretion of substances that exhibit favorable influence on oviductal environment, which prime gametes adhesion and viability, fertilization, and early embryo journey.
Collapse
|
18
|
Soe ZY, Prajuabjinda O, Myint PK, Gaowa A, Kawamoto E, Park EJ, Shimaoka M. Talin-2 regulates integrin functions in exosomes. Biochem Biophys Res Commun 2019; 512:429-434. [PMID: 30879762 DOI: 10.1016/j.bbrc.2019.03.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 10/27/2022]
Abstract
Integrins on exosomes have been shown to mediate binding to recipient cells, potentially playing important roles in controlling exosomal internalization and organ distributions. Although the ability of cellular integrins to mediate cell adhesion is known to be regulated by the cytoplasmic adaptor protein talin, whether the activity of exosomal integrins is similarly regulated by talin remains to be elucidated. Here we have studied this question in T-cell exosomes that surface express the integrins αLβ2 and α4β7. T-cells and T-cell exosomes engineered to lack talin-2 showed reduced binding to the integrin ligand ICAM-1 and MAdCAM-1 compared with control T-cells and exosomes, despite the fact that those T cells and exosomes express intact levels of the other isoform talin-1. In addition, talin-2-deficient T-cell exosomes were less efficiently internalized by endothelial cells, compared with control exosomes. These results suggest that the mechanisms of talin-mediated integrin regulation operate similarly in cells and exosomes.
Collapse
Affiliation(s)
- Zay Yar Soe
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Onmanee Prajuabjinda
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Phyoe Kyawe Myint
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Arong Gaowa
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan; Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan.
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
19
|
|
20
|
Zhang SM, Yu LL, Qu T, Hu Y, Yuan DZ, Zhang S, Xu Q, Zhao YB, Zhang JH, Yue LM. The Changes of Cytoskeletal Proteins Induced by the Fast Effect of Estrogen in Mouse Blastocysts and Its Roles in Implantation. Reprod Sci 2017; 24:1639-1646. [PMID: 28299994 DOI: 10.1177/1933719117697126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
It is necessary for estrogen to activate mouse blastocysts, so that they can attach to endometrial epithelium in implantation and in our previous research, we have proved estrogen can induce a fast increase in intracellular calcium of mouse blastocysts through acting on G protein-coupled receptor 30 (GPR30), which further promotes their implantation. Moreover, there has been evidence that cytoskeletal proteins are involved in integrin-mediated adhesion of many kinds of cells, which also plays an important role in implantation. To prove estrogen induces rapidly the changes of cytoskeletal proteins in mouse blastocysts and its roles in implantation, we first used immunofluorescence staining and laser confocal microscopy to investigate the fast effect of estrogen on the expression and localization of cytoskeletal proteins in mouse blastocysts. Second, we used electroporation associated with RNA interference to knock down one of the important cytoskeletal proteins, talin, in the mouse blastocyst cells to investigate the fast effect of estrogen on the localization of integrins and the binding activity of integrins with their ligand fibronectin (FN). At last, mouse blastocysts with different treatments were cultured with FN or uterine epithelial cell line Ishikawa in vitro, respectively, and transferred into the bilateral uterine horns of recipient mice, to study the role of the fast effect of estrogen on cytoskeletal proteins in blastocysts adhesion and implantation. Our results indicated that estradiol (E2), E2 conjugated with bovine serum album (E2-BSA) and G-1 (a GPR30-specific agonist) could induce cytoskeletal protein talin, vinculin, and actin to cluster in the mouse blastocysts, while G15 (a GPR30-specific antagonist) and BAPTA (a calcium chelator) may block this effect induced by E2-BSA. Furthermore, E2-BSA could induce the clustering and relocalization of integrin β1 and β3 and increase the FN-binding activity of integrins in blastocyst cells, while E2-BSA could not induce these effects in the blastocysts pretreated with talin-small interfering RNA (siRNA). Meanwhile, the adhesion rate and implantation rate of blastocysts pretreated with talin-siRNA were significantly lower than those pretreated with control-siRNA. We provided the first evidence that the fast effect of estrogen might cause the clustering of the cytoskeletal proteins in mouse blastocyst cells and further induce the changes of localization and functional activity of integrins in the blastocyst cells, which play important roles in blastocyst implantation.
Collapse
Affiliation(s)
- Shi-Mao Zhang
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- 2 Chengdu Women and Children's Central Hospital, Chengdu, Sichuan, China
| | - Lin-Lin Yu
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- 2 Chengdu Women and Children's Central Hospital, Chengdu, Sichuan, China
| | - Ting Qu
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Ying Hu
- 3 Department of Obstetrics &Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, China
| | - Dong-Zhi Yuan
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Sheng Zhang
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Qian Xu
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - You-Bo Zhao
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jin-Hu Zhang
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Li-Min Yue
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Strohmeyer N, Bharadwaj M, Costell M, Fässler R, Müller DJ. Fibronectin-bound α5β1 integrins sense load and signal to reinforce adhesion in less than a second. NATURE MATERIALS 2017; 16:1262-1270. [PMID: 29115292 DOI: 10.1038/nmat5023] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 10/02/2017] [Indexed: 05/23/2023]
Abstract
Integrin-mediated mechanosensing of the extracellular environment allows cells to control adhesion and signalling. Whether cells sense and respond to force immediately upon ligand-binding is unknown. Here, we report that during adhesion initiation, fibroblasts respond to mechanical load by strengthening integrin-mediated adhesion to fibronectin (FN) in a biphasic manner. In the first phase, which depends on talin and kindlin as well as on the actin nucleators Arp2/3 and mDia, FN-engaged α5β1 integrins activate focal adhesion kinase (FAK) and c-Src in less than 0.5 s to steeply strengthen α5β1- and αV-class integrin-mediated adhesion. When the mechanical load exceeds a certain threshold, fibroblasts decrease adhesion and initiate the second phase, which is characterized by less steep adhesion strengthening. This unique, biphasic cellular adhesion response is mediated by α5β1 integrins, which form catch bonds with FN and signal to FN-binding integrins to reinforce cell adhesion much before visible adhesion clusters are formed.
Collapse
Affiliation(s)
- Nico Strohmeyer
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| | - Mitasha Bharadwaj
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| | - Mercedes Costell
- Department of Biochemistry and Molecular Biology, Estructura de Reserca Interdisciplinar en Biotechnologia i Biomedicina, Universitat de València, 46100 Burjassot, Spain
| | - Reinhard Fässler
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, 82152 Martinsried, Germany
| | - Daniel J Müller
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| |
Collapse
|
22
|
Jiao Y, Tan S, Xiong J. Proteomic changes of CD4 +/CD25 +/forkhead box p3 + regulatory T cells in a 30-day rat model of sepsis survival. Exp Ther Med 2017; 14:5619-5628. [PMID: 29285101 DOI: 10.3892/etm.2017.5233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/20/2017] [Indexed: 02/07/2023] Open
Abstract
Sepsis is defined as life threatening organ dysfunction arising from a dysregulated host response to infection. The outcomes of sepsis include early mortality, delayed mortality and recovery, and depend on the inflammatory response. Previous studies have demonstrated that regulatory T cells (Tregs) are important in determining the outcome of sepsis, as their suppressive function serves a role in maintaining immune homeostasis. However, Treg-mediated immunosuppression during the course of sepsis remains unclear and little is known about the survival of patients following diagnosis. Studying the survivors of sepsis may explain the mechanisms of natural recovery. Therefore, a 30-day rat model of sepsis survival was established in the current study. Cluster of differentiation CD4+/CD25+/forkhead box p3+ Tregs were isolated from the blood and spleens of rats undergoing cecal ligation and puncture or sham surgery, using flow cytometry. Proteomic analysis was performed using nano high-performance liquid chromatography-mass spectrometry. Several different biological pathways associated with uncommon differentially-expressed proteins were identified in the blood and spleen survivor and sham groups. Extracellular-regulated kinase/mitogen-activated protein kinase, as well as integrin and actin cytoskeletal pathway elements, including Ras-related protein 1b, talin 1 and filamin A, were associated with Tregs in the blood. Pathway elements associated with cell cycle regulators in the B-cell translocation gene family of proteins, tumor necrosis factor receptor superfamily member 4, Hippo signaling, P70-S6 kinase 1, phosphatidylinositol 3-kinase/protein kinase B signaling and 1,25-dihydroxyvitamin D3 biosynthesis were associated with Tregs from the spleen including phosphatase 2A activator regulatory factor 4, histone arginine methyltransferase, CD4, major histocompatibility complex class I antigens, 14-3-3 protein θ and nicotinamide adenine dinucleotide phosphate cytochrome P450 reductase. These results explain the mechanism by which Tregs naturally recover and indicates that Tregs in the blood and spleen vary. Differentially-expressed proteins serving a role in these pathways provide additional insight for the identification of new targets for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Yuxia Jiao
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Siqi Tan
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Junyu Xiong
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
23
|
Ruisu K, Meier R, Kask K, Tõnissoo T, Velling T, Pooga M. RIC8A is essential for the organisation of actin cytoskeleton and cell-matrix interaction. Exp Cell Res 2017; 357:181-191. [PMID: 28526238 DOI: 10.1016/j.yexcr.2017.05.012] [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: 01/04/2017] [Revised: 05/09/2017] [Accepted: 05/14/2017] [Indexed: 01/07/2023]
Abstract
RIC8A functions as a chaperone and guanine nucleotide exchange factor for a subset of G protein α subunits. Multiple G protein subunits mediate various signalling events that regulate cell adhesion and migration and the involvement of RIC8A in some of these processes has been demonstrated. We have previously shown that the deficiency of RIC8A causes a failure in mouse gastrulation and neurogenesis - major events in embryogenesis that rely on proper association of cells with the extracellular matrix (ECM) and involve active cell migration. To elaborate on these findings, we used Ric8a-/- mouse embryonic stem cells and Ric8a-deficient mouse embryonic fibroblasts, and found that RIC8A plays an important role in the organisation and remodelling of actin cytoskeleton and cell-ECM association. Ric8a-deficient cells were able to attach to different ECM components, but were unable to spread correctly, and did not form stress fibres or focal adhesion complexes. We also found that the presence of RIC8A is necessary for the activation of β1 integrins and integrin-mediated cell migration.
Collapse
Affiliation(s)
- Katrin Ruisu
- Department of Developmental Biology, Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St., Tartu 51010, Estonia.
| | - Riho Meier
- Department of Developmental Biology, Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St., Tartu 51010, Estonia; Competence Centre on Health Technologies, Tiigi 61b, 50410 Tartu, Estonia
| | - Keiu Kask
- Department of Developmental Biology, Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St., Tartu 51010, Estonia
| | - Tambet Tõnissoo
- Department of Developmental Biology, Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St., Tartu 51010, Estonia; Competence Centre on Health Technologies, Tiigi 61b, 50410 Tartu, Estonia
| | - Teet Velling
- Department of Developmental Biology, Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St., Tartu 51010, Estonia; Competence Centre on Health Technologies, Tiigi 61b, 50410 Tartu, Estonia
| | - Margus Pooga
- Department of Developmental Biology, Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St., Tartu 51010, Estonia; Competence Centre on Health Technologies, Tiigi 61b, 50410 Tartu, Estonia
| |
Collapse
|
24
|
Li Y, Luo X, Sun Y, Cui Z, Liu Y, Liu R, Guo X. High Stoichiometry Phosphorylation of Talin at T144/T150 or S446 Produces Contrasting Effects on Calpain-mediated Talin Cleavage and Cell Migration. J Cancer 2016; 7:1645-1652. [PMID: 27698901 PMCID: PMC5039385 DOI: 10.7150/jca.14192] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/13/2016] [Indexed: 12/20/2022] Open
Abstract
Focal adhesions are large multi-protein complexes that serve as the linkage between extracellular matrix (ECM) and actin cytoskeleton and control the network of signaling cascades underlying cell migration. Talin plays a key role in focal adhesion turnover, and calpain-mediated proteolysis of talin is central to focal adhesion disassembly, but its regulation is not well elucidated. Here we demonstrate that talin phosphorylation at three high stoichiometry sites on its head domain, T144 and T150, or S446, have contrasting effects on calpain-mediated cleavage of talin and cell migration by using site-directed mutagenesis to inhibit phosphorylation. Expression of talinT144A+T150A stimulated calpain-mediated cleavage of talin and accelerated focal adhesion disassembly, whereas expression of talinS446A fully inhibited talin cleavage by calpain, preventing focal adhesion disassembly. A large decrease in phospho-threonine or phospho-serine levels was seen with talinT144A+T150A or talinS446A respectively, while more active ERK was present in talinT144A+T150A than in talinS446A. Cell adhesion and transwell assays using uniformly expressing cells showed that expression of talinT144A+T150A or talinS446A have opposing effects on cell adhesion and migration. These findings define and highlight the integral role of site-specific high stoichiometry phosphorylation of talin in regulating calpain-mediated cleavage of talin and focal adhesion disassembly, thus controlling adhesion stability, cell adhesion and ultimately, cell migration.
Collapse
Affiliation(s)
- Youjun Li
- Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Xiaoyong Luo
- Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yang Sun
- Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Zhenyi Cui
- Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yizhou Liu
- Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Rushi Liu
- Laboratory of Medical Molecular and Immunological Diagnostics, College of Medicine, Hunan Normal University, Changsha 410013, China
| | - Xiangrong Guo
- Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
25
|
Kumar A, Ouyang M, Van den Dries K, McGhee EJ, Tanaka K, Anderson MD, Groisman A, Goult BT, Anderson KI, Schwartz MA. Talin tension sensor reveals novel features of focal adhesion force transmission and mechanosensitivity. J Cell Biol 2016; 213:371-83. [PMID: 27161398 PMCID: PMC4862330 DOI: 10.1083/jcb.201510012] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 04/05/2016] [Indexed: 12/12/2022] Open
Abstract
Integrin-dependent adhesions are mechanosensitive structures in which talin mediates a linkage to actin filaments either directly or indirectly by recruiting vinculin. Here, we report the development and validation of a talin tension sensor. We find that talin in focal adhesions is under tension, which is higher in peripheral than central adhesions. Tension on talin is increased by vinculin and depends mainly on actin-binding site 2 (ABS2) within the middle of the rod domain, rather than ABS3 at the far C terminus. Unlike vinculin, talin is under lower tension on soft substrates. The difference between central and peripheral adhesions requires ABS3 but not vinculin or ABS2. However, differential stiffness sensing by talin requires ABS2 but not vinculin or ABS3. These results indicate that central versus peripheral adhesions must be organized and regulated differently, and that ABS2 and ABS3 have distinct functions in spatial variations and stiffness sensing. Overall, these results shed new light on talin function and constrain models for cellular mechanosensing.
Collapse
Affiliation(s)
- Abhishek Kumar
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511
| | - Mingxing Ouyang
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511
| | - Koen Van den Dries
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511
| | - Ewan James McGhee
- Beatson Institute for Cancer Research, Glasgow G20 0TZ, Scotland, UK
| | - Keiichiro Tanaka
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511
| | - Marie D Anderson
- School of Biosciences, University of Kent, Canterbury CT2 7NZ, England, UK
| | - Alexander Groisman
- Department of Physics, University of California, San Diego, La Jolla, CA 92093
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury CT2 7NZ, England, UK
| | - Kurt I Anderson
- Beatson Institute for Cancer Research, Glasgow G20 0TZ, Scotland, UK
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06511 Department of Cell Biology, Yale University, New Haven, CT 06520 Department of Biomedical Engineering, Yale University, New Haven, CT 06520
| |
Collapse
|
26
|
Yao F, Kausalya JP, Sia YY, Teo ASM, Lee WH, Ong AGM, Zhang Z, Tan JHJ, Li G, Bertrand D, Liu X, Poh HM, Guan P, Zhu F, Pathiraja TN, Ariyaratne PN, Rao J, Woo XY, Cai S, Mulawadi FH, Poh WT, Veeravalli L, Chan CS, Lim SS, Leong ST, Neo SC, Choi PSD, Chew EGY, Nagarajan N, Jacques PÉ, So JBY, Ruan X, Yeoh KG, Tan P, Sung WK, Hunziker W, Ruan Y, Hillmer AM. Recurrent Fusion Genes in Gastric Cancer: CLDN18-ARHGAP26 Induces Loss of Epithelial Integrity. Cell Rep 2015; 12:272-85. [PMID: 26146084 DOI: 10.1016/j.celrep.2015.06.020] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 04/21/2015] [Accepted: 06/06/2015] [Indexed: 12/21/2022] Open
Abstract
Genome rearrangements, a hallmark of cancer, can result in gene fusions with oncogenic properties. Using DNA paired-end-tag (DNA-PET) whole-genome sequencing, we analyzed 15 gastric cancers (GCs) from Southeast Asians. Rearrangements were enriched in open chromatin and shaped by chromatin structure. We identified seven rearrangement hot spots and 136 gene fusions. In three out of 100 GC cases, we found recurrent fusions between CLDN18, a tight junction gene, and ARHGAP26, a gene encoding a RHOA inhibitor. Epithelial cell lines expressing CLDN18-ARHGAP26 displayed a dramatic loss of epithelial phenotype and long protrusions indicative of epithelial-mesenchymal transition (EMT). Fusion-positive cell lines showed impaired barrier properties, reduced cell-cell and cell-extracellular matrix adhesion, retarded wound healing, and inhibition of RHOA. Gain of invasion was seen in cancer cell lines expressing the fusion. Thus, CLDN18-ARHGAP26 mediates epithelial disintegration, possibly leading to stomach H(+) leakage, and the fusion might contribute to invasiveness once a cell is transformed.
Collapse
Affiliation(s)
- Fei Yao
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore; The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore
| | - Jaya P Kausalya
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, Singapore 138673, Singapore
| | - Yee Yen Sia
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore; The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore
| | - Audrey S M Teo
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore; The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore
| | - Wah Heng Lee
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Alicia G M Ong
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, Singapore 138673, Singapore
| | - Zhenshui Zhang
- Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Joanna H J Tan
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Guoliang Li
- National Key Laboratory of Crop Genetic Improvement, Center for Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Denis Bertrand
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Xingliang Liu
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Huay Mei Poh
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Peiyong Guan
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore; School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Feng Zhu
- The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Thushangi Nadeera Pathiraja
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore; The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore
| | - Pramila N Ariyaratne
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Jaideepraj Rao
- Department of General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Xing Yi Woo
- Personal Genomic Solutions, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Shaojiang Cai
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Fabianus H Mulawadi
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Wan Ting Poh
- Personal Genomic Solutions, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Lavanya Veeravalli
- Research Computing, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Chee Seng Chan
- Research Computing, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Seong Soo Lim
- Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - See Ting Leong
- Genome Technology and Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Say Chuan Neo
- Genome Technology and Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Poh Sum D Choi
- Genome Technology and Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Elaine G Y Chew
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Niranjan Nagarajan
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | | | - Jimmy B Y So
- The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; National University Health System, Singapore 119228, Singapore
| | - Xiaoan Ruan
- Personal Genomic Solutions, Genome Institute of Singapore, Singapore 138672, Singapore; Genome Technology and Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Khay Guan Yeoh
- The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; National University Health System, Singapore 119228, Singapore
| | - Patrick Tan
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore; The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore; Duke-NUS Graduate Medical School, Singapore 169857, Singapore; Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Wing-Kin Sung
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore; School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Walter Hunziker
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, Singapore 138673, Singapore; Department of Physiology, National University of Singapore, Singapore 117597, Singapore.
| | - Yijun Ruan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.
| | - Axel M Hillmer
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore; The Singapore Gastric Cancer Consortium, National University of Singapore, Singapore 119228, Singapore.
| |
Collapse
|
27
|
Kiss A, Gong X, Kowalewski JM, Shafqat-Abbasi H, Strömblad S, Lock JG. Non-monotonic cellular responses to heterogeneity in talin protein expression-level. Integr Biol (Camb) 2015; 7:1171-85. [PMID: 26000342 DOI: 10.1039/c4ib00291a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Talin is a key cell-matrix adhesion component with a central role in regulating adhesion complex maturation, and thereby various cellular properties including adhesion and migration. However, knockdown studies have produced inconsistent findings regarding the functional influence of talin in these processes. Such discrepancies may reflect non-monotonic responses to talin expression-level variation that are not detectable via canonical "binary" comparisons of aggregated control versus knockdown cell populations. Here, we deployed an "analogue" approach to map talin influence across a continuous expression-level spectrum, which we extended with sub-maximal RNAi-mediated talin depletion. Applying correlative imaging to link live cell and fixed immunofluorescence data on a single cell basis, we related per cell talin levels to per cell measures quantitatively defining an array of cellular properties. This revealed both linear and non-linear correspondences between talin expression and cellular properties, including non-monotonic influences over cell shape, adhesion complex-F-actin association and adhesion localization. Furthermore, we demonstrate talin level-dependent changes in networks of correlations among adhesion/migration properties, particularly in relation to cell migration speed. Importantly, these correlation networks were strongly affected by talin expression heterogeneity within the natural range, implying that this endogenous variation has a broad, quantitatively detectable influence. Overall, we present an accessible analogue method that reveals complex dependencies on talin expression-level, thereby establishing a framework for considering non-linear and non-monotonic effects of protein expression-level heterogeneity in cellular systems.
Collapse
Affiliation(s)
- Alexa Kiss
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Hälsov. 7-9, G-building floor 6, S-141 83 Huddinge, Sweden.
| | | | | | | | | | | |
Collapse
|
28
|
Ye TM, Pang RT, Leung CO, Chiu JF, Yeung WS. Two-dimensional liquid chromatography with tandem mass spectrometry–based proteomic characterization of endometrial luminal epithelial surface proteins responsible for embryo implantation. Fertil Steril 2015; 103:853-61.e3. [DOI: 10.1016/j.fertnstert.2014.12.110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 12/14/2014] [Accepted: 12/16/2014] [Indexed: 10/24/2022]
|
29
|
Nanda SY, Hoang T, Patel P, Zhang H. Vinculin regulates assembly of talin: β3 integrin complexes. J Cell Biochem 2014; 115:1206-16. [PMID: 24446374 DOI: 10.1002/jcb.24772] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/17/2014] [Indexed: 11/09/2022]
Abstract
Vinculin is a talin-binding protein that promotes integrin-mediated cell adhesion, but the mechanisms are not understood. Because talin is a direct activator of integrins, we asked whether and how vinculin regulates the formation of integrin: talin complexes. We report that VD1 (aa 1-258) and its talin-binding mutant, VD1A50I, bind directly and equally to several β integrin cytoplasmic tails (βCT). Results from competition assays show that VD1, but not VD1A50I, inhibits the interaction of talin (Tn) and talin rod (TnR), but not talin head (TnH) with β3CT. The inhibition observed could be the result of VD1 binding to one or more of the 11 vinculin binding sites (VBSs) in the TnR domain. Our studies demonstrate that VD1 binding to amino acids 482-911, a VBS rich region, in TnR perturbs the interaction of rod with β3CT. The integrin activation assays done using CHOA5 cells show that activated vinculin enhances αIIbβ3 integrin activation and that the effect is dependent on talin. The TnR domain however shows no integrin activation unlike TnH that shows enhanced integrin activation. The overall results indicate that activated vinculin promotes talin-mediated integrin activation by binding to accessible VBSs in TnR and thus displacing the TnR from the β3 subunit. The study presented, defines a novel direct interaction of VD1 with β3CT and provides an attractive explanation for vinculin's ability to potentiate integrin-mediated cell adhesion through directly binding to both TnR and the integrin cytoplasmic tail.
Collapse
Affiliation(s)
- Suman Yadav Nanda
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | | | | | | |
Collapse
|
30
|
Zhang X, Moore SW, Iskratsch T, Sheetz MP. N-WASP-directed actin polymerization activates Cas phosphorylation and lamellipodium spreading. J Cell Sci 2014; 127:1394-405. [PMID: 24481817 DOI: 10.1242/jcs.134692] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Tyrosine phosphorylation of the substrate domain of Cas (CasSD) correlates with increased cell migration in healthy and diseased cells. Here, we address the mechanism leading to the phosphorylation of CasSD in the context of fibronectin-induced early spreading of fibroblasts. We have previously demonstrated that mechanical stretching of CasSD exposes phosphorylation sites for Src family kinases (SFKs). Surprisingly, phosphorylation of CasSD was independent of myosin contractile activity but dependent on actin polymerization. Furthermore, we found that CasSD phosphorylation in the early stages of cell spreading required: (1) integrin anchorage and integrin-mediated activation of SFKs, (2) association of Cas with focal adhesion kinase (FAK), and (3) N-WASP-driven actin-assembly activity. These findings, and analyses of the interactions of the Cas domains, indicate that the N-terminus of Cas associates with the FAK-N-WASP complex at the protrusive edge of the cell and that the C-terminus of Cas associates with the immobilized integrin-SFK cluster. Thus, extension of the leading edge mediated by actin polymerization could stretch Cas during early cell spreading, priming it for phosphorylation.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY 10027, USA
| | | | | | | |
Collapse
|
31
|
Calderwood DA, Campbell ID, Critchley DR. Talins and kindlins: partners in integrin-mediated adhesion. Nat Rev Mol Cell Biol 2013; 14:503-17. [PMID: 23860236 PMCID: PMC4116690 DOI: 10.1038/nrm3624] [Citation(s) in RCA: 457] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Integrin receptors provide a dynamic, tightly-regulated link between the extracellular matrix (or cellular counter-receptors) and intracellular cytoskeletal and signalling networks, enabling cells to sense and respond to their chemical and physical environment. Talins and kindlins, two families of FERM-domain proteins, bind the cytoplasmic tail of integrins, recruit cytoskeletal and signalling proteins involved in mechanotransduction and synergize to activate integrin binding to extracellular ligands. New data reveal the domain structure of full-length talin, provide insights into talin-mediated integrin activation and show that RIAM recruits talin to the plasma membrane, whereas vinculin stabilizes talin in cell-matrix junctions. How kindlins act is less well-defined, but disease-causing mutations show that kindlins are also essential for integrin activation, adhesion, cell spreading and signalling.
Collapse
Affiliation(s)
- David A Calderwood
- Departments of Pharmacology and of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Iain D Campbell
- Department of Biochemistry, University of Oxford, S. Parks Rd., Oxford, OX1 3QU, UK
| | - David R Critchley
- Department of Biochemistry, University of Leicester, Leicester LE1 7RH
| |
Collapse
|
32
|
Lim J, Thompson J, May RC, Hotchin NA, Caron E. Regulator of G-Protein Signalling-14 (RGS14) Regulates the Activation of αMβ2 Integrin during Phagocytosis. PLoS One 2013; 8:e69163. [PMID: 23805333 PMCID: PMC3689692 DOI: 10.1371/journal.pone.0069163] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 06/10/2013] [Indexed: 01/30/2023] Open
Abstract
Integrin-mediated phagocytosis, an important physiological activity undertaken by professional phagocytes, requires bidirectional signalling to/from αMβ2 integrin and involves Rap1 and Rho GTPases. The action of Rap1 and the cytoskeletal protein talin in activating αMβ2 integrins, in a RIAM-independent manner, has been previously shown to be critical during phagocytosis in mammalian phagocytes. However, the events downstream of Rap1 are not clearly understood. Our data demonstrate that one potential Rap1 effector, Regulator of G-Protein Signalling-14 (RGS14), is involved in activating αMβ2. Exogenous expression of RGS14 in COS-7 cells expressing αMβ2 results in increased binding of C3bi-opsonised sheep red blood cells. Consistent with this, knock-down of RGS14 in J774.A1 macrophages results in decreased association with C3bi-opsonised sheep red blood cells. Regulation of αMβ2 function occurs through the R333 residue of the RGS14 Ras/Rap binding domain (RBD) and the F754 residue of β2, residues previously shown to be involved in binding of H-Ras and talin1 head binding prior to αMβ2 activation, respectively. Surprisingly, overexpression of talin2 or RAPL had no effect on αMβ2 regulation. Our results establish for the first time a role for RGS14 in the mechanism of Rap1/talin1 activation of αMβ2 during phagocytosis.
Collapse
Affiliation(s)
- Jenson Lim
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- nanoTherics Ltd., Newcastle under Lyme, Staffordshire, United Kingdom
| | - Jo Thompson
- Royal Devon and Exeter Hospital, Exeter, Devon, United Kingdom
| | - Robin C. May
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Neil A. Hotchin
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Emmanuelle Caron
- Centre for Molecular Microbiology and Infection and Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
33
|
Jevnikar Z, Rojnik M, Jamnik P, Doljak B, Fonovic UP, Kos J. Cathepsin H mediates the processing of talin and regulates migration of prostate cancer cells. J Biol Chem 2013; 288:2201-9. [PMID: 23204516 PMCID: PMC3554893 DOI: 10.1074/jbc.m112.436394] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Indexed: 12/18/2022] Open
Abstract
The cytoskeletal protein talin, an actin- and β-integrin tail-binding protein, plays an important role in cell migration by promoting integrin activation and focal adhesion formation. Here, we show that talin is a substrate for cathepsin H (CtsH), a lysosomal cysteine protease with a strong aminopeptidase activity. Purified active CtsH sequentially cleaved a synthetic peptide representing the N terminus of the talin F0 head domain. The processing of talin by CtsH was determined also in the metastatic PC-3 prostate cancer cell line, which exhibits increased expression of CtsH. The attenuation of CtsH aminopeptidase activity by a specific inhibitor or siRNA-mediated silencing significantly reduced the migration of PC-3 cells on fibronectin and invasion through Matrigel. We found that in migrating PC-3 cells, CtsH was co-localized with talin in the focal adhesions. Furthermore, specific inhibition of CtsH increased the activation of α(v)β(3)-integrin on PC-3 cells. We propose that CtsH-mediated processing of talin might promote cancer cell progression by affecting integrin activation and adhesion strength.
Collapse
Affiliation(s)
- Zala Jevnikar
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia.
| | | | | | | | | | | |
Collapse
|
34
|
Talin couples the actomyosin cortex to the plasma membrane during rear retraction and cytokinesis. Proc Natl Acad Sci U S A 2012; 109:12992-7. [PMID: 22826231 DOI: 10.1073/pnas.1208296109] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Contraction of the cortical actin cytoskeleton underlies both rear retraction in directed cell migration and cytokinesis. Here, we show that talin, a central component of focal adhesions, has a major role in these processes. We found that Dictyostelium talin A colocalized with myosin II in the rear of migrating cells and the cleavage furrow. During directed cell migration, talin A-null cells displayed a long thin tail devoid of actin filaments, whereas additional depletion of SibA, a transmembrane adhesion molecule that binds to talin A, reverted this phenotype, suggesting a requirement of the link between actomyosin and SibA by talin A for rear retraction. Disruptions of talin A also resulted in detachment of the actomyosin contractile ring from the cell membrane and concomitant regression of the cleavage furrow under certain conditions. The C-terminal actin-binding domain (ABD) of talin A exhibited a localization pattern identical to that of full-length talin A. The N-terminal FERM domain was found to bind phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] and phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3,4,5)P3] in vitro. In vivo, however, PtdIns(4,5)P2, which is known to activate talin, is believed to be enriched in the rear of migrating cells and the cleavage furrow in Dictyostelium. From these results, we propose that talin A activated by PtdIns(4,5)P2 in the cell posterior or cleavage furrow links actomyosin cytoskeleton to adhesion molecules or other membrane proteins, and that the force is transmitted through these links to retract the tail during cell migration or to cause efficient ingression of the equator during cytokinesis.
Collapse
|
35
|
Le XF, Almeida MI, Mao W, Spizzo R, Rossi S, Nicoloso MS, Zhang S, Wu Y, Calin GA, Bast RC. Modulation of MicroRNA-194 and cell migration by HER2-targeting trastuzumab in breast cancer. PLoS One 2012; 7:e41170. [PMID: 22829924 PMCID: PMC3400637 DOI: 10.1371/journal.pone.0041170] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/18/2012] [Indexed: 12/27/2022] Open
Abstract
Trastuzumab, a humanized monoclonal antibody directed against the extracellular domain of the HER2 oncoprotein, can effectively target HER2-positive breast cancer through several mechanisms. Although the effects of trastuzumab on cancer cell proliferation, angiogenesis and apoptosis have been investigated in depth, the effect of trastuzumab on microRNA (miRNA) has not been extensively studied. We have performed miRNA microarray profiling before and after trastuzumab treatment in SKBr3 and BT474 human breast cancer cells that overexpress HER2. We found that trastuzumab treatment of SKBr3 cells significantly decreased five miRNAs and increased three others, whereas treatment of BT474 cells significantly decreased two miRNAs and increased nine. The only change in miRNA expression observed in both cell lines following trastuzumab treatment was upregulation of miRNA-194 (miR-194) that was further validated in vitro and in vivo. Forced expression of miR-194 in breast cancer cells that overexpress HER2 produced no effect on apoptosis, modest inhibition of proliferation, significant inhibition of cell migration/invasion in vitro and significant inhibition of xenograft growth in vivo. Conversely, knockdown of miR-194 promoted cell migration. Increased miR-194 expression markedly reduced levels of the cytoskeletal protein talin2 and specifically inhibited luciferase reporter activity of a talin2 wild-type 3'-untranslated region, but not that of a mutant reporter, indicating that talin2 is a direct downstream target of miR-194. Trastuzumab treatment inhibited breast cancer cell migration and reduced talin2 expression in vitro and in vivo. Knockdown of talin2 inhibited cell migration/invasion. Knockdown of trastuzumab-induced miR-194 expression with a miR-194 inhibitor compromised trastuzumab-inhibited cell migration in HER2-overexpressing breast cancer cells. Consequently, trastuzumab treatment upregulates miR-194 expression and may exert its cell migration-inhibitory effect through miR-194-mediated downregulation of cytoskeleton protein talin2 in HER2-overexpressing human breast cancer cells.
Collapse
Affiliation(s)
- Xiao-Feng Le
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (XFL); (RCB)
| | - Maria I. Almeida
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Braga, Portugal
| | - Weiqun Mao
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Riccardo Spizzo
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Simona Rossi
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Milena S. Nicoloso
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Shu Zhang
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yun Wu
- Department of Pathology, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - George A. Calin
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Robert C. Bast
- Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail: (XFL); (RCB)
| |
Collapse
|
36
|
Bate N, Gingras AR, Bachir A, Horwitz R, Ye F, Patel B, Goult BT, Critchley DR. Talin contains a C-terminal calpain2 cleavage site important in focal adhesion dynamics. PLoS One 2012; 7:e34461. [PMID: 22496808 PMCID: PMC3319578 DOI: 10.1371/journal.pone.0034461] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 02/15/2012] [Indexed: 12/31/2022] Open
Abstract
Talin is a large (∼2540 residues) dimeric adaptor protein that associates with the integrin family of cell adhesion molecules in cell-extracellular matrix junctions (focal adhesions; FAs), where it both activates integrins and couples them to the actin cytoskeleton. Calpain2-mediated cleavage of talin between the head and rod domains has previously been shown to be important in FA turnover. Here we identify an additional calpain2-cleavage site that removes the dimerisation domain from the C-terminus of the talin rod, and show that an E2492G mutation inhibits calpain cleavage at this site in vitro, and increases the steady state levels of talin1 in vivo. Expression of a GFP-tagged talin1 E2492G mutant in CHO.K1 cells inhibited FA turnover and the persistence of cell protrusion just as effectively as a L432G mutation that inhibits calpain cleavage between the talin head and rod domains. Moreover, incorporation of both mutations into a single talin molecule had an additive effect clearly demonstrating that calpain cleavage at both the N- and C-terminal regions of talin contribute to the regulation of FA dynamics. However, the N-terminal site was more sensitive to calpain cleavage suggesting that lower levels of calpain are required to liberate the talin head and rod fragments than are needed to clip off the C-terminal dimerisation domain. The talin head and rod liberated by calpain2 cleavage have recently been shown to play roles in an integrin activation cycle important in FA turnover and in FAK-dependent cell cycle progression respectively. The half-life of the talin head is tightly regulated by ubiquitination and we suggest that removal of the C-terminal dimerisation domain from the talin rod may provide a mechanism both for terminating the signalling function of the talin rod and indeed for inactivating full-length talin thereby promoting FA turnover at the rear of the cell.
Collapse
Affiliation(s)
- Neil Bate
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | | | - Alexia Bachir
- Department of Cell Biology, Univeristy of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Rick Horwitz
- Department of Cell Biology, Univeristy of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Feng Ye
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Bipin Patel
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | - Benjamin T. Goult
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | - David R. Critchley
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
- * E-mail:
| |
Collapse
|
37
|
Wang P, Ballestrem C, Streuli CH. The C terminus of talin links integrins to cell cycle progression. ACTA ACUST UNITED AC 2012; 195:499-513. [PMID: 22042621 PMCID: PMC3206343 DOI: 10.1083/jcb.201104128] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Talin recruits and activates focal adhesion proteins required for cell cycle progression. Integrins are cell adhesion receptors that sense the extracellular matrix (ECM) environment. One of their functions is to regulate cell fate decisions, although the question of how integrins initiate intracellular signaling is not fully resolved. In this paper, we examine the role of talin, an adapter protein at cell–matrix attachment sites, in outside-in signaling. We used lentiviral small hairpin ribonucleic acid to deplete talin in mammary epithelial cells. These cells still attached to the ECM in an integrin-dependent manner and spread. They had a normal actin cytoskeleton, but vinculin, paxillin, focal adhesion kinase (FAK), and integrin-linked kinase were not recruited to adhesion sites. Talin-deficient cells showed proliferation defects, and reexpressing a tail portion of the talin rod, but not its head domain, restored integrin-mediated FAK phosphorylation, suppressed p21 expression, and rescued cell cycle. Thus, talin recruits and activates focal adhesion proteins required for proliferation via the C terminus of its rod domain. Our study reveals a new function for talin, which is to link integrin adhesions with cell cycle progression.
Collapse
Affiliation(s)
- Pengbo Wang
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, England, UK
| | | | | |
Collapse
|
38
|
Sen S, Ng WP, Kumar S. Contributions of talin-1 to glioma cell-matrix tensional homeostasis. J R Soc Interface 2011; 9:1311-7. [PMID: 22158841 DOI: 10.1098/rsif.2011.0567] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The ability of cells to adapt their mechanical properties to those of the surrounding microenvironment (tensional homeostasis) has been implicated in the progression of a variety of solid tumours, including the brain tumour glioblastoma multiforme (GBM). GBM tumour cells are highly sensitive to extracellular matrix (ECM) stiffness and overexpress a variety of focal adhesion proteins, such as talin. While talin has been shown to play critical early roles in integrin-based force-sensing in non-tumour cells, it remains unclear whether this protein contributes to tensional homeostasis in GBM cells. Here, we investigate the role of the talin isoform talin-1 in enabling human GBM cells to adapt to ECM stiffness. We show that human GBM cells express talin-1, and we use RNA interference to suppress talin-1 expression without affecting levels of talin-2, vinculin or phosphorylated focal adhesion kinase. Knockdown of talin-1 strongly reduces both cell spreading area and random migration speed but does not significantly affect overall focal adhesion size distributions. Most strikingly, atomic force microscopy indentation reveals that talin-1 suppression compromises adaptation of cell stiffness to changes in ECM stiffness. Together, these data support a role for talin-1 in the maintenance of tensional homeostasis in GBM and suggest a functional role for enriched talin expression in this tumour.
Collapse
Affiliation(s)
- Shamik Sen
- Department of Bioengineering, University of California, 274A Stanley Hall no. 1762, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
39
|
Sanz-Herrera JA, Reina-Romo E. Cell-biomaterial mechanical interaction in the framework of tissue engineering: insights, computational modeling and perspectives. Int J Mol Sci 2011; 12:8217-44. [PMID: 22174660 PMCID: PMC3233466 DOI: 10.3390/ijms12118217] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Revised: 10/19/2011] [Accepted: 11/02/2011] [Indexed: 01/09/2023] Open
Abstract
Tissue engineering is an emerging field of research which combines the use of cell-seeded biomaterials both in vitro and/or in vivo with the aim of promoting new tissue formation or regeneration. In this context, how cells colonize and interact with the biomaterial is critical in order to get a functional tissue engineering product. Cell-biomaterial interaction is referred to here as the phenomenon involved in adherent cells attachment to the biomaterial surface, and their related cell functions such as growth, differentiation, migration or apoptosis. This process is inherently complex in nature involving many physico-chemical events which take place at different scales ranging from molecular to cell body (organelle) levels. Moreover, it has been demonstrated that the mechanical environment at the cell-biomaterial location may play an important role in the subsequent cell function, which remains to be elucidated. In this paper, the state-of-the-art research in the physics and mechanics of cell-biomaterial interaction is reviewed with an emphasis on focal adhesions. The paper is focused on the different models developed at different scales available to simulate certain features of cell-biomaterial interaction. A proper understanding of cell-biomaterial interaction, as well as the development of predictive models in this sense, may add some light in tissue engineering and regenerative medicine fields.
Collapse
Affiliation(s)
- Jose A. Sanz-Herrera
- School of Engineering, University of Seville, Camino de los descubrimientos s/n, 41092 Seville, Spain; E-Mail:
| | - Esther Reina-Romo
- School of Engineering, University of Seville, Camino de los descubrimientos s/n, 41092 Seville, Spain; E-Mail:
| |
Collapse
|
40
|
PIPKIγ regulates focal adhesion dynamics and colon cancer cell invasion. PLoS One 2011; 6:e24775. [PMID: 21931851 PMCID: PMC3171478 DOI: 10.1371/journal.pone.0024775] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 08/17/2011] [Indexed: 11/19/2022] Open
Abstract
Focal adhesion assembly and disassembly are essential for cell migration and cancer invasion, but the detailed molecular mechanisms regulating these processes remain to be elucidated. Phosphatidylinositol phosphate kinase type Iγ (PIPKIγ) binds talin and is required for focal adhesion formation in EGF-stimulated cells, but its role in regulating focal adhesion dynamics and cancer invasion is poorly understood. We show here that overexpression of PIPKIγ promoted focal adhesion formation, whereas cells expressing either PIPKIγ(K188,200R) or PIPKIγ(D316K), two kinase-dead mutants, had much fewer focal adhesions than those expressing WT PIPKIγ in CHO-K1 cells and HCT116 colon cancer cells. Furthermore, overexpression of PIPKIγ, but not PIPKIγ(K188,200R), resulted in an increase in both focal adhesion assembly and disassembly rates. Depletion of PIPKIγ by using shRNA strongly inhibited formation of focal adhesions in HCT116 cells. Overexpression of PIPKIγ(K188,200R) or depletion of PIPKIγ reduced the strength of HCT116 cell adhesion to fibronection and inhibited the invasive capacities of HCT116 cells. PIPKIγ depletion reduced PIP₂ levels to ∼40% of control and PIP₃ to undetectable levels, and inhibited vinculin localizing to focal adhesions. Taken together, PIPKIγ positively regulates focal adhesion dynamics and cancer invasion, most probably through PIP₂-mediated vinculin activation.
Collapse
|
41
|
Liu J, He X, Qi Y, Tian X, Monkley SJ, Critchley DR, Corbett SA, Lowry SF, Graham AM, Li S. Talin1 regulates integrin turnover to promote embryonic epithelial morphogenesis. Mol Cell Biol 2011; 31:3366-77. [PMID: 21670148 PMCID: PMC3147785 DOI: 10.1128/mcb.01403-10] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 05/21/2011] [Indexed: 12/17/2022] Open
Abstract
Talin is a cytoskeletal protein that binds to integrin β cytoplasmic tails and regulates integrin activation. Talin1 ablation in mice disrupts gastrulation and causes embryonic lethality. However, the role of talin in mammalian epithelial morphogenesis is poorly understood. Here we demonstrate that embryoid bodies (EBs) differentiated from talin1-null embryonic stem cells are defective in integrin adhesion complex assembly, epiblast elongation, and lineage differentiation. These defects are accompanied by a significant reduction in integrin β1 protein levels due to accelerated degradation through an MG-132-sensitive proteasomal pathway. Overexpression of integrin β1 or MG-132 treatment in mutant EBs largely rescues the phenotype. In addition, epiblast cells isolated from talin1-null EBs exhibit impaired cell spreading and focal adhesion formation. Transfection of the mutant cells with green fluorescent protein (GFP)-tagged wild-type but not mutant talin1 that is defective in integrin binding normalizes integrin β1 protein levels and restores focal adhesion formation. Significantly, cell adhesion and spreading are also improved by overexpression of integrin β1. All together, these results suggest that talin1 binding to integrin promotes epiblast adhesion and morphogenesis in part by preventing integrin β1 degradation.
Collapse
Affiliation(s)
- Jie Liu
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| | - Xiaowen He
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| | - Yanmei Qi
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| | - Xiaoxiang Tian
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| | - Susan J. Monkley
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | - David R. Critchley
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | - Siobhan A. Corbett
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| | - Stephen F. Lowry
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| | - Alan M. Graham
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| | - Shaohua Li
- Department of Surgery, Robert Wood Johnson Medical School-UMDNJ, New Brunswick, New Jersey 08903
| |
Collapse
|
42
|
Abstract
The development of multicellular organisms, as well as maintenance of organ architecture and function, requires robust regulation of cell fates. This is in part achieved by conserved signaling pathways through which cells process extracellular information and translate this information into changes in proliferation, differentiation, migration, and cell shape. Gene deletion studies in higher eukaryotes have assigned critical roles for components of the extracellular matrix (ECM) and their cellular receptors in a vast number of developmental processes, indicating that a large proportion of this signaling is regulated by cell-ECM interactions. In addition, genetic alterations in components of this signaling axis play causative roles in several human diseases. This review will discuss what genetic analyses in mice and lower organisms have taught us about adhesion signaling in development and disease.
Collapse
Affiliation(s)
- Sara A Wickström
- Paul Gerson Una Group, Skin Homeostasis and Ageing, Max Planck Institute for Biology of Ageing, 50937 Cologne, Germany.
| | | | | |
Collapse
|
43
|
Weng J, Liao M, Zou S, Bao J, Zhou J, Qu L, Feng R, Feng X, Zhao Z, Jing Z. Downregulation of FHL1 Expression in Thoracic Aortic Dissection: Implications in Aortic Wall Remodeling and Pathogenesis of Thoracic Aortic Dissection. Ann Vasc Surg 2011; 25:240-7. [DOI: 10.1016/j.avsg.2010.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 09/29/2010] [Accepted: 10/04/2010] [Indexed: 11/28/2022]
|
44
|
Kopp PM, Bate N, Hansen TM, Brindle NPJ, Praekelt U, Debrand E, Coleman S, Mazzeo D, Goult BT, Gingras AR, Pritchard CA, Critchley DR, Monkley SJ. Studies on the morphology and spreading of human endothelial cells define key inter- and intramolecular interactions for talin1. Eur J Cell Biol 2010; 89:661-73. [PMID: 20605055 PMCID: PMC2958305 DOI: 10.1016/j.ejcb.2010.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 05/14/2010] [Accepted: 05/17/2010] [Indexed: 01/07/2023] Open
Abstract
Talin binds to and activates integrins and is thought to couple them to cytoskeletal actin. However, functional studies on talin have been restricted by the fact that most cells express two talin isoforms. Here we show that human umbilical vein endothelial cells (HUVEC) express only talin1, and that talin1 knockdown inhibited focal adhesion (FA) assembly preventing the cells from maintaining a spread morphology, a phenotype that was rescued by GFP-mouse talin1. Thus HUVEC offer an ideal model system in which to conduct talin structure/function studies. Talin contains an N-terminal FERM domain (comprised of F1, F2 and F3 domains) and a C-terminal flexible rod. The F3 FERM domain binds β-integrin tails, and mutations in F3 that inhibited integrin binding (W359A) or activation (L325R) severely compromised the ability of GFP-talin1 to rescue the talin1 knockdown phenotype despite the presence of a second integrin-binding site in the talin rod. The talin rod contains several actin-binding sites (ABS), and mutations in the C-terminal ABS that reduced actin-binding impaired talin1 function, whereas those that increased binding resulted in more stable FAs. The results show that both the N-terminal integrin and C-terminal actin-binding functions of talin are essential to cell spreading and FA assembly. Finally, mutations that relieve talin auto-inhibition resulted in the rapid and excessive production of FA, highlighting the importance of talin regulation within the cell.
Collapse
Affiliation(s)
- Petra M Kopp
- Department of Biochemistry, University of Leicester, Leicester, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Gingras AR, Bate N, Goult BT, Patel B, Kopp PM, Emsley J, Barsukov IL, Roberts GCK, Critchley DR. Central region of talin has a unique fold that binds vinculin and actin. J Biol Chem 2010; 285:29577-87. [PMID: 20610383 PMCID: PMC2937989 DOI: 10.1074/jbc.m109.095455] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 05/13/2010] [Indexed: 12/22/2022] Open
Abstract
Talin is an adaptor protein that couples integrins to F-actin. Structural studies show that the N-terminal talin head contains an atypical FERM domain, whereas the N- and C-terminal parts of the talin rod include a series of α-helical bundles. However, determining the structure of the central part of the rod has proved problematic. Residues 1359-1659 are homologous to the MESDc1 gene product, and we therefore expressed this region of talin in Escherichia coli. The crystal structure shows a unique fold comprised of a 5- and 4-helix bundle. The 5-helix bundle is composed of nonsequential helices due to insertion of the 4-helix bundle into the loop at the C terminus of helix α3. The linker connecting the bundles forms a two-stranded anti-parallel β-sheet likely limiting the relative movement of the two bundles. Because the 5-helix bundle contains the N and C termini of this module, we propose that it is linked by short loops to adjacent bundles, whereas the 4-helix bundle protrudes from the rod. This suggests the 4-helix bundle has a unique role, and its pI (7.8) is higher than other rod domains. Both helical bundles contain vinculin-binding sites but that in the isolated 5-helix bundle is cryptic, whereas that in the isolated 4-helix bundle is constitutively active. In contrast, both bundles are required for actin binding. Finally, we show that the MESDc1 protein, which is predicted to have a similar fold, is a novel actin-binding protein.
Collapse
Affiliation(s)
- Alexandre R. Gingras
- From the Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN
| | - Neil Bate
- From the Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN
| | - Benjamin T. Goult
- From the Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN
| | - Bipin Patel
- From the Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN
| | - Petra M. Kopp
- From the Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN
| | - Jonas Emsley
- the Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, and
| | - Igor L. Barsukov
- the School of Biological Sciences, University of Liverpool, BioSciences Building, Liverpool L69 7ZB, United Kingdom
| | - Gordon C. K. Roberts
- From the Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN
| | - David R. Critchley
- From the Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN
| |
Collapse
|
46
|
Mace EM, Zhang J, Siminovitch KA, Takei F. Elucidation of the integrin LFA-1-mediated signaling pathway of actin polarization in natural killer cells. Blood 2010; 116:1272-9. [PMID: 20472831 DOI: 10.1182/blood-2009-12-261487] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The leukocyte integrin LFA-1 is critical for natural killer (NK) cell cytotoxicity as it mediates NK-cell adhesion to target cells and generates activating signals that lead to polarization of the actin cytoskeleton. However, the LFA-1-mediated signaling pathway is not fully understood. Here, we examined the subcellular localization of actin-associated proteins in wild-type, talin-deficient, and Wiskott-Aldrich Syndrome protein (WASP)-deficient NK cells bound to beads coated with the LFA-1 ligand intercellular adhesion molecule-1 (ICAM-1). In addition, we carried out coimmunoprecipitation analyses and also used a pharmacologic reagent to reduce the level of phosphatidylinositol-4,5-bisphosphate (PIP(2)). The results revealed the following signaling pathways. Upon ICAM-1 binding to LFA-1, talin redistributes to the site of LFA-1 ligation and initiates 2 signaling pathways. First, talin recruits the actin nucleating protein complex Arp2/3 via constitutive association of vinculin with talin and Arp2/3. Second, talin also associates with type I phosphatidylinositol 4-phosphate 5-kinase (PIPKI) and binding of LFA-1 to ICAM-1 results in localized increase in PIP(2). This increase in PIP(2) recruits WASP to the site of LFA-1 ligation where WASP promotes Arp2/3-mediated actin polymerization. These processes are critical for the initiation of NK cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Emily M Mace
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC
| | | | | | | |
Collapse
|
47
|
Falet H, Pollitt AY, Begonja AJ, Weber SE, Duerschmied D, Wagner DD, Watson SP, Hartwig JH. A novel interaction between FlnA and Syk regulates platelet ITAM-mediated receptor signaling and function. ACTA ACUST UNITED AC 2010; 207:1967-79. [PMID: 20713593 PMCID: PMC2931168 DOI: 10.1084/jem.20100222] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Filamin A (FlnA) cross-links actin filaments and connects the Von Willebrand factor receptor GPIb-IX-V to the underlying cytoskeleton in platelets. Because FlnA deficiency is embryonic lethal, mice lacking FlnA in platelets were generated by breeding FlnAloxP/loxP females with GATA1-Cre males. FlnAloxP/y GATA1-Cre males have a macrothrombocytopenia and increased tail bleeding times. FlnA-null platelets have decreased expression and altered surface distribution of GPIbα because they lack the normal cytoskeletal linkage of GPIbα to underlying actin filaments. This results in ∼70% less platelet coverage on collagen-coated surfaces at shear rates of 1,500/s, compared with wild-type platelets. Unexpectedly, however, immunoreceptor tyrosine-based activation motif (ITAM)- and ITAM-like–mediated signals are severely compromised in FlnA-null platelets. FlnA-null platelets fail to spread and have decreased α-granule secretion, integrin αIIbβ3 activation, and protein tyrosine phosphorylation, particularly that of the protein tyrosine kinase Syk and phospholipase C–γ2, in response to stimulation through the collagen receptor GPVI and the C-type lectin-like receptor 2. This signaling defect was traced to the loss of a novel FlnA–Syk interaction, as Syk binds to FlnA at immunoglobulin-like repeat 5. Our findings reveal that the interaction between FlnA and Syk regulates ITAM- and ITAM-like–containing receptor signaling and platelet function.
Collapse
Affiliation(s)
- Hervé Falet
- Division of Translational Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Nieves B, Jones CW, Ward R, Ohta Y, Reverte CG, LaFlamme SE. The NPIY motif in the integrin beta1 tail dictates the requirement for talin-1 in outside-in signaling. J Cell Sci 2010; 123:1216-26. [PMID: 20332112 PMCID: PMC2848110 DOI: 10.1242/jcs.056549] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2010] [Indexed: 12/11/2022] Open
Abstract
Protein interactions with the integrin beta-subunit cytoplasmic domain (beta-tail) are essential for adhesion-dependent processes, including cell spreading and the connection of integrins with actin filaments at adhesion sites. Talin-1 binds to the conserved membrane-proximal NPxY motif of beta-tails (NPIY in beta1 integrin) promoting the inside-out activation of integrins and providing a linkage between integrins and the actin cytoskeleton. Here, we characterize the role of interactions between talin-1 and beta-tail downstream of integrin activation, in the context of recombinant integrins containing either the wild type (WT) or the (YA) mutant beta1A tail, with a tyrosine to alanine substitution in the NPIY motif. In addition to inhibiting integrin activation, the YA mutation suppresses cell spreading, integrin signaling, focal adhesion and stress-fiber formation, as well as microtubule assembly. Constitutive activation of the mutant integrin restores these integrin-dependent processes, bringing into question the importance of the NPIY motif downstream of integrin activation. Depletion of talin-1 using TLN1 siRNA demonstrated that talin-1 is required for cell spreading, focal adhesion and stress-fiber formation, as well as microtubule assembly, even when cells are adhered by constitutively activated WT integrins. Depletion of talin-1 does not inhibit these processes when cells are adhered by constitutively activated mutant integrins, suggesting that the binding of an inhibitory protein to the NPIY motif negatively regulates integrin function when talin-1 is depleted. We identified filamin A (FLNa) as this inhibitory protein; it binds to the beta1A tail in an NPIY-dependent manner and inhibition of FLNa expression in talin-1-depleted cells restores integrin function when cells are adhered by constitutively activated WT integrins. FLNa binds FilGAP, which is a negative regulator of Rac activation. Expression of the dominant inhibitory mutant, FilGAP(DeltaGAP), which lacks GAP activity restores spreading in cells adhered by constitutively activated integrins containing the beta1A tail, but not by integrins containing the beta1D tail, which is known to bind poorly to FLNa. Together, these results suggest that the binding of talin-1 to the NPIY motif is required downstream of integrin activation to promote cell spreading by preventing the inappropriate recruitment of FLNa and FilGAP to the beta1A tail. Our studies emphasize the importance of understanding the mechanisms that regulate the differential binding FLNa and talin-1 to the beta1 tail downstream of integrin activation in promoting integrin function.
Collapse
Affiliation(s)
- Bethsaida Nieves
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| | - Christopher W. Jones
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| | - Rachel Ward
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| | - Yasutaka Ohta
- Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| | - Carlos G. Reverte
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| | - Susan E. LaFlamme
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| |
Collapse
|
49
|
Conti FJ, Monkley SJ, Wood MR, Critchley DR, Müller U. Talin 1 and 2 are required for myoblast fusion, sarcomere assembly and the maintenance of myotendinous junctions. Development 2009; 136:3597-606. [PMID: 19793892 PMCID: PMC2761109 DOI: 10.1242/dev.035857] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2009] [Indexed: 01/08/2023]
Abstract
Talin 1 and 2 connect integrins to the actin cytoskeleton and regulate the affinity of integrins for ligands. In skeletal muscle, talin 1 regulates the stability of myotendinous junctions (MTJs), but the function of talin 2 in skeletal muscle is not known. Here we show that MTJ integrity is affected in talin 2-deficient mice. Concomitant ablation of talin 1 and 2 leads to defects in myoblast fusion and sarcomere assembly, resembling defects in muscle lacking beta1 integrins. Talin 1/2-deficient myoblasts express functionally active beta1 integrins, suggesting that defects in muscle development are not primarily caused by defects in ligand binding, but rather by disruptions of the interaction of integrins with the cytoskeleton. Consistent with this finding, assembly of integrin adhesion complexes is perturbed in the remaining muscle fibers of talin 1/2-deficient mice. We conclude that talin 1 and 2 are crucial for skeletal muscle development, where they regulate myoblast fusion, sarcomere assembly and the maintenance of MTJs.
Collapse
Affiliation(s)
- Francesco J Conti
- The Scripps Research Institute, Department of Cell Biology and Institute of Childhood and Neglected Diseases, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
50
|
Boettner B, Van Aelst L. Control of cell adhesion dynamics by Rap1 signaling. Curr Opin Cell Biol 2009; 21:684-93. [PMID: 19615876 PMCID: PMC2841981 DOI: 10.1016/j.ceb.2009.06.004] [Citation(s) in RCA: 203] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 05/16/2009] [Accepted: 06/17/2009] [Indexed: 10/20/2022]
Abstract
Individual cells in their particular environments adhere to the extracellular matrix (ECM) and their neighbours via integrin-containing and cadherin-containing complexes, respectively. The dynamics of these interactions regulate the formation and maintenance of complex tissues. An expanding body of evidence accentuates the role of the small Rap1 GTPase and its associated signaling network in many of these processes. In this review we will discuss more recently revealed roles of Rap1 signaling by primarily focusing on functions of the Rap1 effectors RIAM, KRIT-1/CCM1 and AF-6/Afadin in junctional regulation of the vascular system and in epithelial cells. Furthermore, we will describe novel findings on the Rap activator PDZ-GEF in the regulation of cell-cell adhesion between epithelial cells and within a stem cell niche.
Collapse
Affiliation(s)
- Benjamin Boettner
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| | | |
Collapse
|