1
|
Biertümpfel C, Yamada Y, Vasquez-Montes V, Truong TV, Cada AK, Mizuno N. Biochemical and structural bases for talin ABSs-F-actin interactions. Proc Natl Acad Sci U S A 2025; 122:e2405922122. [PMID: 39903122 PMCID: PMC11831117 DOI: 10.1073/pnas.2405922122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Focal adhesions (FAs) are large intracellular macromolecular assemblies that play a critical role in cell polarization and migration. Talin serves as a direct connection between integrin receptor and actomyosin cytoskeleton within FAs. Talin contains three actin-binding sites (ABS1-3) that engage discreetly during the development of FAs, thus acting as a critical player in FA initiation and maturation. However, the molecular basis of the ABS-F-actin interactions remains unknown. Here, we explore interactions of ABSs with F-actin to understand the multivalent behavior of talin. Particularly, the cryo-EM structure of the F-actin-ABS3 complex at 2.9 Å shows ABS3 spanning through two actin monomers along the filament axis, each occupied by the R13 rod subdomain and the DD domain. The dimerization of ABS3 occurs through the DD domain where both protomers interact on the actin surface, and the dimerization of talin to the actin surface is necessary for the engagement to F-actin. The R13 helical bundle is distorted upon binding to F-actin and releases the H1 helix from the rest of the bundle. This phenomenon has also been observed with other tension-sensing proteins like vinculin and α-catenin, highlighting that unfolding is relevant for its force sensing activity. On the contrary, ABS2 (R4R8 subdomains), which is thought to be critical for the maintenance of mature FAs, had multiple F-actin-binding regions within ABS2 and the binding likely occurred by these subdomains running through the surface of F-actin, thus strengthening the interactions upon the maturation of FAs.
Collapse
Affiliation(s)
- Christian Biertümpfel
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20892
| | - Yurika Yamada
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20892
| | - Victor Vasquez-Montes
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20892
| | - Thien Van Truong
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20892
| | - A. King Cada
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20892
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD20892
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD20892
| |
Collapse
|
2
|
Song L, Shi X, Kovacs L, Han W, John J, Barman SA, Dong Z, Lucas R, Fulton DJR, Verin AD, Su Y. Calpain Promotes LPS-induced Lung Endothelial Barrier Dysfunction via Cleavage of Talin. Am J Respir Cell Mol Biol 2023; 69:678-688. [PMID: 37639326 PMCID: PMC10704117 DOI: 10.1165/rcmb.2023-0009oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
Acute lung injury (ALI) is characterized by lung vascular endothelial cell (EC) barrier compromise resulting in increased endothelial permeability and pulmonary edema. The infection of gram-negative bacteria that produce toxins like LPS is one of the major causes of ALI. LPS activates Toll-like receptor 4, leading to cytoskeleton reorganization, resulting in lung endothelial barrier disruption and pulmonary edema in ALI. However, the signaling pathways that lead to the cytoskeleton reorganization and lung microvascular EC barrier disruption remain largely unexplored. Here we show that LPS induces calpain activation and talin cleavage into head and rod domains and that inhibition of calpain attenuates talin cleavage, RhoA activation, and pulmonary EC barrier disruption in LPS-treated human lung microvascular ECs in vitro and lung EC barrier disruption and pulmonary edema induced by LPS in ALI in vivo. Moreover, overexpression of calpain causes talin cleavage and RhoA activation, myosin light chain (MLC) phosphorylation, and increases in actin stress fiber formation. Furthermore, knockdown of talin attenuates LPS-induced RhoA activation and MLC phosphorylation and increased stress fiber formation and mitigates LPS-induced lung microvascular endothelial barrier disruption. Additionally, overexpression of talin head and rod domains increases RhoA activation, MLC phosphorylation, and stress fiber formation and enhances lung endothelial barrier disruption. Finally, overexpression of cleavage-resistant talin mutant reduces LPS-induced increases in MLC phosphorylation in human lung microvascular ECs and attenuates LPS-induced lung microvascular endothelial barrier disruption. These results provide the first evidence that calpain mediates LPS-induced lung microvascular endothelial barrier disruption in ALI via cleavage of talin.
Collapse
Affiliation(s)
| | | | - Laszlo Kovacs
- Department of Pharmacology & Toxicology
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | | | - Joseph John
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | | | - Zheng Dong
- Department of Cellular Biology and Anatomy, and
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - David J. R. Fulton
- Department of Pharmacology & Toxicology
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - Alexander D. Verin
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - Yunchao Su
- Department of Pharmacology & Toxicology
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
3
|
Sadhanasatish T, Augustin K, Windgasse L, Chrostek-Grashoff A, Rief M, Grashoff C. A molecular optomechanics approach reveals functional relevance of force transduction across talin and desmoplakin. SCIENCE ADVANCES 2023; 9:eadg3347. [PMID: 37343090 DOI: 10.1126/sciadv.adg3347] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/17/2023] [Indexed: 06/23/2023]
Abstract
Many mechanobiological processes that govern development and tissue homeostasis are regulated on the level of individual molecular linkages, and a number of proteins experiencing piconewton-scale forces in cells have been identified. However, under which conditions these force-bearing linkages become critical for a given mechanobiological process is often still unclear. Here, we established an approach to revealing the mechanical function of intracellular molecules using molecular optomechanics. When applied to the integrin activator talin, the technique provides direct evidence that its role as a mechanical linker is indispensable for the maintenance of cell-matrix adhesions and overall cell integrity. Applying the technique to desmoplakin shows that mechanical engagement of desmosomes to intermediate filaments is expendable under homeostatic conditions yet strictly required for preserving cell-cell adhesion under stress. These results reveal a central role of talin and desmoplakin as mechanical linkers in cell adhesion structures and demonstrate that molecular optomechanics is a powerful tool to investigate the molecular details of mechanobiological processes.
Collapse
Affiliation(s)
- Tanmay Sadhanasatish
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| | - Katharina Augustin
- Center for Protein Assemblies and Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Lukas Windgasse
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| | - Anna Chrostek-Grashoff
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| | - Matthias Rief
- Center for Protein Assemblies and Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Carsten Grashoff
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| |
Collapse
|
4
|
Bhattacharjya S. The structural basis of β2 integrin intra-cellular multi-protein complexes. Biophys Rev 2022; 14:1183-1195. [PMID: 36345283 PMCID: PMC9636337 DOI: 10.1007/s12551-022-00995-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/24/2022] [Indexed: 01/03/2023] Open
Abstract
In multicellular organisms, cell adhesion is a pivotal physiological process which is essential for cell-cell communications, cell migration, and interactions with extracellular matrix. Integrins, a family of large hetero-dimeric type I membrane proteins, are known for driving cell adhesion functions. Among 24 different integrins, four β2 integrins, αL β2, αM β2, αX β2 and αD β2, are specific for cell adhesion and migration of leukocytes. Many cytosolic proteins interact with short cytosolic tails (CTs) of β2 and other integrins which are essential in bi-directional signaling processes. Further, phosphorylation of CTs of integrins regulates binding of intra-cellular proteins and signaling systems. In this review, recent advances in structures and interactions of multi-protein complexes of integrin tails, with a focus on β2 integrin, and cytosolic proteins are discussed along with a proposed future direction.
Collapse
Affiliation(s)
- Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551 Singapore
| |
Collapse
|
5
|
Zanjani LS, Vafaei S, Abolhasani M, Fattahi F, Madjd Z. Prognostic value of Talin-1 in renal cell carcinoma and its association with B7-H3. Cancer Biomark 2022; 35:269-292. [DOI: 10.3233/cbm-220018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
METHODS: Talin-1 protein was demonstrated as a potential prognostic marker in renal cell carcinoma (RCC) using bioinformatics analysis. We, therefore, examined the protein expression levels and prognostic significance of Talin-1 with a clinical follow-up in a total of 269 tissue specimens from three important subtypes of RCC and 30 adjacent normal samples using immunohistochemistry. Then, we used combined analysis with B7-H3 to investigate higher prognostic values. RESULTS: The results showed that high membranous and cytoplasmic expression of Talin-1 was significantly associated with advanced nucleolar grade, microvascular invasion, histological tumor necrosis, and invasion to Gerota’s fascia in clear cell RCC (ccRCC). In addition, high membranous and cytoplasmic expression of Talin-1 was found to be associated with significantly poorer disease-specific survival (DSS) and progression-free survival (PFS). Moreover, increased cytoplasmic expression of Talin-1High/B7-H3High compared to the other phenotypes was associated with tumor aggressiveness and progression of the disease, and predicted a worse clinical outcome, which may be an effective biomarker to identify ccRCC patients at high risk of recurrence and metastasis. CONCLUSIONS: Collectively, these observations indicate that Talin-1 is an important molecule involved in the spread and progression of ccRCC when expressed particularly in the cytoplasm and may serve as a novel prognostic biomarker in this subtype. Furthermore, a combined analysis of Talin-1/B7-H3 indicated an effective biomarker to predict the progression of disease and prognosis in ccRCC.
Collapse
Affiliation(s)
- Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Somayeh Vafaei
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Integrin Regulators in Neutrophils. Cells 2022; 11:cells11132025. [PMID: 35805108 PMCID: PMC9266208 DOI: 10.3390/cells11132025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Neutrophils are the most abundant leukocytes in humans and are critical for innate immunity and inflammation. Integrins are critical for neutrophil functions, especially for their recruitment to sites of inflammation or infections. Integrin conformational changes during activation have been heavily investigated but are still not fully understood. Many regulators, such as talin, Rap1-interacting adaptor molecule (RIAM), Rap1, and kindlin, are critical for integrin activation and might be potential targets for integrin-regulating drugs in treating inflammatory diseases. In this review, we outline integrin activation regulators in neutrophils with a focus on the above critical regulators, as well as newly discovered modulators that are involved in integrin activation.
Collapse
|
7
|
Gautam D, Kailashiya J, Tiwari A, Chaurasia RN, Annarapu GK, Guchhait P, Dash D. Fibrinogen Mitigates Prion-Mediated Platelet Activation and Neuronal Cell Toxicity. Front Cell Dev Biol 2022; 10:834016. [PMID: 35386203 PMCID: PMC8977893 DOI: 10.3389/fcell.2022.834016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/17/2022] [Indexed: 11/23/2022] Open
Abstract
Prion peptide (PrP) misfolds to infectious scrapie isoform, the β pleat-rich insoluble fibrils responsible for neurodegeneration and fatal conformational diseases in humans. The amino acid sequence 106–126 from prion proteins, PrP(106–126), is highly amyloidogenic and implicated in prion-induced pathologies. Here, we report a novel interaction between PrP(106–126) and the thrombogenic plasma protein fibrinogen that can lead to mitigation of prion-mediated pro-thrombotic responses in human platelets as well as significant decline in neuronal toxicity. Thus, prior exposure to fibrinogen-restrained PrP-induced rise in cytosolic calcium, calpain activation, and shedding of extracellular vesicles in platelets while it, too, averted cytotoxicity of neuronal cells triggered by prion peptide. Interestingly, PrP was found to accelerate fibrin-rich clot formation, which was resistant to plasmin-mediated fibrinolysis, consistent with enhanced thrombus stability provoked by PrP. We propose that PrP-fibrinogen interaction can be clinically exploited further for prevention and management of infectious prion related disorders. Small molecules or peptides mimicking PrP-binding sites on fibrinogen can potentially mitigate PrP-induced cellular toxicity while also preventing the negative impact of PrP on fibrin clot formation and lysis.
Collapse
Affiliation(s)
- Deepa Gautam
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jyotsna Kailashiya
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Arundhati Tiwari
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rameshwar Nath Chaurasia
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Gowtham K. Annarapu
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Debabrata Dash,
| |
Collapse
|
8
|
Zhao Y, Yue S, Zhou X, Guo J, Ma S, Chen Q. O-GlcNAc transferase promotes the nuclear localization of the focal adhesion-associated protein Zyxin to regulate UV-induced cell death. J Biol Chem 2022; 298:101776. [PMID: 35227760 PMCID: PMC8988012 DOI: 10.1016/j.jbc.2022.101776] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 11/18/2022] Open
Abstract
Zyxin is a zinc-binding phosphoprotein known to regulate cell migration, adhesion, and cell survival. Zyxin also plays a role in signal transduction between focal adhesions and the nuclear compartment. However, the mechanism of Zyxin shuttling to nucleus is still unclear. Here, we identify that the GlcNAc transferase (O-linked GlcNAc [O-GlcNAc] transferase) can O-GlcNAcylate Zyxin and regulate its nuclear localization. We show that O-GlcNAc transferase O-GlcNAcylates Zyxin at two residues, serine 169 (Ser-169) and Ser-246. In addition, O-GlcNAcylation of Ser-169, but not Ser-246, enhances its interaction with 14-3-3γ, which is a phosphoserine/threonine-binding protein and is reported to bind with phosphorylated Zyxin. Furthermore, we found that 14-3-3γ could promote the nuclear localization of Zyxin after Ser-169 O-GlcNAcylation by affecting the function of the N-terminal nuclear export signal sequence; functionally, UV treatment increases the O-GlcNAcylation of Zyxin, which may enhance the nuclear location of Zyxin. Finally, Zyxin in the nucleus maintains homeodomain-interacting protein kinase 2 stability and promotes UV-induced cell death. In conclusion, we uncover that the nuclear localization of Zyxin can be regulated by its O-GlcNAcylation, and that this protein may regulate UV-induced cell death.
Collapse
|
9
|
Zhang P, Azizi L, Kukkurainen S, Gao T, Baikoghli M, Jacquier MC, Sun Y, Määttä JAE, Cheng RH, Wehrle-Haller B, Hytönen VP, Wu J. Crystal structure of the FERM-folded talin head reveals the determinants for integrin binding. Proc Natl Acad Sci U S A 2020; 117:32402-32412. [PMID: 33288722 PMCID: PMC7768682 DOI: 10.1073/pnas.2014583117] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Binding of the intracellular adapter proteins talin and its cofactor, kindlin, to the integrin receptors induces integrin activation and clustering. These processes are essential for cell adhesion, migration, and organ development. Although the talin head, the integrin-binding segment in talin, possesses a typical FERM-domain sequence, a truncated form has been crystallized in an unexpected, elongated form. This form, however, lacks a C-terminal fragment and possesses reduced β3-integrin binding. Here, we present a crystal structure of a full-length talin head in complex with the β3-integrin tail. The structure reveals a compact FERM-like conformation and a tightly associated N-P-L-Y motif of β3-integrin. A critical C-terminal poly-lysine motif mediates FERM interdomain contacts and assures the tight association with the β3-integrin cytoplasmic segment. Removal of the poly-lysine motif or disrupting the FERM-folded configuration of the talin head significantly impairs integrin activation and clustering. Therefore, structural characterization of the FERM-folded active talin head provides fundamental understanding of the regulatory mechanism of integrin function.
Collapse
Affiliation(s)
- Pingfeng Zhang
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Latifeh Azizi
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Sampo Kukkurainen
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Tong Gao
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616
| | - Marie-Claude Jacquier
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211 Geneva 4, Switzerland
| | - Yijuan Sun
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Juha A E Määttä
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211 Geneva 4, Switzerland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland;
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Jinhua Wu
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111;
| |
Collapse
|
10
|
Boujemaa-Paterski R, Martins B, Eibauer M, Beales CT, Geiger B, Medalia O. Talin-activated vinculin interacts with branched actin networks to initiate bundles. eLife 2020; 9:e53990. [PMID: 33185186 PMCID: PMC7682986 DOI: 10.7554/elife.53990] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
Vinculin plays a fundamental role in integrin-mediated cell adhesion. Activated by talin, it interacts with diverse adhesome components, enabling mechanical coupling between the actin cytoskeleton and the extracellular matrix. Here we studied the interactions of activated full-length vinculin with actin and the way it regulates the organization and dynamics of the Arp2/3 complex-mediated branched actin network. Through a combination of surface patterning and light microscopy experiments we show that vinculin can bundle dendritic actin networks through rapid binding and filament crosslinking. We show that vinculin promotes stable but flexible actin bundles having a mixed-polarity organization, as confirmed by cryo-electron tomography. Adhesion-like synthetic design of vinculin activation by surface-bound talin revealed that clustered vinculin can initiate and immobilize bundles from mobile Arp2/3-branched networks. Our results provide a molecular basis for coordinate actin bundle formation at nascent adhesions.
Collapse
Affiliation(s)
- Rajaa Boujemaa-Paterski
- Department of Biochemistry, University of ZurichZurichSwitzerland
- Université Grenoble AlpesGrenobleFrance
| | - Bruno Martins
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Matthias Eibauer
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Charlie T Beales
- Department of Biochemistry, University of ZurichZurichSwitzerland
| | - Benjamin Geiger
- Department of Immunology, Weizmann Institute of ScienceRehovotIsrael
| | - Ohad Medalia
- Department of Biochemistry, University of ZurichZurichSwitzerland
| |
Collapse
|
11
|
Kukkurainen S, Azizi L, Zhang P, Jacquier MC, Baikoghli M, von Essen M, Tuukkanen A, Laitaoja M, Liu X, Rahikainen R, Orłowski A, Jänis J, Määttä JAE, Varjosalo M, Vattulainen I, Róg T, Svergun D, Cheng RH, Wu J, Hytönen VP, Wehrle-Haller B. The F1 loop of the talin head domain acts as a gatekeeper in integrin activation and clustering. J Cell Sci 2020; 133:jcs239202. [PMID: 33046605 PMCID: PMC10679385 DOI: 10.1242/jcs.239202] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 07/31/2020] [Indexed: 12/15/2022] Open
Abstract
Integrin activation and clustering by talin are early steps of cell adhesion. Membrane-bound talin head domain and kindlin bind to the β integrin cytoplasmic tail, cooperating to activate the heterodimeric integrin, and the talin head domain induces integrin clustering in the presence of Mn2+ Here we show that kindlin-1 can replace Mn2+ to mediate β3 integrin clustering induced by the talin head, but not that induced by the F2-F3 fragment of talin. Integrin clustering mediated by kindlin-1 and the talin head was lost upon deletion of the flexible loop within the talin head F1 subdomain. Further mutagenesis identified hydrophobic and acidic motifs in the F1 loop responsible for β3 integrin clustering. Modeling, computational and cysteine crosslinking studies showed direct and catalytic interactions of the acidic F1 loop motif with the juxtamembrane domains of α- and β3-integrins, in order to activate the β3 integrin heterodimer, further detailing the mechanism by which the talin-kindlin complex activates and clusters integrins. Moreover, the F1 loop interaction with the β3 integrin tail required the newly identified compact FERM fold of the talin head, which positions the F1 loop next to the inner membrane clasp of the talin-bound integrin heterodimer.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sampo Kukkurainen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Latifeh Azizi
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Pingfeng Zhang
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Marie-Claude Jacquier
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, 1 Shields Ave, Davis, CA 95616, USA
| | - Magdaléna von Essen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Anne Tuukkanen
- EMBL Hamburg c/o DESY, European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Mikko Laitaoja
- Department of Chemistry, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland
| | - Xiaonan Liu
- Proteomics Unit, Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Rolle Rahikainen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Adam Orłowski
- Proteomics Unit, Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Janne Jänis
- Department of Chemistry, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland
| | - Juha A E Määttä
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Markku Varjosalo
- Proteomics Unit, Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Ilpo Vattulainen
- Computational Physics Laboratory, Tampere University, FI-33520 Tampere, Finland
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Tomasz Róg
- Computational Physics Laboratory, Tampere University, FI-33520 Tampere, Finland
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
| | - Dmitri Svergun
- EMBL Hamburg c/o DESY, European Molecular Biology Laboratory, Notkestrasse 85, 22607 Hamburg, Germany
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, 1 Shields Ave, Davis, CA 95616, USA
| | - Jinhua Wu
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| |
Collapse
|
12
|
Kumar R, Donakonda S, Müller SA, Lichtenthaler SF, Bötzel K, Höglinger GU, Koeglsperger T. Basic Fibroblast Growth Factor 2-Induced Proteome Changes Endorse Lewy Body Pathology in Hippocampal Neurons. iScience 2020; 23:101349. [PMID: 32707433 PMCID: PMC7381695 DOI: 10.1016/j.isci.2020.101349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/11/2020] [Accepted: 07/03/2020] [Indexed: 12/15/2022] Open
Abstract
Hippocampal Lewy body pathology (LBP) is associated with changes in neurotrophic factor signaling and neuronal energy metabolism. LBP progression is attributed to the aggregation of α-synuclein (α-Syn) and its cell-to-cell transmission via extracellular vehicles (EVs). We recently discovered an enhanced EV release in basic fibroblast growth factor (bFGF)-treated hippocampal neurons. Here, we examined the EV and cell lysate proteome changes in bFGF-treated hippocampal neurons. We identified n = 2,310 differentially expressed proteins (DEPs) induced by bFGF. We applied weighted protein co-expression network analysis (WPCNA) to generate protein modules from DEPs and mapped them to published LBP datasets. This approach revealed n = 532 LBP-linked DEPs comprising key α-Syn-interacting proteins, LBP-associated RNA-binding proteins (RBPs), and neuronal ion channels and receptors that can impact LBP onset and progression. In summary, our deep proteomic analysis affirms the potential influence of bFGF signaling on LBP-related proteome changes and associated molecular interactions.
Collapse
Affiliation(s)
- Rohit Kumar
- German Center for Neurodegenerative Diseases (DZNE), 81337 Munich, Germany; Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Department of Neurology, Ludwig Maximilian University, 81377 Munich, Germany.
| | - Sainitin Donakonda
- Institute of Immunology and Experimental Oncology, Technical University of Munich, 81675 Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), 81337 Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), 81337 Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Kai Bötzel
- Department of Neurology, Ludwig Maximilian University, 81377 Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), 81337 Munich, Germany; Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Department of Neurology, Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Thomas Koeglsperger
- German Center for Neurodegenerative Diseases (DZNE), 81337 Munich, Germany; Department of Neurology, Ludwig Maximilian University, 81377 Munich, Germany.
| |
Collapse
|
13
|
Chen C, Manso AM, Ross RS. Talin and Kindlin as Integrin-Activating Proteins: Focus on the Heart. Pediatr Cardiol 2019; 40:1401-1409. [PMID: 31367953 PMCID: PMC7590617 DOI: 10.1007/s00246-019-02167-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/18/2019] [Indexed: 01/11/2023]
Abstract
Integrin receptors enable cells to sense and respond to their chemical and physical environment. As a class of membrane receptors, they provide a dynamic, tightly regulated link between the extracellular matrix or cellular counter-receptors and intracellular cytoskeletal and signaling networks. They enable transmission of mechanical force across the plasma membrane, and particularly for cardiomyocytes, may sense the mechanical load placed on cells. Talins and Kindlins are two families of FERM-domain proteins which bind the cytoplasmic tail of integrins, recruit cytoskeletal and signaling proteins involved in mechano-transduction, and those which synergize to activate integrins, allowing the integrins to physically change and bind to extracellular ligands. In this review, we will discuss the roles of talin and kindlin, particularly as integrin activators, with a focus on cardiac myocytes.
Collapse
Affiliation(s)
- Chao Chen
- Department of Medicine/Cardiology, UCSD School of Medicine, La Jolla, CA, 92093, USA
- Department of Medicine/Cardiology, Veterans Administration Healthcare, San Diego, CA, 92161, USA
| | - Ana Maria Manso
- Department of Medicine/Cardiology, UCSD School of Medicine, La Jolla, CA, 92093, USA
- Department of Medicine/Cardiology, Veterans Administration Healthcare, San Diego, CA, 92161, USA
| | - Robert S Ross
- Department of Medicine/Cardiology, UCSD School of Medicine, La Jolla, CA, 92093, USA.
- Department of Medicine/Cardiology, Veterans Administration Healthcare, San Diego, CA, 92161, USA.
- University of California, San Diego, Biomedical Research Facility 2, Room 2A-17, 9500 Gilman Drive #0613-C, La Jolla, CA, 92093-0613, USA.
| |
Collapse
|
14
|
Onochie OE, Onyejose AJ, Rich CB, Trinkaus-Randall V. The Role of Hypoxia in Corneal Extracellular Matrix Deposition and Cell Motility. Anat Rec (Hoboken) 2019; 303:1703-1716. [PMID: 30861330 DOI: 10.1002/ar.24110] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 11/29/2018] [Accepted: 12/17/2018] [Indexed: 12/24/2022]
Abstract
The cornea is an excellent model tissue to study how cells adapt to periods of hypoxia as it is naturally exposed to diurnal fluxes in oxygen. It is avascular, transparent, and highly innervated. In certain pathologies, such as diabetes, limbal stem cell deficiency, or trauma, the cornea may be exposed to hypoxia for variable lengths of time. Due to its avascularity, the cornea requires atmospheric oxygen, and a reduction in oxygen availability can impair its physiology and function. We hypothesize that hypoxia alters membrane stiffness and the deposition of matrix proteins, leading to changes in cell migration, focal adhesion formation, and wound repair. Two systems-a 3D corneal organ culture model and polyacrylamide substrates of varying stiffness-were used to examine the response of corneal epithelium to normoxic and hypoxic environments. Exposure to hypoxia alters the deposition of the matrix proteins such as laminin and Type IV collagen. In addition, previous studies had shown a change in fibronectin after injury. Studies performed on matrix-coated acrylamide substrates ranging from 0.2 to 50 kPa revealed stiffness-dependent changes in cell morphology. The localization, number, and length of paxillin pY118- and vinculin pY1065-containing focal adhesions were different in wounded corneas and in human corneal epithelial cells incubated in hypoxic environments. Overall, these results demonstrate that low-oxygenated environments modify the composition of the extracellular matrix, basal lamina stiffness, and focal adhesion dynamics, leading to alterations in the function of the cornea. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Obianamma E Onochie
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Anwuli J Onyejose
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts
| | - Celeste B Rich
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Vickery Trinkaus-Randall
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts.,Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
15
|
Turley TN, Theis JL, Sundsbak RS, Evans JM, O'Byrne MM, Gulati R, Tweet MS, Hayes SN, Olson TM. Rare Missense Variants in TLN1 Are Associated With Familial and Sporadic Spontaneous Coronary Artery Dissection. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 12:e002437. [PMID: 30888838 DOI: 10.1161/circgen.118.002437] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Spontaneous coronary artery dissection (SCAD) is an uncommon idiopathic disorder predominantly affecting young, otherwise healthy women. Rare familial cases reveal a genetic predisposition to disease. The aim of this study was to identify a novel susceptibility gene for SCAD. METHODS Whole-exome sequencing was performed in a family comprised of 3 affected individuals and filtered to identify rare, predicted deleterious, segregating variants. Immunohistochemical staining was used to evaluate protein expression of the identified candidate gene. The prevalence and spectrum of rare (<0.1%) variants within binding domains was determined by next-generation sequencing or denaturing high-performance liquid chromatography in a sporadic SCAD cohort of 675 unrelated individuals. RESULTS We identified a rare heterozygous missense variant within a highly conserved β-integrin-binding domain of TLN1 segregating with familial SCAD. TLN1 encodes talin 1-a large cytoplasmic protein of the integrin adhesion complex that links the actin cytoskeleton and extracellular matrix. Consistent with high mRNA expression in arterial tissues, robust immunohistochemical staining of talin 1 was demonstrated in coronary arteries. Nine additional rare heterozygous missense variants in TLN1 were identified in 10 sporadic cases. Incomplete penetrance, suggesting genetic or environmental modifiers of this episodic disorder, was evident in the familial case and 5 individuals with sporadic SCAD from whom parental DNA was available. CONCLUSIONS Our findings reveal TLN1 as a disease-associated gene in familial and sporadic SCAD and, together with abnormal vascular phenotypes reported in animal models of talin 1 disruption, implicate impaired structural integrity of the coronary artery cytoskeleton in SCAD susceptibility.
Collapse
Affiliation(s)
- Tamiel N Turley
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology and Experimental Therapeutics (T.N.T.), Mayo Clinic, Rochester, MN.,Cardiovascular Genetics Research Laboratory (T.N.T., J.L.T., R.S.S., T.M.O.), Mayo Clinic, Rochester, MN
| | - Jeanne L Theis
- Cardiovascular Genetics Research Laboratory (T.N.T., J.L.T., R.S.S., T.M.O.), Mayo Clinic, Rochester, MN
| | - Rhianna S Sundsbak
- Cardiovascular Genetics Research Laboratory (T.N.T., J.L.T., R.S.S., T.M.O.), Mayo Clinic, Rochester, MN
| | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (J.M.E., M.M.O.), Mayo Clinic, Rochester, MN
| | - Megan M O'Byrne
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (J.M.E., M.M.O.), Mayo Clinic, Rochester, MN
| | - Rajiv Gulati
- Department of Cardiovascular Medicine (R.G., M.S.T., S.N.H., T.M.O.), Mayo Clinic, Rochester, MN
| | - Marysia S Tweet
- Department of Cardiovascular Medicine (R.G., M.S.T., S.N.H., T.M.O.), Mayo Clinic, Rochester, MN
| | - Sharonne N Hayes
- Department of Cardiovascular Medicine (R.G., M.S.T., S.N.H., T.M.O.), Mayo Clinic, Rochester, MN
| | - Timothy M Olson
- Cardiovascular Genetics Research Laboratory (T.N.T., J.L.T., R.S.S., T.M.O.), Mayo Clinic, Rochester, MN.,Department of Cardiovascular Medicine (R.G., M.S.T., S.N.H., T.M.O.), Mayo Clinic, Rochester, MN.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine (T.M.O.), Mayo Clinic, Rochester, MN
| |
Collapse
|
16
|
The extracellular matrix-myosin pathway in mechanotransduction: from molecule to tissue. Emerg Top Life Sci 2018; 2:727-737. [PMID: 33530663 PMCID: PMC7289002 DOI: 10.1042/etls20180043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 12/23/2022]
Abstract
Mechanotransduction via the extracellular matrix (ECM)–myosin pathway is involved in determining cell morphology during development and in coupling external transient mechanical stimuli to the reorganization of the cytoskeleton. Here, we present a review on the molecular mechanisms involved in this pathway and how they influence cellular development and organization. We investigate key proteins involved in the ECM–myosin pathway and discuss how specific binding events and conformational changes under force are related to mechanical signaling. We connect these molecular mechanisms with observed morphological changes at the cellular and organism level. Finally, we propose a model encompassing the biomechanical signals along the ECM–myosin pathway and how it could be involved in cell adhesion, cell migration, and tissue architecture.
Collapse
|
17
|
Kanoldt V, Fischer L, Grashoff C. Unforgettable force – crosstalk and memory of mechanosensitive structures. Biol Chem 2018; 400:687-698. [DOI: 10.1515/hsz-2018-0328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/11/2018] [Indexed: 12/11/2022]
Abstract
Abstract
The ability of cells to sense and respond to mechanical stimuli is crucial for many developmental and homeostatic processes, while mechanical dysfunction of cells has been associated with numerous pathologies including muscular dystrophies, cardiovascular defects and epithelial disorders. Yet, how cells detect and process mechanical information is still largely unclear. In this review, we outline major mechanisms underlying cellular mechanotransduction and we summarize the current understanding of how cells integrate information from distinct mechanosensitive structures to mediate complex mechanoresponses. We also discuss the concept of mechanical memory and describe how cells store information on previous mechanical events for different periods of time.
Collapse
Affiliation(s)
- Verena Kanoldt
- Group of Molecular Mechanotransduction , Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
| | - Lisa Fischer
- Group of Molecular Mechanotransduction , Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
| | - Carsten Grashoff
- Group of Molecular Mechanotransduction , Max Planck Institute of Biochemistry , 82152 Martinsried , Germany
- Department of Quantitative Cell Biology , Institute of Molecular Cell Biology, University of Münster , 48149 Münster , Germany
| |
Collapse
|
18
|
Mykuliak VV, Haining AWM, von Essen M, del Río Hernández A, Hytönen VP. Mechanical unfolding reveals stable 3-helix intermediates in talin and α-catenin. PLoS Comput Biol 2018; 14:e1006126. [PMID: 29698481 PMCID: PMC5940241 DOI: 10.1371/journal.pcbi.1006126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/08/2018] [Accepted: 04/06/2018] [Indexed: 11/18/2022] Open
Abstract
Mechanical stability is a key feature in the regulation of structural scaffolding proteins and their functions. Despite the abundance of α-helical structures among the human proteome and their undisputed importance in health and disease, the fundamental principles of their behavior under mechanical load are poorly understood. Talin and α-catenin are two key molecules in focal adhesions and adherens junctions, respectively. In this study, we used a combination of atomistic steered molecular dynamics (SMD) simulations, polyprotein engineering, and single-molecule atomic force microscopy (smAFM) to investigate unfolding of these proteins. SMD simulations revealed that talin rod α-helix bundles as well as α-catenin α-helix domains unfold through stable 3-helix intermediates. While the 5-helix bundles were found to be mechanically stable, a second stable conformation corresponding to the 3-helix state was revealed. Mechanically weaker 4-helix bundles easily unfolded into a stable 3-helix conformation. The results of smAFM experiments were in agreement with the findings of the computational simulations. The disulfide clamp mutants, designed to protect the stable state, support the 3-helix intermediate model in both experimental and computational setups. As a result, multiple discrete unfolding intermediate states in the talin and α-catenin unfolding pathway were discovered. Better understanding of the mechanical unfolding mechanism of α-helix proteins is a key step towards comprehensive models describing the mechanoregulation of proteins.
Collapse
Affiliation(s)
- Vasyl V. Mykuliak
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Alexander William M. Haining
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Magdaléna von Essen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Armando del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
- * E-mail: (AdRH); (VPH)
| | - Vesa P. Hytönen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
- * E-mail: (AdRH); (VPH)
| |
Collapse
|
19
|
Ringer P, Colo G, Fässler R, Grashoff C. Sensing the mechano-chemical properties of the extracellular matrix. Matrix Biol 2017; 64:6-16. [DOI: 10.1016/j.matbio.2017.03.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
|
20
|
Baxter NJ, Zacharchenko T, Barsukov IL, Williamson MP. Pressure-Dependent Chemical Shifts in the R3 Domain of Talin Show that It Is Thermodynamically Poised for Binding to Either Vinculin or RIAM. Structure 2017; 25:1856-1866.e2. [PMID: 29153504 DOI: 10.1016/j.str.2017.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/13/2017] [Accepted: 10/26/2017] [Indexed: 12/25/2022]
Abstract
Talin mediates attachment of the cell to the extracellular matrix. It is targeted by the Rap1 effector RIAM to focal adhesion sites and subsequently undergoes force-induced conformational opening to recruit the actin-interacting protein vinculin. The conformational switch involves the talin R3 domain, which binds RIAM when closed and vinculin when open. Here, we apply pressure to R3 and measure 1H, 15N, and 13C chemical shift changes, which are fitted using a simple model, and indicate that R3 is only 50% closed: the closed form is a four-helix bundle, while in the open state helix 1 is twisted out. Strikingly, a mutant of R3 that binds RIAM with an affinity similar to wild-type but more weakly to vinculin is shown to be 0.84 kJ mol-1 more stable when closed. These results demonstrate that R3 is thermodynamically poised to bind either RIAM or vinculin, and thus constitutes a good mechanosensitive switch.
Collapse
Affiliation(s)
- Nicola J Baxter
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK
| | - Thomas Zacharchenko
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - Igor L Barsukov
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - Mike P Williamson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
21
|
Zhuang B, Hu Y, Fan X, Li M, Zhu J, Liu H, Cao L, Liang D, Zhang J, Yu Z, Han S. Peptidomic Analysis of Maternal Serum to Identify Biomarker Candidates for Prenatal Diagnosis of Tetralogy of Fallot. J Cell Biochem 2017; 119:468-477. [DOI: 10.1002/jcb.26204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/08/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Bin Zhuang
- Department of PediatricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
- Fourth Clinical Medicine CollegeNanjing Medical UniversityNanjing210029China
| | - Yin Hu
- Department of PediatricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
- Fourth Clinical Medicine CollegeNanjing Medical UniversityNanjing210029China
| | - Xuemei Fan
- Department of ObstetricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| | - Mengmeng Li
- Department of PediatricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| | - Jingai Zhu
- Department of PediatricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| | - Heng Liu
- Department of PediatricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
- Fourth Clinical Medicine CollegeNanjing Medical UniversityNanjing210029China
| | - Li Cao
- Department of UltrasoundObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| | - Dong Liang
- Department of Prenatal DiagnosisObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| | - Jingjing Zhang
- Department of Prenatal DiagnosisObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| | - Zhangbin Yu
- Department of PediatricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| | - Shuping Han
- Department of PediatricsObstetrics and Gynecology Hospital Affiliated to Nanjing Medical UniversityNanjing210004China
| |
Collapse
|
22
|
Maki K, Nakao N, Adachi T. Nano-mechanical characterization of tension-sensitive helix bundles in talin rod. Biochem Biophys Res Commun 2017; 484:372-377. [PMID: 28131835 DOI: 10.1016/j.bbrc.2017.01.127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 01/11/2023]
Abstract
Tension-induced exposure of a cryptic signaling binding site is one of the most fundamental mechanisms in molecular mechanotransduction. Helix bundles in rod domains of talin, a tension-sensing protein at focal adhesions, unfurl under tension to expose cryptic vinculin binding sites. Although the difference in their mechanical stabilities would determine which helix bundle is tension-sensitive, their respective mechanical behaviors under tension have not been characterized. In this study, we evaluated the mechanical behaviors of residues 486-654 and 754-889 of talin, which form helix bundles with low and high tension-sensitivity, by employing AFM nano-tensile testing. As a result, residues 754-889 exhibited lower unfolding energy for complete unfolding than residues 486-654. In addition, we found that residues 754-889 transition into intermediate conformations under lower tension than residues 486-654. Furthermore, residues 754-889 showed shorter persistence length in the intermediate conformation than residues 486-654, suggesting that residues 754-889 under tension exhibit separated α-helices, while residues 486-654 assume a compact conformation with inter-helix interactions. Therefore, we suggest that residues 754-889 of talin work as a tension-sensitive domain to recruit vinculin at the early stage of focal adhesion development, while residues 486-654 contribute to rather robust tension-sensitivity by recruiting vinculin under high tension.
Collapse
Affiliation(s)
- Koichiro Maki
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Yoshida Honmachi, Sakyo, Kyoto 606-8501, Japan
| | - Nobuhiko Nakao
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Yoshida Honmachi, Sakyo, Kyoto 606-8501, Japan
| | - Taiji Adachi
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto 606-8507, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Yoshida Honmachi, Sakyo, Kyoto 606-8501, Japan.
| |
Collapse
|
23
|
Ye X, McLean MA, Sligar SG. Phosphatidylinositol 4,5-Bisphosphate Modulates the Affinity of Talin-1 for Phospholipid Bilayers and Activates Its Autoinhibited Form. Biochemistry 2016; 55:5038-48. [PMID: 27548281 DOI: 10.1021/acs.biochem.6b00497] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Integrins are vital transmembrane receptors that mediate cell-cell and cell-extracellular matrix interactions and signaling. Talin is a 270 kDa protein and is considered a key regulator of integrin activity. The interaction between talin and integrin is commonly regarded as the final step of inside-out activation. In the cytosol, talin adopts an autoinhibited conformation, in which the C-terminal rod domain binds the N-terminal head domain, preventing the interactions of the head domain with the membrane surface and the integrin cytoplasmic domain. It has long been suggested that the presence of phosphatidylinositol 4,5-bisphosphate (PIP2) at focal adhesions plays a role in activating talin. However, a detailed picture and mechanism of PIP2 activation of autoinhibited talin remains elusive. Here, we use a fluorescence resonance energy transfer-based binding assay to measure the affinity of talin and lipid bilayers harboring anionic lipids. Results show that the R9 and R12R13 segments of the talin rod domain inhibit the binding of the talin head domain (THD) to anionic lipid bilayers. In contrast, we show that the binding of the THD to bilayers containing PIP2 is insensitive to the presence of the inhibitor domains, thereby directly implicating PIP2 as an effective activator of talin. Furthermore, we have mapped the activation to the interaction of PIP2 with the F2F3 domain of the talin head, showing that PIP2 plays a critical role in the regulation of the autoinhibited form of talin and stimulates recruitment of talin to the membrane, which is essential for integrin inside-out signaling.
Collapse
Affiliation(s)
- Xin Ye
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois , Urbana, Illinois 61801, United States
| | - Mark A McLean
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois , Urbana, Illinois 61801, United States
| | - Stephen G Sligar
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois , Urbana, Illinois 61801, United States
| |
Collapse
|
24
|
Uehara K, Uehara A. Differentiated localizations of phosphorylated focal adhesion kinase in endothelial cells of rat splenic sinus. Cell Tissue Res 2016; 364:611-622. [PMID: 26846226 DOI: 10.1007/s00441-015-2350-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/16/2015] [Indexed: 01/03/2023]
Abstract
The splenic sinus endothelium adhering via adherens junctions and tight junctions regulates the passage of blood cells through the splenic cord. Focal adhesion kinase (FAK) regulates the focal adhesion complex in the basal part of endothelial cells and is an integrated component of cell-cell adhesion, depending on its phosphorylation status. The objectives of this study are to assess the localization of FAK phosphorylated at tyrosine residues and the related proteins of integrin β5, talin, paxillin, p130Cas, vinculin, RhoA, Rac1, Rac2, Cdc42 and VE-cadherin, in the sinus endothelial cells of rat spleen and to examine the roles of FAK in regulating endothelial adhesion and the passage of blood cells. Immunofluorescence microscopy of tissue cryosections revealed that FAK was localized in the entire circumference of sinus endothelial cells and FAK phosphorylated at Try397 residue (pFAKy397) and pFAKy576 were precisely localized in the adherens junctions of the endothelial cells, whereas pFAKy925 was localized in the basal part of the endothelial cells. Paxillin and vinculin were prominently localized in the basal part of the endothelial cells. Integrin β5, talin and p130Cas were colocalized with FAK in the entire circumference of sinus endothelial cells. RhoA, Rac2 and Cdc42 were localized in the entire circumference of sinus endothelial cells close to FAK, stress fibers and cortical actin filaments. Immunogold electron microscopy revealed that pFAKy397 and pFAKy576 were colocalized with VE-cadherin, RhoA, Rac2 and Cdc42 in the adherens junctions of the endothelial cells. Possible functional roles of FAK in splenic sinus endothelial cells are also discussed.
Collapse
Affiliation(s)
- Kiyoko Uehara
- Department of Cell Biology, Fukuoka University School of Medicine, Jonan-ku, Fukuoka, 814-0180, Japan.
| | - Akira Uehara
- Department of Physiology, Fukuoka University School of Medicine, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
25
|
Golji J, Mofrad MRK. The talin dimer structure orientation is mechanically regulated. Biophys J 2015; 107:1802-1809. [PMID: 25418161 DOI: 10.1016/j.bpj.2014.08.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 06/30/2014] [Accepted: 08/27/2014] [Indexed: 01/09/2023] Open
Abstract
Formation of a stable cell-substrate contact can be regulated by mechanical force, especially at the focal adhesion. Individual proteins that make up the focal adhesions, such as talin, can exhibit mechanosensing. We previously described one mode of talin mechanosensing in which the vinculin-binding site of talin is exposed after force-induced stretch of a single talin rod domain. Here, we describe a second mode of talin mechanosensing in which the talin dimer itself can adopt different orientations in response to mechanical stimulation. Using molecular dynamics models, we demonstrate that the C-terminus region of the talin dimer is flexible mainly at the linker between the dimerization helices and the nearby actin-binding helical bundle. Our molecular dynamics simulations reveal two possible orientations of the talin dimer at its C-terminus. The extracellular matrix (ECM)-bound integrins cross-linked by talin can be forced apart leading to an elongated orientation of the talin dimer, and the ECM-bound integrins can be forced together by the ECM producing a collapsed orientation of the talin dimer. Formation of the elongated orientation is shown to be more favorable. Switching between the two talin dimer orientations constitutes a mode of mechanosensing.
Collapse
Affiliation(s)
- Javad Golji
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California; Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California.
| |
Collapse
|
26
|
Meller J, Rogozin IB, Poliakov E, Meller N, Bedanov-Pack M, Plow EF, Qin J, Podrez EA, Byzova TV. Emergence and subsequent functional specialization of kindlins during evolution of cell adhesiveness. Mol Biol Cell 2014; 26:786-96. [PMID: 25540429 PMCID: PMC4325847 DOI: 10.1091/mbc.e14-08-1294] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Kindlins are integrin-interacting proteins essential for integrin-mediated cell adhesiveness. In this study, we focused on the evolutionary origin and functional specialization of kindlins as a part of the evolutionary adaptation of cell adhesive machinery. Database searches revealed that many members of the integrin machinery (including talin and integrins) existed before kindlin emergence in evolution. Among the analyzed species, all metazoan lineages—but none of the premetazoans—had at least one kindlin-encoding gene, whereas talin was present in several premetazoan lineages. Kindlin appears to originate from a duplication of the sequence encoding the N-terminal fragment of talin (the talin head domain) with a subsequent insertion of the PH domain of separate origin. Sequence analysis identified a member of the actin filament-associated protein 1 (AFAP1) superfamily as the most likely origin of the kindlin PH domain. The functional divergence between kindlin paralogues was assessed using the sequence swap (chimera) approach. Comparison of kindlin 2 (K2)/kindlin 3 (K3) chimeras revealed that the F2 subdomain, in particular its C-terminal part, is crucial for the differential functional properties of K2 and K3. The presence of this segment enables K2 but not K3 to localize to focal adhesions. Sequence analysis of the C-terminal part of the F2 subdomain of K3 suggests that insertion of a variable glycine-rich sequence in vertebrates contributed to the loss of constitutive K3 targeting to focal adhesions. Thus emergence and subsequent functional specialization of kindlins allowed multicellular organisms to develop additional tissue-specific adaptations of cell adhesiveness.
Collapse
Affiliation(s)
- Julia Meller
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Igor B Rogozin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894
| | - Eugenia Poliakov
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Nahum Meller
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Mark Bedanov-Pack
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894
| | - Edward F Plow
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jun Qin
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Eugene A Podrez
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Tatiana V Byzova
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
27
|
Abstract
Biological mechano-transduction and force-dependent changes scale from protein conformation (â„« to nm) to cell organization and multi-cell function (mm to cm) to affect cell organization, fate, and homeostasis. External forces play complex roles in cell organization, fate, and homeostasis. Changes in these forces, or how cells respond to them, can result in abnormal embryonic development and diseases in adults. How cells sense and respond to these mechanical stimuli requires an understanding of the biophysical principles that underlie changes in protein conformation and result in alterations in the organization and function of cells and tissues. Here, we discuss mechano-transduction as it applies to protein conformation, cellular organization, and multi-cell (tissue) function.
Collapse
Affiliation(s)
- Beth L. Pruitt
- Department of Mechanical Engineering, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| | - Alexander R. Dunn
- Department of Chemical Engineering, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| | - William I. Weis
- Department of Structural Biology, Stanford University, Stanford, California, United States of America
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| | - W. James Nelson
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- Department of Biology, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| |
Collapse
|
28
|
Mechanical activation of vinculin binding to talin locks talin in an unfolded conformation. Sci Rep 2014; 4:4610. [PMID: 24714394 PMCID: PMC3980218 DOI: 10.1038/srep04610] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/21/2014] [Indexed: 11/08/2022] Open
Abstract
The force-dependent interaction between talin and vinculin plays a crucial role in the initiation and growth of focal adhesions. Here we use magnetic tweezers to characterise the mechano-sensitive compact N-terminal region of the talin rod, and show that the three helical bundles R1–R3 in this region unfold in three distinct steps consistent with the domains unfolding independently. Mechanical stretching of talin R1–R3 enhances its binding to vinculin and vinculin binding inhibits talin refolding after force is released. Mutations that stabilize R3 identify it as the initial mechano-sensing domain in talin, unfolding at ∼5 pN, suggesting that 5 pN is the force threshold for vinculin binding and adhesion progression.
Collapse
|
29
|
Hytönen VP, Wehrle-Haller B. Protein conformation as a regulator of cell–matrix adhesion. Phys Chem Chem Phys 2014; 16:6342-57. [DOI: 10.1039/c3cp54884h] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Conformational changes within proteins play key roles in the regulation of cell–matrix adhesion. We discuss the mechanisms involved in conformational regulation, including mechanical signals, posttranslational modifications and intrinsically disordered proteins.
Collapse
Affiliation(s)
- Vesa P. Hytönen
- University of Tampere
- Institute of Biomedical Technology and BioMediTech
- 33520 Tampere, Finland
- Fimlab Laboratories
- 33014 Tampere, Finland
| | - Bernhard Wehrle-Haller
- University of Geneva
- Department of Cell Physiology and Metabolism
- Centre Médical Universitaire
- 1211 Geneva 4, Switzerland
| |
Collapse
|
30
|
Gkourogianni A, Egot M, Koloka V, Moussis V, Tsikaris V, Panou-Pomonis E, Sakarellos-Daitsiotis M, Bachelot-Loza C, Tsoukatos DC. Palmitoylated peptide, being derived from the carboxyl-terminal sequence of the integrin αIIbcytoplasmic domain, inhibits talin binding to αIIbβ3. Platelets 2013; 25:619-27. [DOI: 10.3109/09537104.2013.850588] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
31
|
Wang Y, Wang C, Hou Z, Miao K, Zhao H, Wang R, Guo M, Wu Z, Tian J, An L. Comparative analysis of proteomic profiles between endometrial caruncular and intercaruncular areas in ewes during the peri-implantation period. J Anim Sci Biotechnol 2013; 4:39. [PMID: 24093944 PMCID: PMC3892124 DOI: 10.1186/2049-1891-4-39] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 09/23/2013] [Indexed: 01/24/2023] Open
Abstract
The endometrium of sheep consists of plenty of raised aglandular areas called caruncular (C), and intensely glandular intercaruncular areas (IC). In order to better understand the endometrium involved mechanisms of implantation, we used LC-MS/MS technique to profile the proteome of ovine endometrial C areas and IC areas separately during the peri-implantation period, and then compared the proteomic profiles between these two areas. We successfully detected 1740 and 1813 proteins in C areas and IC areas respectively. By comparing the proteome of these two areas, we found 170 differentially expressed proteins (DEPs) (P < 0.05), functional bioinformatics analysis showed these DEPs were mainly involved in growth and remodeling of endometrial tissue, cell adhesion and protein transport, and so on. Our study, for the first time, provided a proteomic reference for elucidating the differences between C and IC areas, as an integrated function unit respectively, during the peri-implantation period. The results could help us to better understand the implantation in the ewes. In addition, we established a relatively detailed protein database of ovine endometrium, which provide a unique reference for further studies.
Collapse
Affiliation(s)
- Yang Wang
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Chao Wang
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Zhuocheng Hou
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, China Agricultural University, Beijing 100193, China
| | - Kai Miao
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Haichao Zhao
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Rui Wang
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Min Guo
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Zhonghong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Jianhui Tian
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| | - Lei An
- Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National engineering laboratory for animal breeding, College of Animal Sciences and Technology, China Agricultural University, No.2 Yuanmingyuan Xi Lu, Haidian, Beijing 100193, China
| |
Collapse
|
32
|
Goult BT, Zacharchenko T, Bate N, Tsang R, Hey F, Gingras AR, Elliott PR, Roberts GCK, Ballestrem C, Critchley DR, Barsukov IL. RIAM and vinculin binding to talin are mutually exclusive and regulate adhesion assembly and turnover. J Biol Chem 2013; 288:8238-8249. [PMID: 23389036 PMCID: PMC3605642 DOI: 10.1074/jbc.m112.438119] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/20/2013] [Indexed: 12/02/2022] Open
Abstract
Talin activates integrins, couples them to F-actin, and recruits vinculin to focal adhesions (FAs). Here, we report the structural characterization of the talin rod: 13 helical bundles (R1-R13) organized into a compact cluster of four-helix bundles (R2-R4) within a linear chain of five-helix bundles. Nine of the bundles contain vinculin-binding sites (VBS); R2R3 are atypical, with each containing two VBS. Talin R2R3 also binds synergistically to RIAM, a Rap1 effector involved in integrin activation. Biochemical and structural data show that vinculin and RIAM binding to R2R3 is mutually exclusive. Moreover, vinculin binding requires domain unfolding, whereas RIAM binds the folded R2R3 double domain. In cells, RIAM is enriched in nascent adhesions at the leading edge whereas vinculin is enriched in FAs. We propose a model in which RIAM binding to R2R3 initially recruits talin to membranes where it activates integrins. As talin engages F-actin, force exerted on R2R3 disrupts RIAM binding and exposes the VBS, which recruit vinculin to stabilize the complex.
Collapse
Affiliation(s)
- Benjamin T Goult
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Thomas Zacharchenko
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - Neil Bate
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Ricky Tsang
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Fiona Hey
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Alexandre R Gingras
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Paul R Elliott
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - Gordon C K Roberts
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - David R Critchley
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom.
| | - Igor L Barsukov
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, 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
|
The leucocyte β2 (CD18) integrins: the structure, functional regulation and signalling properties. Biosci Rep 2012; 32:241-69. [PMID: 22458844 DOI: 10.1042/bsr20110101] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Leucocytes are highly motile cells. Their ability to migrate into tissues and organs is dependent on cell adhesion molecules. The integrins are a family of heterodimeric transmembrane cell adhesion molecules that are also signalling receptors. They are involved in many biological processes, including the development of metazoans, immunity, haemostasis, wound healing and cell survival, proliferation and differentiation. The leucocyte-restricted β2 integrins comprise four members, namely αLβ2, αMβ2, αXβ2 and αDβ2, which are required for a functional immune system. In this paper, the structure, functional regulation and signalling properties of these integrins are reviewed.
Collapse
|
35
|
Kahner BN, Kato H, Banno A, Ginsberg MH, Shattil SJ, Ye F. Kindlins, integrin activation and the regulation of talin recruitment to αIIbβ3. PLoS One 2012; 7:e34056. [PMID: 22457811 PMCID: PMC3311585 DOI: 10.1371/journal.pone.0034056] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 02/21/2012] [Indexed: 11/18/2022] Open
Abstract
Talins and kindlins bind to the integrin β3 cytoplasmic tail and both are required for effective activation of integrin αIIbβ3 and resulting high-affinity ligand binding in platelets. However, binding of the talin head domain alone to β3 is sufficient to activate purified integrin αIIbβ3 in vitro. Since talin is localized to the cytoplasm of unstimulated platelets, its re-localization to the plasma membrane and to the integrin is required for activation. Here we explored the mechanism whereby kindlins function as integrin co-activators. To test whether kindlins regulate talin recruitment to plasma membranes and to αIIbβ3, full-length talin and kindlin recruitment to β3 was studied using a reconstructed CHO cell model system that recapitulates agonist-induced αIIbβ3 activation. Over-expression of kindlin-2, the endogenous kindlin isoform in CHO cells, promoted PAR1-mediated and talin-dependent ligand binding. In contrast, shRNA knockdown of kindlin-2 inhibited ligand binding. However, depletion of kindlin-2 by shRNA did not affect talin recruitment to the plasma membrane, as assessed by sub-cellular fractionation, and neither over-expression of kindlins nor depletion of kindlin-2 affected talin interaction with αIIbβ3 in living cells, as monitored by bimolecular fluorescence complementation. Furthermore, talin failed to promote kindlin-2 association with αIIbβ3 in CHO cells. In addition, purified talin and kindlin-3, the kindlin isoform expressed in platelets, failed to promote each other's binding to the β3 cytoplasmic tail in vitro. Thus, kindlins do not promote initial talin recruitment to αIIbβ3, suggesting that they co-activate integrin through a mechanism independent of recruitment.
Collapse
Affiliation(s)
- Bryan N Kahner
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | | | | | | | | |
Collapse
|
36
|
Sandri-Goldin RM. The many roles of the highly interactive HSV protein ICP27, a key regulator of infection. Future Microbiol 2012; 6:1261-77. [PMID: 22082288 DOI: 10.2217/fmb.11.119] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Human herpes viruses cause an array of illnesses ranging from cancers for Epstein?Barr virus and Kaposi?s sarcoma-associated herpes virus, to painful skin lesions, and more rarely, keratitis and encephalitis for HSV. All herpes viruses encode a multifunctional protein, typified by HSV ICP27, which plays essential roles in viral infection. ICP27 functions in all stages of mRNA biogenesis from transcription, RNA processing and export through to translation. ICP27 has also been implicated in nuclear protein quality control, cell cycle control, activation of stress signaling pathways and prevention of apoptosis. ICP27 interacts with many proteins and it binds RNA. This article focuses on how ICP27 performs its many roles and highlights similarities with its homologs, which could be targets for antiviral intervention.
Collapse
Affiliation(s)
- Rozanne M Sandri-Goldin
- Department of Microbiology & Molecular Genetics, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
37
|
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
|
38
|
Whitman SA, Cover C, Yu L, Nelson DL, Zarnescu DC, Gregorio CC. Desmoplakin and talin2 are novel mRNA targets of fragile X-related protein-1 in cardiac muscle. Circ Res 2011; 109:262-71. [PMID: 21659647 PMCID: PMC3163600 DOI: 10.1161/circresaha.111.244244] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 06/02/2011] [Indexed: 01/31/2023]
Abstract
RATIONALE The proper function of cardiac muscle requires the precise assembly and interactions of numerous cytoskeletal and regulatory proteins into specialized structures that orchestrate contraction and force transmission. Evidence suggests that posttranscriptional regulation is critical for muscle function, but the mechanisms involved remain understudied. OBJECTIVE To investigate the molecular mechanisms and targets of the muscle-specific fragile X mental retardation, autosomal homolog 1 (FXR1), an RNA binding protein whose loss leads to perinatal lethality in mice and cardiomyopathy in zebrafish. METHODS AND RESULTS Using RNA immunoprecipitation approaches we found that desmoplakin and talin2 mRNAs associate with FXR1 in a complex. In vitro assays indicate that FXR1 binds these mRNA targets directly and represses their translation. Fxr1 KO hearts exhibit an up-regulation of desmoplakin and talin2 proteins, which is accompanied by severe disruption of desmosome as well as costamere architecture and composition in the heart, as determined by electron microscopy and deconvolution immunofluorescence analysis. CONCLUSIONS Our findings reveal the first direct mRNA targets of FXR1 in striated muscle and support translational repression as a novel mechanism for regulating heart muscle development and function, in particular the assembly of specialized cytoskeletal structures.
Collapse
Affiliation(s)
- Samantha A. Whitman
- Department of Cellular and Molecular Medicine and the Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona 85724
| | - Cathleen Cover
- Department of Cellular and Molecular Medicine and the Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona 85724
| | - Lily Yu
- Department of Cellular and Molecular Medicine and the Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona 85724
| | - David L. Nelson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Daniela C. Zarnescu
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona 85721
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and the Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona 85724
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona 85721
| |
Collapse
|
39
|
Kim HR, Liu K, Roberts TJ, Hai CM. Length-dependent modulation of cytoskeletal remodeling and mechanical energetics in airway smooth muscle. Am J Respir Cell Mol Biol 2011; 44:888-97. [PMID: 20705939 PMCID: PMC3135848 DOI: 10.1165/rcmb.2010-0144oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Accepted: 07/11/2010] [Indexed: 12/12/2022] Open
Abstract
Actin cytoskeletal remodeling is an important mechanism of airway smooth muscle (ASM) contraction. We tested the hypothesis that mechanical strain modulates the cholinergic receptor-mediated cytoskeletal recruitment of actin-binding and integrin-binding proteins in intact airway smooth muscle, thereby regulating the mechanical energetics of airway smooth muscle. We found that the carbachol-stimulated cytoskeletal recruitment of actin-related protein-3 (Arp3), metavinculin, and talin were up-regulated at short muscle lengths and down-regulated at long muscle lengths, suggesting that the actin cytoskeleton--integrin complex becomes enriched in cross-linked and branched actin filaments in shortened ASM. The mechanical energy output/input ratio during sinusoidal length oscillation was dependent on muscle length, oscillatory amplitude, and cholinergic activation. The enhancing effect of cholinergic stimulation on mechanical energy output/input ratio at short and long muscle lengths may be explained by the length-dependent modulation of cytoskeletal recruitment and crossbridge cycling, respectively. We postulate that ASM functions as a hybrid biomaterial, capable of switching between operating as a cytoskeleton-based mechanical energy store at short muscle lengths to operating as an actomyosin-powered mechanical energy generator at long muscle lengths. This postulate predicts that targeting the signaling molecules involved in cytoskeletal recruitment may provide a novel approach to dilating collapsed airways in obstructive airway disease.
Collapse
Affiliation(s)
- Hak Rim Kim
- Department of Molecular Pharmacology, Physiology, and Biotechnology, and Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island; and Department of Pharmacology, College of Medicine, Dankook University, Cheonan-si, Chungnam, Republic of Korea
| | - Katrina Liu
- Department of Molecular Pharmacology, Physiology, and Biotechnology, and Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island; and Department of Pharmacology, College of Medicine, Dankook University, Cheonan-si, Chungnam, Republic of Korea
| | - Thomas J. Roberts
- Department of Molecular Pharmacology, Physiology, and Biotechnology, and Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island; and Department of Pharmacology, College of Medicine, Dankook University, Cheonan-si, Chungnam, Republic of Korea
| | - Chi-Ming Hai
- Department of Molecular Pharmacology, Physiology, and Biotechnology, and Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island; and Department of Pharmacology, College of Medicine, Dankook University, Cheonan-si, Chungnam, Republic of Korea
| |
Collapse
|
40
|
A proteomic and cellular analysis of uropods in the pathogen Entamoeba histolytica. PLoS Negl Trop Dis 2011; 5:e1002. [PMID: 21483708 PMCID: PMC3071361 DOI: 10.1371/journal.pntd.0001002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 03/07/2011] [Indexed: 11/19/2022] Open
Abstract
Exposure of Entamoeba histolytica to specific ligands induces cell polarization via the activation of signalling pathways and cytoskeletal elements. The process leads to formation of a protruding pseudopod at the front of the cell and a retracting uropod at the rear. In the present study, we show that the uropod forms during the exposure of trophozoites to serum isolated from humans suffering of amoebiasis. To investigate uropod assembly, we used LC-MS/MS technology to identify protein components in isolated uropod fractions. The galactose/N-acetylgalactosamine lectin, the immunodominant antigen M17 (which is specifically recognized by serum from amoeba-infected persons) and a few other cells adhesion-related molecules were primarily involved. Actin-rich cytoskeleton components, GTPases from the Rac and Rab families, filamin, α-actinin and a newly identified ezrin-moesin-radixin protein were the main factors found to potentially interact with capped receptors. A set of specific cysteine proteases and a serine protease were enriched in isolated uropod fractions. However, biological assays indicated that cysteine proteases are not involved in uropod formation in E. histolytica, a fact in contrast to the situation in human motile immune cells. The surface proteins identified here are testable biomarkers which may be either recognized by the immune system and/or released into the circulation during amoebiasis.
Collapse
|
41
|
Kotecki M, Zeiger AS, Van Vliet K, Herman IM. Calpain- and talin-dependent control of microvascular pericyte contractility and cellular stiffness. Microvasc Res 2010; 80:339-48. [PMID: 20709086 PMCID: PMC2981705 DOI: 10.1016/j.mvr.2010.07.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 07/26/2010] [Accepted: 07/30/2010] [Indexed: 01/09/2023]
Abstract
Pericytes surround capillary endothelial cells and exert contractile forces modulating microvascular tone and endothelial growth. We previously described pericyte contractile phenotype to be Rho GTPase- and α-smooth muscle actin (αSMA)-dependent. However, mechanisms mediating adhesion-dependent shape changes and contractile force transduction remain largely equivocal. We now report that the neutral cysteine protease, calpain, modulates pericyte contractility and cellular stiffness via talin, an integrin-binding and F-actin associating protein. Digital imaging and quantitative analyses of living cells reveal significant perturbations in contractile force transduction detected via deformation of silicone substrata, as well as perturbations of mechanical stiffness in cellular contractile subdomains quantified via atomic force microscope (AFM)-enabled nanoindentation. Pericytes overexpressing GFP-tagged talin show significantly enhanced contractility (~two-fold), which is mitigated when either the calpain-cleavage resistant mutant talin L432G or vinculin are expressed. Moreover, the cell-penetrating, calpain-specific inhibitor termed CALPASTAT reverses talin-enhanced, but not Rho GTP-dependent, contractility. Interestingly, our analysis revealed that CALPASTAT, but not its inactive mutant, alters contractile cell-driven substrata deformations while increasing mechanical stiffness of subcellular contractile regions of these pericytes. Altogether, our results reveal that calpain-dependent cleavage of talin modulates cell contractile dynamics, which in pericytes may prove instrumental in controlling normal capillary function or microvascular pathophysiology.
Collapse
Affiliation(s)
- Maciej Kotecki
- Department of Physiology, and The Center for Innovations in Wound Healing Research, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111 USA
| | - Adam S. Zeiger
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
| | - Krystyn Van Vliet
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 USA
| | - Ira M. Herman
- Department of Physiology, and The Center for Innovations in Wound Healing Research, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111 USA
| |
Collapse
|
42
|
Head-to-tail intramolecular interaction of herpes simplex virus type 1 regulatory protein ICP27 is important for its interaction with cellular mRNA export receptor TAP/NXF1. mBio 2010; 1. [PMID: 21060739 PMCID: PMC2975367 DOI: 10.1128/mbio.00268-10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 10/15/2010] [Indexed: 01/07/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) protein ICP27 has many important functions during infection that are achieved through interactions with a number of cellular proteins. In its role as a viral RNA export protein, ICP27 interacts with TAP/NXF1, the cellular mRNA export receptor, and both the N and C termini of ICP27 must be intact for this interaction to take place. Here we show by bimolecular fluorescence complementation (BiFC) that ICP27 interacts directly with TAP/NXF1 during infection, and this interaction failed to occur with an ICP27 mutant bearing substitutions of serines for cysteines at positions 483 and 488 in the C-terminal zinc finger. Recently, we showed that ICP27 undergoes a head-to-tail intramolecular interaction, which could make the N- and C-terminal regions accessible for binding to TAP/NXF1. To determine the importance of intramolecular association of ICP27 to its interaction with TAP/NXF1, we performed BiFC-based fluorescence resonance energy transfer (FRET) by acceptor photobleaching. BiFC-based FRET showed that the interaction between ICP27 and TAP/NXF1 occurred in living cells upon head-to-tail intramolecular association of ICP27, further establishing that TAP/NXF1 interacts with both the N and C termini of ICP27. ICP27 is a key regulatory protein during herpes simplex virus type 1 (HSV-1) infection. ICP27 interacts with a number of cellular proteins, and an important question asks how these interactions are regulated during infection. We showed previously that ICP27 undergoes a head-to-tail intramolecular interaction, and here we show that the cellular mRNA export receptor protein TAP/NXF1 interacts with ICP27 after its head-to-tail association. Several proteins that interact with ICP27 require that the N and C termini of ICP27 be intact. These results demonstrate that the head-to-tail interaction of ICP27 may regulate some of its protein interactions perhaps through alternating between open and closed configurations.
Collapse
|
43
|
Elliott PR, Goult BT, Kopp PM, Bate N, Grossmann JG, Roberts GC, Critchley DR, Barsukov IL. The Structure of the talin head reveals a novel extended conformation of the FERM domain. Structure 2010; 18:1289-99. [PMID: 20947018 PMCID: PMC2977851 DOI: 10.1016/j.str.2010.07.011] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 07/16/2010] [Accepted: 07/18/2010] [Indexed: 11/29/2022]
Abstract
FERM domains are found in a diverse superfamily of signaling and adaptor proteins at membrane interfaces. They typically consist of three separately folded domains (F1, F2, F3) in a compact cloverleaf structure. The crystal structure of the N-terminal head of the integrin-associated cytoskeletal protein talin reported here reveals a novel FERM domain with a linear domain arrangement, plus an additional domain F0 packed against F1. While F3 binds β-integrin tails, basic residues in F1 and F2 are required for membrane association and for integrin activation. We show that these same residues are also required for cell spreading and focal adhesion assembly in cells. We suggest that the extended conformation of the talin head allows simultaneous binding to integrins via F3 and to PtdIns(4,5)P2-enriched microdomains via basic residues distributed along one surface of the talin head, and that these multiple interactions are required to stabilize integrins in the activated state.
Collapse
Affiliation(s)
- Paul R. Elliott
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Benjamin T. Goult
- Department of Biochemistry, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Petra M. Kopp
- Department of Biochemistry, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Neil Bate
- Department of Biochemistry, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - J. Günter Grossmann
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Gordon C.K. Roberts
- Department of Biochemistry, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - David R. Critchley
- Department of Biochemistry, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 9HN, UK
| | - Igor L. Barsukov
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| |
Collapse
|
44
|
Mitsios JV, Prévost N, Kasirer-Friede A, Gutierrez E, Groisman A, Abrams CS, Wang Y, Litvinov RI, Zemljic-Harpf A, Ross RS, Shattil SJ. What is vinculin needed for in platelets? J Thromb Haemost 2010; 8:2294-304. [PMID: 20670372 PMCID: PMC2965783 DOI: 10.1111/j.1538-7836.2010.03998.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
UNLABELLED Summary. BACKGROUND Vinculin links integrins to the cell cytoskeleton by virtue of its binding to proteins such as talin and F-actin. It has been implicated in the transmission of mechanical forces from the extracellular matrix to the cytoskeleton of migrating cells. Vinculin's function in platelets is unknown. OBJECTIVE To determine whether vinculin is required for the functions of platelets and their major integrin, α(IIb) β(3) . METHODS The murine vinculin gene (Vcl) was deleted in the megakaryocyte/platelet lineage by breeding Vcl fl/fl mice with Pf4-Cre mice. Platelet and integrin functions were studied in vivo and ex vivo. RESULTS Vinculin was undetectable in platelets from Vcl fl/fl Cre(+) mice, as determined by immunoblotting and fluorescence microscopy. Vinculin-deficient megakaryocytes exhibited increased membrane tethers in response to mechanical pulling on α(IIb) β(3) with laser tweezers, suggesting that vinculin helps to maintain membrane cytoskeleton integrity. Surprisingly, vinculin-deficient platelets displayed normal agonist-induced fibrinogen binding to α(IIb) β(3) , aggregation, spreading, actin polymerization/organization, clot retraction and the ability to form a procoagulant surface. Furthermore, vinculin-deficient platelets adhered to immobilized fibrinogen or collagen normally, under both static and flow conditions. Tail bleeding times were prolonged in 59% of vinculin-deficient mice. However, these mice exhibited no spontaneous bleeding and they formed occlusive platelet thrombi comparable to those in wild-type littermates in response to carotid artery injury with FeCl(3) . CONCLUSION Despite promoting membrane cytoskeleton integrity when mechanical force is applied to α(IIb) β(3) , vinculin is not required for the traditional functions of α(IIb) β(3) or the platelet actin cytoskeleton.
Collapse
Affiliation(s)
- John V. Mitsios
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Nicolas Prévost
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ana Kasirer-Friede
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Edgar Gutierrez
- Department of Physics, University of California, San Diego, La Jolla, CA 92093
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, CA 92093
| | - Charles S. Abrams
- Department of Medicine and Cell, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Yanfeng Wang
- Department of Medicine and Cell, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Rustem I. Litvinov
- Department of Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Alice Zemljic-Harpf
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- VA Healthcare San Diego, San Diego, CA 92161
| | - Robert S. Ross
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- VA Healthcare San Diego, San Diego, CA 92161
| | - Sanford J. Shattil
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
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
|
|
47
|
Goult BT, Gingras AR, Bate N, Barsukov IL, Critchley DR, Roberts GC. The domain structure of talin: residues 1815-1973 form a five-helix bundle containing a cryptic vinculin-binding site. FEBS Lett 2010; 584:2237-41. [PMID: 20399778 PMCID: PMC2887493 DOI: 10.1016/j.febslet.2010.04.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 03/24/2010] [Accepted: 04/08/2010] [Indexed: 11/24/2022]
Abstract
Talin is a large flexible rod-shaped protein that activates the integrin family of cell adhesion molecules and couples them to cytoskeletal actin. Its rod region consists of a series of helical bundles. Here we show that residues 1815-1973 form a 5-helix bundle, with a topology unique to talin which is optimally suited for formation of a long rod such as talin. This is much more stable than the 4-helix (1843-1973) domain described earlier and as a result its vinculin binding sequence is inaccessible to vinculin at room temperature, with implications for the overall mechanism of the talin-vinculin interaction.
Collapse
Affiliation(s)
- Benjamin T. Goult
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester, LE1 9HN, UK
| | - Alexandre R. Gingras
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester, LE1 9HN, UK
| | - Neil Bate
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester, LE1 9HN, UK
| | - Igor L. Barsukov
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - David R. Critchley
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester, LE1 9HN, UK
| | - Gordon C.K. Roberts
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester, LE1 9HN, UK
| |
Collapse
|
48
|
Goult BT, Bouaouina M, Elliott PR, Bate N, Patel B, Gingras AR, Grossmann JG, Roberts GCK, Calderwood DA, Critchley DR, Barsukov IL. Structure of a double ubiquitin-like domain in the talin head: a role in integrin activation. EMBO J 2010; 29:1069-80. [PMID: 20150896 PMCID: PMC2845276 DOI: 10.1038/emboj.2010.4] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 01/11/2010] [Indexed: 01/13/2023] Open
Abstract
Talin is a 270-kDa protein that activates integrins and couples them to cytoskeletal actin. Talin contains an N-terminal FERM domain comprised of F1, F2 and F3 domains, but it is atypical in that F1 contains a large insert and is preceded by an extra domain F0. Although F3 contains the binding site for beta-integrin tails, F0 and F1 are also required for activation of beta1-integrins. Here, we report the solution structures of F0, F1 and of the F0F1 double domain. Both F0 and F1 have ubiquitin-like folds joined in a novel fixed orientation by an extensive charged interface. The F1 insert forms a loop with helical propensity, and basic residues predicted to reside on one surface of the helix are required for binding to acidic phospholipids and for talin-mediated activation of beta1-integrins. This and the fact that basic residues on F2 and F3 are also essential for integrin activation suggest that extensive interactions between the talin FERM domain and acidic membrane phospholipids are required to orientate the FERM domain such that it can activate integrins.
Collapse
Affiliation(s)
- Benjamin T Goult
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Mohamed Bouaouina
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | - Paul R Elliott
- School of Biological Sciences, University of Liverpool, Liverpool, UK
| | - Neil Bate
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Bipin Patel
- Department of Biochemistry, University of Leicester, Leicester, UK
| | | | | | | | - David A Calderwood
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, CT, USA
| | | | - Igor L Barsukov
- School of Biological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
49
|
Herpes simplex virus 1 regulatory protein ICP27 undergoes a head-to-tail intramolecular interaction. J Virol 2010; 84:4124-35. [PMID: 20164236 DOI: 10.1128/jvi.02319-09] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) regulatory protein ICP27 is a multifunction functional protein that interacts with many cellular proteins. A number of the proteins with which ICP27 interacts require that both the N and C termini of ICP27 are intact. These include RNA polymerase II, TAP/NXF1, and Hsc70. We tested the possibility that the N and C termini of ICP27 could undergo a head-to-tail intramolecular interaction that exists in open and closed configurations for different binding partners. Here, we show by bimolecular fluorescence complementation (BiFC) assays and fluorescence resonance energy transfer (FRET) by acceptor photobleaching that ICP27 undergoes a head-to-tail intramolecular interaction but not head-to-tail or tail-to-tail intermolecular interactions. Substitution mutations in the N or C termini showed that the leucine-rich region (LRR) in the N terminus and the zinc finger-like region in the C terminus must be intact for intramolecular interactions. A recombinant virus, vNC-Venus-ICP27, was constructed, and this virus was severely impaired for virus replication. The expression of NC-Venus-ICP27 protein was delayed compared to ICP27 expression in wild-type HSV-1 infection, but NC-Venus-ICP27 was abundantly expressed at late times of infection. Because the renaturation of the Venus fluorescent protein results in a covalent bonding of the two halves of the Venus molecule, the head-to-tail interaction of NC-Venus-ICP27 locks ICP27 in a closed configuration. We suggest that the population of locked ICP27 molecules is not able to undergo further protein-protein interactions.
Collapse
|
50
|
Domadia PN, Li YF, Bhunia A, Mohanram H, Tan SM, Bhattacharjya S. Functional and structural characterization of the talin F0F1 domain. Biochem Biophys Res Commun 2009; 391:159-65. [PMID: 19903453 DOI: 10.1016/j.bbrc.2009.11.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 11/05/2009] [Indexed: 10/20/2022]
Abstract
The globular head domain of talin, a large multi-domain cytoplasmic protein, is required for inside-out activation of the integrins, a family of heterodimeric transmembrane cell adhesion molecules. Talin head contains a FERM domain that is composed of F1, F2, and F3 subdomains. A F0 subdomain is located N-terminus to F1. The F3 contains a canonical phosphotyrosine binding (PTB) fold that directly interacts with the membrane proximal NPxY/F motif in the integrin beta cytoplasmic tail. This interaction is stabilized by the F2 that interacts with the lipid head-groups of the plasma membrane. In comparison to F2 and F3, the properties of the F0F1 remains poorly characterized. Here, we showed that F0F1 is essential for talin-induced activation of integrin alphaLbeta2 (LFA-1). F0F1 has a high content of beta-sheet secondary structure, and it tends to homodimerize that may provide stability against proteolysis and chaotrope induced unfolding.
Collapse
Affiliation(s)
- Prerna N Domadia
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
| | | | | | | | | | | |
Collapse
|