1
|
Thomasy SM, Leonard BC, Greiner MA, Skeie JM, Raghunathan VK. Squishy matters - Corneal mechanobiology in health and disease. Prog Retin Eye Res 2024; 99:101234. [PMID: 38176611 PMCID: PMC11193890 DOI: 10.1016/j.preteyeres.2023.101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
The cornea, as a dynamic and responsive tissue, constantly interacts with mechanical forces in order to maintain its structural integrity, barrier function, transparency and refractive power. Cells within the cornea sense and respond to various mechanical forces that fundamentally regulate their morphology and fate in development, homeostasis and pathophysiology. Corneal cells also dynamically regulate their extracellular matrix (ECM) with ensuing cell-ECM crosstalk as the matrix serves as a dynamic signaling reservoir providing biophysical and biochemical cues to corneal cells. Here we provide an overview of mechanotransduction signaling pathways then delve into the recent advances in corneal mechanobiology, focusing on the interplay between mechanical forces and responses of the corneal epithelial, stromal, and endothelial cells. We also identify species-specific differences in corneal biomechanics and mechanotransduction to facilitate identification of optimal animal models to study corneal wound healing, disease, and novel therapeutic interventions. Finally, we identify key knowledge gaps and therapeutic opportunities in corneal mechanobiology that are pressing for the research community to address especially pertinent within the domains of limbal stem cell deficiency, keratoconus and Fuchs' endothelial corneal dystrophy. By furthering our understanding corneal mechanobiology, we can contextualize discoveries regarding corneal diseases as well as innovative treatments for them.
Collapse
Affiliation(s)
- Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States; California National Primate Research Center, Davis, CA, United States.
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States
| | - Mark A Greiner
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | - Jessica M Skeie
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | | |
Collapse
|
2
|
Protein dynamics at invadopodia control invasion-migration transitions in melanoma cells. Cell Death Dis 2023; 14:190. [PMID: 36899008 PMCID: PMC10006204 DOI: 10.1038/s41419-023-05704-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/12/2023]
Abstract
Cell invasion is a highly complex process that requires the coordination of cell migration and degradation of the extracellular matrix. In melanoma cells, as in many highly invasive cancer cell types these processes are driven by the regulated formation of adhesives structures such as focal adhesions and invasive structures like invadopodia. Structurally, focal adhesion and invadopodia are quite distinct, yet they share many protein constituents. However, quantitative understanding of the interaction of invadopodia with focal adhesion is lacking, and how invadopodia turn-over is associated with invasion-migration transition cycles remains unknown. In this study, we investigated the role of Pyk2, cortactin and Tks5 in invadopodia turnover and their relation with focal adhesions. We found that active Pyk2 and cortactin are localised at both focal adhesions and invadopodia. At invadopodia, localisation of active Pyk2 is correlated with ECM degradation. During invadopodia disassembly, Pyk2 and cortactin but not Tks5 are often relocated at nearby nascent adhesions. We also show that during ECM degradation, cell migration is reduced which is likely related to the sharing of common molecules within the two structures. Finally, we found that the dual FAK/Pyk2 inhibitor PF-431396 inhibits both focal adhesion and invadopodia activities thereby reducing both migration and ECM degradation.
Collapse
|
3
|
Chen XR, Igumenova TI. Regulation of eukaryotic protein kinases by Pin1, a peptidyl-prolyl isomerase. Adv Biol Regul 2023; 87:100938. [PMID: 36496344 PMCID: PMC9992314 DOI: 10.1016/j.jbior.2022.100938] [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: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
The peptidyl-prolyl isomerase Pin1 cooperates with proline-directed kinases and phosphatases to regulate multiple oncogenic pathways. Pin1 specifically recognizes phosphorylated Ser/Thr-Pro motifs in proteins and catalyzes their cis-trans isomerization. The Pin1-catalyzed conformational changes determine the stability, activity, and subcellular localization of numerous protein substrates. We conducted a survey of eukaryotic protein kinases that are regulated by Pin1 and whose Pin1 binding sites have been identified. Our analyses reveal that Pin1 target sites in kinases do not fall exclusively within the intrinsically disordered regions of these enzymes. Rather, they fall into three groups based on their location: (i) within the catalytic kinase domain, (ii) in the C-terminal kinase region, and (iii) in regulatory domains. Some of the kinases downregulated by Pin1 activity are tumor-suppressing, and all kinases upregulated by Pin1 activity are functionally pro-oncogenic. These findings further reinforce the rationale for developing Pin1-specific inhibitors as attractive pharmaceuticals for cancer therapy.
Collapse
Affiliation(s)
- Xiao-Ru Chen
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Tatyana I Igumenova
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
4
|
TNS1: Emerging Insights into Its Domain Function, Biological Roles, and Tumors. BIOLOGY 2022; 11:biology11111571. [PMID: 36358270 PMCID: PMC9687257 DOI: 10.3390/biology11111571] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 01/25/2023]
Abstract
Tensins are a family of cellular-adhesion constituents that have been extensively studied. They have instrumental roles in the pathogenesis of numerous diseases. The mammalian tensin family comprises four members: tensin1 (TNS1), tensin2, tensin3, and tensin4. Among them, TNS1 has recently received attention from researchers because of its structural properties. TNS1 engages in various biological processes, such as cell adhesion, polarization, migration, invasion, proliferation, apoptosis, and mechano-transduction, by interacting with various partner proteins. Moreover, the abnormal expression of TNS1 in vivo is associated with the development of various diseases, especially tumors. Interestingly, the role of TNS1 in different tumors is still controversial. Here, we systematically summarize three aspects of TNS1: the gene structure, the biological processes underlying its action, and the dual regulatory role of TNS1 in different tumors through different mechanisms, of which we provide the first overview.
Collapse
|
5
|
Merryweather D, Moxon SR, Capel AJ, Hooper NM, Lewis MP, Roach P. Impact of type-1 collagen hydrogel density on integrin-linked morphogenic response of SH-SY5Y neuronal cells. RSC Adv 2021; 11:33124-33135. [PMID: 35493559 PMCID: PMC9042137 DOI: 10.1039/d1ra05257h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022] Open
Abstract
Cellular metabolism and behaviour is closely linked to cytoskeletal tension and scaffold mechanics. In the developing nervous system functional connectivity is controlled by the interplay between chemical and mechanical cues that initiate programs of cell behaviour. Replication of functional connectivity in neuronal populations in vitro has proven a technical challenge due to the absence of many systems of biomechanical regulation that control directional outgrowth in vivo. Here, a 3D culture system is explored by dilution of a type I collagen hydrogel to produce variation in gel stiffness. Hydrogel scaffold remodelling was found to be linked to gel collagen concentration, with a greater degree of gel contraction occurring at lower concentrations. Gel mechanics were found to evolve over the culture period according to collagen concentration. Less concentrated gels reduced in stiffness, whilst a biphasic pattern of increasing and then decreasing stiffness was observed at higher concentrations. Analysis of these cultures by PCR revealed a program of shifting integrin expression and highly variable activity in key morphogenic signal pathways, such as mitogen-associated protein kinase, indicating genetic impact of biomaterial interactions via mechano-regulation. Gel contraction at lower concentrations was also found to be accompanied by an increase in average collagen fibre diameter. Minor changes in biomaterial mechanics result in significant changes in programmed cell behaviour, resulting in adoption of markedly different cell morphologies and ability to remodel the scaffold. Advanced understanding of cell-biomaterial interactions, over short and long-term culture, is of critical importance in the development of novel tissue engineering strategies for the fabrication of biomimetic 3D neuro-tissue constructs. Simple methods of tailoring the initial mechanical environment presented to SH-SY5Y populations in 3D can lead to significantly different programs of network development over time.
Collapse
Affiliation(s)
- D Merryweather
- Department of Chemistry, Loughborough University Leicestershire LE11 3TU UK
| | - S R Moxon
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre Manchester M13 9PL UK
| | - A J Capel
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University Leicestershire LE11 3TU UK
| | - N M Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre Manchester M13 9PL UK
| | - M P Lewis
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University Leicestershire LE11 3TU UK
| | - P Roach
- Department of Chemistry, Loughborough University Leicestershire LE11 3TU UK
| |
Collapse
|
6
|
Michael E, Polydorides S, Promponas VJ, Skourides P, Archontis G. Recognition of LD motifs by the focal adhesion targeting domains of focal adhesion kinase and proline-rich tyrosine kinase 2-beta: Insights from molecular dynamics simulations. Proteins 2020; 89:29-52. [PMID: 32776636 DOI: 10.1002/prot.25992] [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: 04/16/2020] [Revised: 06/21/2020] [Accepted: 07/26/2020] [Indexed: 12/13/2022]
Abstract
The focal adhesion kinase (FAK) and the proline-rich tyrosine kinase 2-beta (PYK2) are implicated in cancer progression and metastasis and represent promising biomarkers and targets for cancer therapy. FAK and PYK2 are recruited to focal adhesions (FAs) via interactions between their FA targeting (FAT) domains and conserved segments (LD motifs) on the proteins Paxillin, Leupaxin, and Hic-5. A promising new approach for the inhibition of FAK and PYK2 targets interactions of the FAK domains with proteins that promote localization at FAs. Advances toward this goal include the development of surface plasmon resonance, heteronuclear single quantum coherence nuclear magnetic resonance (HSQC-NMR) and fluorescence polarization assays for the identification of fragments or compounds interfering with the FAK-Paxillin interaction. We have recently validated this strategy, showing that Paxillin mimicking polypeptides with 2 to 3 LD motifs displace FAK from FAs and block kinase-dependent and independent functions of FAK, including downstream integrin signaling and FA localization of the protein p130Cas. In the present work we study by all-atom molecular dynamics simulations the recognition of peptides with the Paxillin and Leupaxin LD motifs by the FAK-FAT and PYK2-FAT domains. Our simulations and free-energy analysis interpret experimental data on binding of Paxillin and Leupaxin LD motifs at FAK-FAT and PYK2-FAT binding sites, and assess the roles of consensus LD regions and flanking residues. Our results can assist in the design of effective inhibitory peptides of the FAK-FAT: Paxillin and PYK2-FAT:Leupaxin complexes and the construction of pharmacophore models for the discovery of potential small-molecule inhibitors of the FAK-FAT and PYK2-FAT focal adhesion based functions.
Collapse
Affiliation(s)
- Eleni Michael
- Department of Physics, University of Cyprus, Nicosia, Cyprus
| | | | - Vasilis J Promponas
- Bioinformatics Research Laboratory, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Paris Skourides
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | | |
Collapse
|
7
|
Seo Y, Park J, Choi W, Ju Son D, Sung Kim Y, Kim MK, Yoon BE, Pyee J, Tae Hong J, Go YM, Park H. Antiatherogenic Effect of Resveratrol Attributed to Decreased Expression of ICAM-1 (Intercellular Adhesion Molecule-1). Arterioscler Thromb Vasc Biol 2020; 39:675-684. [PMID: 30786743 DOI: 10.1161/atvbaha.118.312201] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objective- Increasing evidence shows that resveratrol has antiatherogenic effects, but its underlying mechanisms are unknown. Thus, we evaluated the molecular mechanisms underlying the antiatherogenic effect of resveratrol. Approach and Results- Using the previously established mouse atherosclerosis model of partial ligation of the left carotid artery, we evaluated the role of resveratrol in antiatherosclerosis. We attempted to determine the mechanisms associated with focal adhesions using vascular endothelial cells. The results showed that resveratrol stimulated focal adhesion kinase cleavage via resveratrol-increased expression of lactoferrin in endothelial cells. Furthermore, we found that an N-terminal focal adhesion kinase fragment cleaved by resveratrol contained the FERM (band 4.1, ezrin, radixin, and moesin)-kinase domain. Furthermore, resveratrol inhibited lipopolysaccharide-stimulated adhesion of THP-1 human monocytes by decreased expression of ICAM-1 (intercellular adhesion molecule-1). A decreased ICAM-1 level was also observed in the left carotid artery of mice treated with resveratrol. To understand the relationship between resveratrol-induced antiinflammation and focal adhesion disruption, endothelial cells were transfected with FERM-kinase. Ectopically expressed FERM-kinase, the resveratrol-cleaved focal adhesion kinase fragment, was found in the nuclear fraction and inhibited the transcription level of icam-1 via the Nrf2 (nuclear factor erythroid 2-related factor 2)-antioxidant response element complex. Finally, ectopically expressed FERM-kinase blocked tumor necrosis factor-α- or IL- (interleukin) stimulated monocytic binding to endothelial cells. Conclusions- Our results show that resveratrol inhibits the expression of ICAM-1 via transcriptional regulation of the FERM-kinase and Nrf2 interaction, thereby blocking monocyte adhesion. These suppressive effects on the inflammatory mechanism suggest that resveratrol delayed the onset of atherosclerosis.
Collapse
Affiliation(s)
- Youngsik Seo
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| | - Jinsun Park
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| | - Woosoung Choi
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, South Korea (D.J.S., J.T.H.)
| | - Yoo Sung Kim
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| | - Min-Kyun Kim
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| | - Bo-Eun Yoon
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| | - Jaeho Pyee
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, South Korea (D.J.S., J.T.H.)
| | - Young-Mi Go
- Pulmonary Medicine, Department of Medicine, Emory University, Atlanta, GA (Y.-M.G.)
| | - Heonyong Park
- From the Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook University, Chungnam, South Korea (Y.S., J. Park, W.C., Y.S.K., M.-K.K., B.-E.Y., J. Pyee, H.P.)
| |
Collapse
|
8
|
Sohn SO, Chay KO. The ATP-dependent RNA helicase, DDX42 interacts with paxillin and regulates apoptosis and polarization of Ba/F3 cells. Anim Cells Syst (Seoul) 2019; 23:1-9. [PMID: 30834153 PMCID: PMC6394298 DOI: 10.1080/19768354.2019.1567580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/24/2018] [Accepted: 01/06/2019] [Indexed: 01/08/2023] Open
Abstract
Paxillin is a focal adhesion adaptor protein, heavily phosphorylated at multiple tyrosine residues, as well as at serine 273 (S273), and is known to be critical for cytoskeleton rearrangement and cell migration. We previously found that paxillin plays a regulatory role in IL-3-dependent survival of Ba/F3 cells, a mouse pro-B cell line. In this study, by using overexpressed His6 tagged-paxillin as a bait, we found that DDX42, a DEAD-box RNA helicase, interacted with paxillin, inhibited apoptosis, and promoted polarization of Ba/F3 cells. His6 tagged-paxillin was stably overexpressed in Ba/F3 cells, pulled-down from cell lysates with Ni+-NTA beads, and analyzed by one-dimensional SDS-PAGE followed by LC–MS. We found that DDX42 co-precipitated with paxillin, as demonstrated by western blotting analysis of His6 tagged-paxillin precipitates with anti-DDX42 antibodies and His6 tagged-DDX42 precipitates with anti-paxillin antibodies. In addition, we observed a preferential interaction of DDX42 with the paxillin mutant, S273A, compared to the S273D mutant. Furthermore, DDX42 overexpression in Ba/F3 cells delayed the apoptosis induced by IL-3 deprivation and promoted restoration of the elongated shape in Ba/F3 cells induced by IL-3 re-supply after a 6 h-deprivation. These results suggested that DDX42 interacts with paxillin and participates in IL-3-dependent cell survival, as well as in the cytoskeletal rearrangements underlying polarization of Ba/F3 cells.
Collapse
Affiliation(s)
- Sung Oh Sohn
- Department of Biochemistry, Medical School, Chonnam National University, Jeollanam-do, Republic of Korea
| | - Kee Oh Chay
- Department of Biochemistry, Medical School, Chonnam National University, Jeollanam-do, Republic of Korea
| |
Collapse
|
9
|
Nah AS, Chay KO. Roles of paxillin phosphorylation in IL-3 withdrawal-induced Ba/F3 cell apoptosis. Genes Genomics 2019; 41:241-248. [PMID: 30604146 DOI: 10.1007/s13258-018-00779-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/19/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Ba/F3, a mouse pro-B cell line, is dependent on IL-3 for its survival and proliferation. IL-3 withdrawal causes cells to round, stop in G1 phase, then undergo apoptosis. Additionally, IL-3 is known to induce tyrosine phosphorylation of paxillin, a scaffold and signaling protein. We previously determined that overexpression of paxillin prohibited Ba/F3 cell apoptosis induced by IL-3 withdrawal. OBJECTIVE Address whether phosphorylation is essential for the anti-apoptotic effect of overexpressed paxillin. METHODS Mutations were introduced into paxillin cDNA at five phosphorylation sites-Y31F, Y40F, Y118F, Y181F, S273A, or S273D. After overexpression of paxillin mutants in Ba/F3 cells, the apoptotic proportions of cell populations were measured by an annexin V conjugation assay while cells were undergoing IL-3 withdrawal. RESULTS The anti-apoptotic effect of paxillin overexpression was abolished by site-directed mutagenesis replacing Y31, Y40, Y118, and Y181 with phenylalanine, and S273 with aspartic acid. In contrast, the mutation replacing S273 with alanine had no effect on the anti-apoptotic effect. CONCLUSION The above results suggest that paxillin-mediated phosphorylation at Y31, Y40, Y118, and Y181 is essential for the anti-apoptotic effect of paxillin overexpression in Ba/F3 cells and contributes to the cell survival signaling pathway triggered by IL-3. Conversely, phosphorylation at S273 is involved in the negative regulation of the anti-apoptotic action of overexpressed paxillin.
Collapse
Affiliation(s)
- Ae Sun Nah
- Department of Biochemistry, Medical School, Chonnam National University, 264, Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - Kee Oh Chay
- Department of Biochemistry, Medical School, Chonnam National University, 264, Seoyang-ro, Hwasun-eup, Hwasun-gun, Jeollanam-do, Republic of Korea.
| |
Collapse
|
10
|
Tamborindeguy MT, Matte BF, Ramos GDO, Alves AM, Bernardi L, Lamers ML. NADPH-oxidase-derived ROS alters cell migration by modulating adhesions dynamics. Biol Cell 2018; 110:225-236. [DOI: 10.1111/boc.201800011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Maurício Tavares Tamborindeguy
- Basic Research Center in Dentistry, Dentistry School; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
- Center of Biotechnology; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
| | - Bibiana Franzen Matte
- Basic Research Center in Dentistry, Dentistry School; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
| | - Grasieli de Oliveira Ramos
- Basic Research Center in Dentistry, Dentistry School; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
- School of Dentistry; University of Oeste de Santa Catarina; Joaçaba SC Brazil
| | - Alessandro Menna Alves
- Basic Research Center in Dentistry, Dentistry School; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
- School of Dentistry; University Center Univates; Lajeado RS Brazil
| | - Lisiane Bernardi
- Basic Research Center in Dentistry, Dentistry School; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
| | - Marcelo Lazzaron Lamers
- Basic Research Center in Dentistry, Dentistry School; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
- Department of Morphological Sciences, Institute of Basic Health Sciences; Federal University of Rio Grande do Sul; Porto Alegre RS Brazil
| |
Collapse
|
11
|
Gulvady AC, Dubois F, Deakin NO, Goreczny GJ, Turner CE. Hic-5 expression is a major indicator of cancer cell morphology, migration, and plasticity in three-dimensional matrices. Mol Biol Cell 2018; 29:1704-1717. [PMID: 29771639 PMCID: PMC6080706 DOI: 10.1091/mbc.e18-02-0092] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The focal adhesion proteins Hic-5 and paxillin have been previously identified as key regulators of MDA-MB-231 breast cancer cell migration and morphologic mesenchymal-amoeboid plasticity in three-dimensional (3D) extracellular matrices (ECMs). However, their respective roles in other cancer cell types have not been evaluated. Herein, utilizing 3D cell-derived matrices and fibronectin-coated one-dimensional substrates, we show that across a variety of cancer cell lines, the level of Hic-5 expression serves as the major indicator of the cells primary morphology, plasticity, and in vitro invasiveness. Domain mapping studies reveal sites critical to the functions of both Hic-5 and paxillin in regulating phenotype, while ectopic expression of Hic-5 in cell lines with low endogenous levels of the protein is sufficient to induce a Rac1-dependent mesenchymal phenotype and, in turn, increase amoeboid-mesenchymal plasticity and invasion. We show that the activity of vinculin, when coupled to the expression of Hic-5 is required for the mesenchymal morphology in the 3D ECM. Taken together, our results identify Hic-5 as a critical modulator of tumor cell phenotype that could be utilized in predicting tumor cell migratory and invasive behavior in vivo.
Collapse
Affiliation(s)
- Anushree C Gulvady
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Fatemeh Dubois
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Nicholas O Deakin
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Gregory J Goreczny
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
12
|
Maxson ME, Naj X, O'Meara TR, Plumb JD, Cowen LE, Grinstein S. Integrin-based diffusion barrier separates membrane domains enabling the formation of microbiostatic frustrated phagosomes. eLife 2018; 7:34798. [PMID: 29553370 PMCID: PMC5897098 DOI: 10.7554/elife.34798] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/16/2018] [Indexed: 12/25/2022] Open
Abstract
Candida albicans hyphae can reach enormous lengths, precluding their internalization by phagocytes. Nevertheless, macrophages engulf a portion of the hypha, generating incompletely sealed tubular phagosomes. These frustrated phagosomes are stabilized by a thick cuff of F-actin that polymerizes in response to non-canonical activation of integrins by fungal glycan. Despite their continuity, the surface and invaginating phagosomal membranes retain a strikingly distinct lipid composition. PtdIns(4,5)P2 is present at the plasmalemma but is not detectable in the phagosomal membrane, while PtdIns(3)P and PtdIns(3,4,5)P3 co-exist in the phagosomes yet are absent from the surface membrane. Moreover, endo-lysosomal proteins are present only in the phagosomal membrane. Fluorescence recovery after photobleaching revealed the presence of a diffusion barrier that maintains the identity of the open tubular phagosome separate from the plasmalemma. Formation of this barrier depends on Syk, Pyk2/Fak and formin-dependent actin assembly. Antimicrobial mechanisms can thereby be deployed, limiting the growth of the hyphae.
Collapse
Affiliation(s)
- Michelle E Maxson
- Program in Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Xenia Naj
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Teresa R O'Meara
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Jonathan D Plumb
- Program in Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Hospital for Sick Children, Toronto, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
13
|
Kpetemey M, Chaudhary P, Van Treuren T, Vishwanatha JK. MIEN1 drives breast tumor cell migration by regulating cytoskeletal-focal adhesion dynamics. Oncotarget 2018; 7:54913-54924. [PMID: 27462783 PMCID: PMC5342390 DOI: 10.18632/oncotarget.10798] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
Migration and invasion enhancer 1 (MIEN1) is an important regulator of cell migration and invasion. MIEN1 overexpression represents an oncogenic event that promotes tumor cell dissemination and metastasis. The underlying mechanism by which MIEN1 regulates migration and invasion has yet to be deciphered. Here, we demonstrate that MIEN1 acts as a cytoskeletal-signaling adapter protein to drive breast cancer cell migration. MIEN1 localization is concentrated underneath the actin-enriched protrusive structures of the migrating breast cancer cells. Depletion of MIEN1 led to the loss of actin-protrusive structures whereas the over-expression of MIEN1 resulted in rich and thick membrane extensions. Knockdown of MIEN1 also decreased the cell-substratum adhesion, suggesting a role for MIEN1 in actin cytoskeletal dynamics. Our results show that MIEN1 supports the transition of G-actin to F-actin polymerization and stabilizes F-actin polymers. Additionally, MIEN1 promotes cellular adhesion and actin dynamics by inducing phosphorylation of FAK at Tyr-925 and reducing phosphorylation of cofilin at Ser-3, which results in breast cancer cell migration. Collectively, our data show that MIEN1 plays an essential role in maintaining the plasticity of the dynamic membrane-associated actin cytoskeleton, which leads to an increase in cell motility. Hence, targeting MIEN1 might represent a promising means to prevent breast tumor metastasis.
Collapse
Affiliation(s)
- Marilyne Kpetemey
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Pankaj Chaudhary
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Timothy Van Treuren
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Jamboor K Vishwanatha
- Department of Molecular and Medical Genetics, Institute for Cancer Research, and The Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
14
|
Rashid M, Belmont J, Carpenter D, Turner CE, Olson EC. Neural-specific deletion of the focal adhesion adaptor protein paxillin slows migration speed and delays cortical layer formation. Development 2017; 144:4002-4014. [PMID: 28935710 DOI: 10.1242/dev.147934] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 09/11/2017] [Indexed: 11/20/2022]
Abstract
Paxillin and Hic-5 are homologous focal adhesion adaptor proteins that coordinate cytoskeletal rearrangements in response to integrin signaling, but their role(s) in cortical development are unknown. Here, we find that Hic-5-deficient mice are postnatal viable with normal cortical layering. Mice with a neural-specific deletion of paxillin are also postnatal viable, but show evidence of a cortical neuron migration delay that is evident pre- and perinatally, but is not detected at postnatal day 35 (P35). This phenotype is not modified by Hic-5 deficiency (double knockout). Specific deletion of paxillin in postmitotic neurons using Nex-Cre-mediated recombination as well as in utero electroporation of a Cre-expression construct identified a cell-autonomous requirement for paxillin in migrating neurons. Paxillin-deficient neurons have shorter leading processes that exhibited multiple swellings in comparison with control. Multiphoton imaging revealed that paxillin-deficient neurons migrate ∼30% slower than control neurons. This phenotype is similar to that produced by deletion of focal adhesion kinase (FAK), a signaling partner of paxillin, and suggests that paxillin and FAK function cell-autonomously to control migrating neuron morphology and speed during cortical development.
Collapse
Affiliation(s)
- Mamunur Rashid
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| | - Judson Belmont
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| | - David Carpenter
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| | - Christopher E Turner
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 E Adams Street, Syracuse, NY 13210, USA
| | - Eric C Olson
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY 13210, USA
| |
Collapse
|
15
|
Bortolotti F, Ruozi G, Falcione A, Doimo S, Dal Ferro M, Lesizza P, Zentilin L, Banks L, Zacchigna S, Giacca M. In Vivo Functional Selection Identifies Cardiotrophin-1 as a Cardiac Engraftment Factor for Mesenchymal Stromal Cells. Circulation 2017; 136:1509-1524. [PMID: 28754835 DOI: 10.1161/circulationaha.117.029003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 07/12/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Transplantation of cells into the infarcted heart has significant potential to improve myocardial recovery; however, low efficacy of cell engraftment still limits therapeutic benefit. Here, we describe a method for the unbiased, in vivo selection of cytokines that improve mesenchymal stromal cell engraftment into the heart both in normal conditions and after myocardial infarction. METHODS An arrayed library of 80 secreted factors, including most of the currently known interleukins and chemokines, were individually cloned into adeno-associated viral vectors. Pools from this library were then used for the batch transduction of bone marrow-derived mesenchymal stromal cells ex vivo, followed by intramyocardial cell administration in normal and infarcted mice. Three weeks after injection, vector genomes were recovered from the few persisting cells and identified by sequencing DNA barcodes uniquely labeling each of the tested cytokines. RESULTS The most effective molecule identified by this competitive engraftment screening was cardiotrophin-1, a member of the interleukin-6 family. Intracardiac injection of mesenchymal stromal cells transiently preconditioned with cardiotrophin-1 preserved cardiac function and reduced infarct size, parallel to the persistence of the transplanted cells in the healing hearts for at least 2 months after injection. Engraftment of cardiotrophin-1-treated mesenchymal stromal cells was consequent to signal transducer and activator of transcription 3-mediated activation of the focal adhesion kinase and its associated focal adhesion complex and the consequent acquisition of adhesive properties by the cells. CONCLUSIONS These results support the feasibility of selecting molecules in vivo for their functional properties with adeno-associated viral vector libraries and identify cardiotrophin-1 as a powerful cytokine promoting cell engraftment and thus improving cell therapy of the infarcted myocardium.
Collapse
Affiliation(s)
- Francesca Bortolotti
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Giulia Ruozi
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Antonella Falcione
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Sara Doimo
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Matteo Dal Ferro
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Pierluigi Lesizza
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Lorena Zentilin
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Lawrence Banks
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Serena Zacchigna
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.)
| | - Mauro Giacca
- From Molecular Medicine (F.B., G.R., A.F., L.Z., M.G.), Tumour Biology (L.B.), and Cardiovascular Biology (S.Z.) Laboratories, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.); and Center for Translational Cardiology, Azienda Sanitaria Integrata di Trieste, Italy (S.D., M.D.F., P.L., S.Z., M.G.).
| |
Collapse
|
16
|
Abstract
Understanding the underlying molecular mechanisms of liver fibrosis is important to develop effective therapy. Herein, we show that focal-adhesion-kinse (FAK) plays a key role in promoting hepatic stellate cells (HSCs) activation in vitro and liver fibrosis progression in vivo. FAK activation is associated with increased expression of α-smooth muscle actin (α-SMA) and collagen in fibrotic live tissues. Transforming growth factor beta-1 (TGF-β1) induces FAK activation in a time and dose dependent manner. FAK activation precedes the α-SMA expression in HSCs. Inhibition of FAK activation blocks the α-SMA and collagen expression, and inhibits the formation of stress fibers in TGF-β1 treated HSCs. Furthermore, inhibition of FAK activation significantly reduces HSC migration and small GTPase activation, and induces apoptotic signaling in TGF-β1 treated HSCs. Importantly, FAK inhibitor attenuates liver fibrosis in vivo and significantly reduces collagen and α-SMA expression in an animal model of liver fibrosis. These data demonstrate that FAK plays an essential role in HSC activation and liver fibrosis progression, and FAK signaling pathway could be a potential target for liver fibrosis.
Collapse
|
17
|
Lakshminarasimhan R, Andreu-Vieyra C, Lawrenson K, Duymich CE, Gayther SA, Liang G, Jones PA. Down-regulation of ARID1A is sufficient to initiate neoplastic transformation along with epigenetic reprogramming in non-tumorigenic endometriotic cells. Cancer Lett 2017; 401:11-19. [PMID: 28483516 DOI: 10.1016/j.canlet.2017.04.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/28/2022]
Abstract
The chromatin remodeler AT-Rich Interactive Domain 1A (ARID1A) is frequently mutated in ovarian clear cell carcinoma (OCCC) and endometriosis precursor lesions. Here, we show that knocking down ARID1A in an immortalized endometriosis cell line is sufficient to induce phenotypic changes indicative of neoplastic transformation as evidenced by higher efficiency of anchorage-independent growth, increased propensity to adhere to collagen, and greater capacity to invade basement membrane extract in vitro. ARID1A knockdown is associated with expression dysregulation of 99 target genes, and many of these expression changes are also observed in primary OCCC tissues. Further, pathway analysis indicates these genes fall within networks highly relevant to tumorigenesis including integrin and paxillin pathways. We demonstrate that the down-regulation of ARID1A does not markedly alter global chromatin accessibility or DNA methylation but unexpectedly, we find strong increases in the active H3K27ac mark in promoter regions and decreases of H3K27ac at potential enhancers. Taken together, these data provide evidence that ARID1A mutation can be an early stage event in the oncogenic transformation of endometriosis cells giving rise to OCCC.
Collapse
Affiliation(s)
- Ranjani Lakshminarasimhan
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Claudia Andreu-Vieyra
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kate Lawrenson
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA
| | - Christopher E Duymich
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Simon A Gayther
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA; Center for Bioinformatics and Functional Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gangning Liang
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Peter A Jones
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Van Andel Research Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
18
|
Antoniades I, Stylianou P, Christodoulou N, Skourides PA. Addressing the Functional Determinants of FAK during Ciliogenesis in Multiciliated Cells. J Biol Chem 2016; 292:488-504. [PMID: 27895123 DOI: 10.1074/jbc.m116.767111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/24/2016] [Indexed: 11/06/2022] Open
Abstract
We previously identified focal adhesion kinase (FAK) as an important regulator of ciliogenesis in multiciliated cells. FAK and other focal adhesion (FA) proteins associate with the basal bodies and their striated rootlets and form complexes named ciliary adhesions (CAs). CAs display similarities with FAs but are established in an integrin independent fashion and are responsible for anchoring basal bodies to the actin cytoskeleton during ciliogenesis as well as in mature multiciliated cells. FAK down-regulation leads to aberrant ciliogenesis due to impaired association between the basal bodies and the actin cytoskeleton, suggesting that FAK is an important regulator of the CA complex. However, the mechanism through which FAK functions in the complex is not clear, and in this study we examined the role of this protein in both ciliogenesis and ciliary function. We show that localization of FAK at CAs depends on interactions taking place at the amino-terminal (FERM) and carboxyl-terminal (FAT) domains and that both domains are required for proper ciliogenesis and ciliary function. Furthermore, we show that an interaction with another CA protein, paxillin, is essential for correct localization of FAK in multiciliated cells. This interaction is indispensable for both ciliogenesis and ciliary function. Finally, we provide evidence that despite the fact that FAK is in the active, open conformation at CAs, its kinase activity is dispensable for ciliogenesis and ciliary function revealing that FAK plays a scaffolding role in multiciliated cells. Overall these data show that the role of FAK at CAs displays similarities but also important differences compared with its role at FAs.
Collapse
Affiliation(s)
- Ioanna Antoniades
- From the Laboratory of Cell Biology and Molecular Embryology, Department of Biological Sciences, University of Cyprus, 1 University Avenue, Nicosia 2109, Cyprus
| | - Panayiota Stylianou
- From the Laboratory of Cell Biology and Molecular Embryology, Department of Biological Sciences, University of Cyprus, 1 University Avenue, Nicosia 2109, Cyprus
| | - Neophytos Christodoulou
- From the Laboratory of Cell Biology and Molecular Embryology, Department of Biological Sciences, University of Cyprus, 1 University Avenue, Nicosia 2109, Cyprus
| | - Paris A Skourides
- From the Laboratory of Cell Biology and Molecular Embryology, Department of Biological Sciences, University of Cyprus, 1 University Avenue, Nicosia 2109, Cyprus
| |
Collapse
|
19
|
Izaguirre-Carbonell J, Kawakubo H, Murata H, Tanabe A, Takeuchi T, Kusayanagi T, Tsukuda S, Hirakawa T, Iwabata K, Kanai Y, Ohta K, Miura M, Sakaguchi K, Matsunaga S, Sahara H, Kamisuki S, Sugawara F. Novel anticancer agent, SQAP, binds to focal adhesion kinase and modulates its activity. Sci Rep 2015; 5:15136. [PMID: 26456697 PMCID: PMC4601023 DOI: 10.1038/srep15136] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/18/2015] [Indexed: 01/03/2023] Open
Abstract
SQAP is a novel and promising anticancer agent that was obtained by structural modifications from a natural compound. SQAP inhibits angiogenesis in vivo resulting in increased hypoxia and reduced tumor volume. In this study, the mechanism by which SQAP modifies the tumor microenvironment was revealed through the application of a T7 phage display screening. This approach identified five SQAP-binding proteins including sterol carrier protein 2, multifunctional enzyme type 2, proteasomal ubiquitin receptor, UV excision repair protein and focal adhesion kinase (FAK). All the interactions were confirmed by surface plasmon resonance analysis. Since FAK plays an important role in cell turnover and angiogenesis, the influence of SQAP on FAK was the principal goal of this study. SQAP decreased FAK phosphorylation and cell migration in human umbilical vein endothelial cells and A549 cancer cells. These findings suggest that inhibition of FAK phosphorylation works as the mechanism for the anti-angiogenesis activity of SQAP.
Collapse
Affiliation(s)
- Jesus Izaguirre-Carbonell
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hirofumi Kawakubo
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hiroshi Murata
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Atsushi Tanabe
- Laboratory of Biology, Azabu University School of Veterinary Medicine, Sagamihara 229-8501, Japan
| | - Toshifumi Takeuchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Tomoe Kusayanagi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Senko Tsukuda
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Takeshi Hirakawa
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kazuki Iwabata
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yoshihiro Kanai
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Keisuke Ohta
- Department of Oral Radiation Oncology, Graduate school, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-85-10, Japan
| | - Masahiko Miura
- Department of Oral Radiation Oncology, Graduate school, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-85-10, Japan
| | - Kengo Sakaguchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Sachihiro Matsunaga
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hiroeki Sahara
- Laboratory of Biology, Azabu University School of Veterinary Medicine, Sagamihara 229-8501, Japan
| | - Shinji Kamisuki
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Fumio Sugawara
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
20
|
Nocula-Lugowska M, Lugowski M, Salgia R, Kossiakoff AA. Engineering Synthetic Antibody Inhibitors Specific for LD2 or LD4 Motifs of Paxillin. J Mol Biol 2015; 427:2532-2547. [PMID: 26087144 DOI: 10.1016/j.jmb.2015.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/01/2015] [Accepted: 06/10/2015] [Indexed: 10/23/2022]
Abstract
Focal adhesion protein paxillin links integrin and growth factor signaling to actin cytoskeleton. Most of paxillin signaling activity is regulated via leucine-rich LD motifs (LD1-LD5) located at the N-terminus. Here, we demonstrate a method to engineer highly selective synthetic antibodies (sABs) against LD2 and LD4 that are binding sites for focal adhesion kinase (FAK) and other proteins. Phage display selections against peptides were used to generate sABs recognizing each LD motif. In the obtained X-ray crystal structures of the LD-sAB complexes, the LD motifs are helical and bind sABs through a hydrophobic side, similarly as in the structures with natural paxillin partners. The sABs are capable of pulling down endogenous paxillin in complex with FAK and can visualize paxillin in focal adhesions in cells. They were also used as selective inhibitors to effectively compete with focal adhesion targeting domain of FAK for the binding to LD2 and LD4. The sABs are tools for investigation of paxillin LD binding "platforms" and are capable of inhibiting paxillin interactions, thereby useful as potential therapeutics in the future.
Collapse
Affiliation(s)
| | - Mateusz Lugowski
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60607, USA
| | - Ravi Salgia
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
21
|
Béraud C, Dormoy V, Danilin S, Lindner V, Béthry A, Hochane M, Coquard C, Barthelmebs M, Jacqmin D, Lang H, Massfelder T. Targeting FAK scaffold functions inhibits human renal cell carcinoma growth. Int J Cancer 2015; 137:1549-59. [PMID: 25809490 DOI: 10.1002/ijc.29522] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 03/06/2015] [Indexed: 11/07/2022]
Abstract
Human conventional renal cell carcinoma (CCC) remains resistant to current therapies. Focal Adhesion Kinase (FAK) is upregulated in many epithelial tumors and clearly implicated in nearly all facets of cancer. However, only few reports have assessed whether FAK may be associated with renal tumorigenesis. In this study, we investigated the potential role of FAK in the growth of human CCC using a panel of CCC cell lines expressing or not the von Hippel-Lindau (VHL) tumor suppressor gene as well as normal/tumoral renal tissue pairs. FAK was found constitutively expressed in human CCC both in culture cells and freshly harvested tumors obtained from patients. We showed that CCC cell growth was dramatically reduced in FAK-depleted cells or after FAK inhibition with various inhibitors and this effect was obtained through inhibition of cell proliferation and induction of cell apoptosis. Additionally, our results indicated that FAK knockdown decreased CCC cell migration and invasion. More importantly, depletion or pharmacological inhibition of FAK substantially inhibited tumor growth in vivo. Interestingly, investigations of the molecular mechanism revealed loss of FAK phosphorylation during renal tumorigenesis impacting multiple signaling pathways. Taken together, our findings reveal a previously uncharacterized role of FAK in CCC whereby FAK exerts oncogenic properties through a non canonical signaling pathway involving its scaffolding kinase-independent properties. Therefore, targeting the FAK scaffold may represent a promising approach for developing innovative and highly specific therapies in human CCC.
Collapse
Affiliation(s)
- Claire Béraud
- Inserm U1113, University of Strasbourg, Strasbourg, France
| | | | | | - Véronique Lindner
- Department of Pathology, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Audrey Béthry
- Inserm U1113, University of Strasbourg, Strasbourg, France
| | - Mazène Hochane
- Inserm U1113, University of Strasbourg, Strasbourg, France
| | | | | | - Didier Jacqmin
- Department of Urology, Nouvel Hôpital Civil De Strasbourg, Strasbourg, France
| | - Hervé Lang
- Department of Urology, Nouvel Hôpital Civil De Strasbourg, Strasbourg, France
| | | |
Collapse
|
22
|
Kim YS, Kim J, Kim KM, Jung DH, Choi S, Kim CS, Kim JS. Myricetin inhibits advanced glycation end product (AGE)-induced migration of retinal pericytes through phosphorylation of ERK1/2, FAK-1, and paxillin in vitro and in vivo. Biochem Pharmacol 2014; 93:496-505. [PMID: 25450667 DOI: 10.1016/j.bcp.2014.09.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 01/24/2023]
Abstract
Advanced glycation end products (AGE) have been implicated in the development of diabetic retinopathy. Characterization of the early stages of diabetic retinopathy is retinal pericytes loss, which is the result of pericytes migration. In this study, we investigated the pathological mechanisms of AGE on the migration of retinal pericytes and confirmed the inhibitory effect of myricetin on migration in vitro and in vivo. Migration assays of bovine retinal pericytes (BRP) were induced using AGE-BSA and phosphorylation of Src, ERK1/2, focal adhesion kinase (FAK-1) and paxillin were determined using immunoblot analysis. Sprague-Dawley rats (6 weeks old) were injected intravitreally with AGE-BSA and morphological and immunohistochemical analysis of p-FAK-1 and p-paxillin were performed in the rat retina. Immunoblot analysis and siRNA transfection were used to study the molecular mechanism of myricetin on BRP migration. AGE-BSA increased BRP migration in a dose-dependent manner via receptor for AGEs (RAGE)-dependent activation of the Src kinase-ERK1/2 pathway. AGE-BSA-induced migration was inhibited by an ERK1/2 specific inhibitor (PD98059), but not by p38 and Jun N-terminal kinase inhibitors. AGE-BSA increased FAK-1 and paxillin phosphorylation in a dose- and time-dependent manner. These increases were attenuated by PD98059 and ERK1/2 siRNA. Phosphorylation of FAK-1 and paxillin was increased in response to AGE-BSA-induced migration of rat retinal pericytes. Myricetin strongly inhibited ERK1/2 phosphorylation and significantly suppressed pericytes migration in AGE-BSA-injected rats. Our results demonstrate that AGE-BSA participated in the pathophysiology of retinal pericytes migration likely through the RAGE-Src-ERK1/2-FAK-1-paxillin signaling pathway. Furthermore, myricetin suppressed phosphorylation of ERK 1/2-FAK-1-paxillin and inhibited pericytes migration.
Collapse
Affiliation(s)
- Young Sook Kim
- Korean Medicine-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, South Korea.
| | - Junghyun Kim
- Korean Medicine-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, South Korea
| | - Ki Mo Kim
- Korean Medicine-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, South Korea
| | - Dong Ho Jung
- Korean Medicine-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, South Korea
| | - Sojin Choi
- Korean Medicine-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, South Korea
| | - Chan-Sik Kim
- Korean Medicine-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, South Korea
| | - Jin Sook Kim
- Korean Medicine-Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, South Korea.
| |
Collapse
|
23
|
Armendáriz BG, Masdeu MDM, Soriano E, Ureña JM, Burgaya F. The diverse roles and multiple forms of focal adhesion kinase in brain. Eur J Neurosci 2014; 40:3573-90. [DOI: 10.1111/ejn.12737] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/25/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Beatriz G. Armendáriz
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Maria del Mar Masdeu
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Eduardo Soriano
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Jesús M. Ureña
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| | - Ferran Burgaya
- Department of Biologia Cellular; Fac Biologia; Universitat de Barcelona; Diagonal, 643 08028 Barcelona Spain
- Parc Científic de Barcelona; Barcelona Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas Ciberned (ISC III); Madrid Spain
| |
Collapse
|
24
|
Fang X, Liu X, Yao L, Chen C, Lin J, Ni P, Zheng X, Fan Q. New insights into FAK phosphorylation based on a FAT domain-defective mutation. PLoS One 2014; 9:e107134. [PMID: 25226367 PMCID: PMC4166415 DOI: 10.1371/journal.pone.0107134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 08/11/2014] [Indexed: 01/27/2023] Open
Abstract
Mounting evidence suggests that the FAK N-terminal (FERM) domain controls FAK phosphorylation and function; however, little is known regarding the role of the C terminal (FAT) domain in FAK regulation. We identified a patient-derived FAK mutant, in which a 27-amino acid segment was deleted from the C-terminal FAT domain (named FAK-Del33). When FAK-Del33 was overexpressed in specific tumor cell lines, Y397 phosphorylation increased compared with that observed in cells expressing FAK-WT. Here, we attempt to unveil the mechanism of this increased phosphorylation. Using cell biology experiments, we show that FAK-Del33 is incapable of co-localizing with paxillin, and has constitutively high Y397 phosphorylation. With a kinase-dead mutation, it showed phosphorylation of FAK-Del33 has enhanced through auto-phosphorylation. It was also demonstrated that phosphorylation of FAK-Del33 is not Src dependent or enhanced intermolecular interactions, and that the hyperphosphorylation can be lowered using increasing amounts of transfected FERM domain. This result suggests that Del33 mutation disrupting of FAT's structural integrity and paxillin binding capacity leads to incapable of targeting Focal adhesions, but has gained the capacity for auto-phosphorylation in cis.
Collapse
Affiliation(s)
- Xuqian Fang
- Department of Clinical Laboratory, Ruijin North Hospital, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
| | - Xiangfan Liu
- Faculty of Medical Laboratory Science, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
| | - Ling Yao
- Department of Biochemistry and Molecular Biology, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
| | - Changqiang Chen
- Department of Clinical Laboratory, Ruijin North Hospital, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
| | - Jiafei Lin
- Department of Clinical Laboratory, Ruijin North Hospital, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
| | - Peihua Ni
- Faculty of Medical Laboratory Science, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
| | - Xinmin Zheng
- Department of Biochemistry and Molecular Biology, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Qishi Fan
- Department of Clinical Laboratory, Ruijin North Hospital, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P. R. China
- * E-mail:
| |
Collapse
|
25
|
Yao L, Li K, Peng W, Lin Q, Li S, Hu X, Zheng X, Shao Z. An aberrant spliced transcript of focal adhesion kinase is exclusively expressed in human breast cancer. J Transl Med 2014; 12:136. [PMID: 24885534 PMCID: PMC4040474 DOI: 10.1186/1479-5876-12-136] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 05/17/2014] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To clarify the roles of a new aberrantly spliced transcript of FAK that lacks exon 26 (denoted -26-exon FAK) in human breast cancers. METHODS Transcripts of FAK expressed in 102 human breast tumor tissues and 52 corresponding normal tissues were analyzed by RT-PCR and DNA sequencing, as well as agarose gel electrophoresis. The cDNA of -26-exon FAK was cloned and expressed in MCF-10A cells, and then the kinase activity, cellular localization and migration capability of FAK were examined by western blotting, immunofluorescent staining and migration assays, respectively. The expression levels of FAK were analyzed by western blotting in MCF-7 cells treated with TNF-α or in MCF-10A cells upon serum deprivation. The MCF-10A cells transfected with a plasmid expressing -26-exon FAK were cultured in serum-free medium and cell apoptosis was analyzed by flow cytometry. RESULTS The -26-exon FAK transcript was exclusively present in human breast tumor tissues and the encoded protein possessed the same kinase activity, cellular localization and cell migration-promoting ability as wild-type FAK. In MCF-7 cells treated with TNF-α, and in MCF-10A cells upon serum deprivation, the -26-exon FAK was resistant to proteolysis while wild-type FAK was largely cleaved. In addition, the -26-exon FAK, but not wild-type FAK, inhibited cell apoptosis. CONCLUSIONS The -26-exon FAK transcript, which is exclusively expressed in human breast tumor tissues, encodes a protein that possesses the same kinase activity and biological function as the wild-type FAK, but because it is resistant to the caspase-mediated cleavage that induces the proteolysis of the wild-type form, it ultimately prevents apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhiming Shao
- Department of Oncology, Breast Cancer Institute, Shanghai Cancer Center, Shanghai Medical College, Fudan University, No,270, Dong'an Road, Shanghai 200032, People's Republic of China.
| |
Collapse
|
26
|
Paxillin and focal adhesion kinase colocalise in human skeletal muscle and its associated microvasculature. Histochem Cell Biol 2014; 142:245-56. [PMID: 24671495 DOI: 10.1007/s00418-014-1212-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/15/2023]
Abstract
Focal adhesion kinase (FAK) and paxillin are functionally linked hormonal- and mechano-sensitive proteins. We aimed to describe paxillin's subcellular distribution using widefield and confocal immunofluorescence microscopy and test the hypothesis that FAK and paxillin colocalise in human skeletal muscle and its associated microvasculature. Percutaneous muscle biopsies were collected from the m. vastus lateralis of seven healthy males, and 5-μm cryosections were stained with anti-paxillin co-incubated with anti-dystrophin to identify the sarcolemma, anti-myosin heavy chain type I for fibre-type differentiation, anti-dihydropyridine receptor to identify T-tubules, lectin UEA-I to identify the endothelium of microvessels and anti-α-smooth muscle actin to identify vascular smooth muscle cells (VSMC). Colocalisation of anti-paxillin with anti-dystrophin or anti-FAK was quantified using Pearson's correlation coefficient on confocal microscopy images. Paxillin was primarily present in (sub)sarcolemmal regions of skeletal muscle fibres where it colocalised with dystrophin (r = 0.414 ± 0.026). The (sub)sarcolemmal paxillin immunofluorescence intensity was ~2.4-fold higher than in sarcoplasmic regions (P < 0.001) with sarcoplasmic paxillin immunofluorescence intensity ~10 % higher in type I than in type II fibres (P < 0.01). In some longitudinally orientated fibres, paxillin formed striations that corresponded to the I-band region. Paxillin immunostaining was highest in endothelial and VSMC and distributed heterogeneously in both cell types. FAK and paxillin colocalised at (sub)sarcolemmal regions and within the microvasculature (r = 0.367 ± 0.036). The first images of paxillin in human skeletal muscle suggest paxillin is present in (sub)sarcolemmal and I-band regions of muscle fibres and within the microvascular endothelium and VSMC. Colocalisation of FAK and paxillin supports their suggested role in hormonal and mechano-sensitive signalling.
Collapse
|
27
|
Antoniades I, Stylianou P, Skourides PA. Making the connection: ciliary adhesion complexes anchor basal bodies to the actin cytoskeleton. Dev Cell 2014; 28:70-80. [PMID: 24434137 DOI: 10.1016/j.devcel.2013.12.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 11/08/2013] [Accepted: 12/06/2013] [Indexed: 01/01/2023]
Abstract
Cilia have been associated with diverse developmental and physiological processes, and defects in cilia underlie a number of genetic conditions. Several lines of evidence support a critical role of the actin cytoskeleton in ciliogenesis and ciliary function. Here, we show that well-characterized focal adhesion (FA) proteins, including FAK, Paxillin, and Vinculin, associate with the basal bodies of multiciliated cells and form complexes (CAs) that interact with the actin cytoskeleton. FAK downregulation leads to ciliogenesis defects similar to those observed when the actin cytoskeleton is disrupted, including defects in basal body migration, docking, and spacing, suggesting that CAs link basal bodies to the actin cytoskeleton. The important role of FA proteins in ciliogenesis leads us to propose that evolutionarily FA proteins, many of which are found in primitive flagellated unicellular eukaryotes, may have originally evolved to perform functions at flagella and were later co-opted for use in cell adhesion.
Collapse
Affiliation(s)
- Ioanna Antoniades
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, Nicosia 2109, Cyprus
| | - Panayiota Stylianou
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, Nicosia 2109, Cyprus
| | - Paris A Skourides
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, Nicosia 2109, Cyprus.
| |
Collapse
|
28
|
Golubovskaya VM. Targeting FAK in human cancer: from finding to first clinical trials. Front Biosci (Landmark Ed) 2014; 19:687-706. [PMID: 24389213 DOI: 10.2741/4236] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
It is twenty years since Focal Adhesion Kinase (FAK) was found to be overexpressed in many types of human cancer. FAK plays an important role in adhesion, spreading, motility, invasion, metastasis, survival, angiogenesis, and recently has been found to play an important role as well in epithelial to mesenchymal transition (EMT), cancer stem cells and tumor microenvironment. FAK has kinase-dependent and kinase independent scaffolding, cytoplasmic and nuclear functions. Several years ago FAK was proposed as a potential therapeutic target; the first clinical trials were just reported, and they supported further studies of FAK as a promising therapeutic target. This review discusses the main functions of FAK in cancer, and specifically focuses on recent novel findings on the role of FAK in cancer stem cells, microenvironment, epithelial-to-mesenchymal transition, invasion, metastasis, and also highlight new approaches of targeting FAK and critically discuss challenges that lie ahead for its targeted therapeutics. The review provides a summary of translational approaches of FAK-targeted and combination therapies and outline perspectives and future directions of FAK research.
Collapse
|
29
|
Grb2 promotes integrin-induced focal adhesion kinase (FAK) autophosphorylation and directs the phosphorylation of protein tyrosine phosphatase α by the Src-FAK kinase complex. Mol Cell Biol 2013; 34:348-61. [PMID: 24248601 DOI: 10.1128/mcb.00825-13] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The integrin-activated Src-focal adhesion kinase (FAK) kinase complex phosphorylates PTPα at Tyr789, initiating PTPα-mediated signaling that promotes cell migration. Recruitment of the BCAR3-Cas complex by PTPα-phospho-Tyr789 at focal adhesions is one mechanism of PTPα signaling. The adaptor protein Grb2 is also recruited by PTPα-phospho-Tyr789, although the role of the PTPα-Grb2 complex in integrin signaling is unknown. We show that silencing Grb2 expression in fibroblasts abolishes PTPα-Tyr789 phosphorylation and that this is due to two unexpected actions of Grb2. First, Grb2 promotes integrin-induced autophosphorylation of FAK-Tyr397. This is impaired in Grb2-depleted cells and prohibits FAK activation and formation of the Src-FAK complex. Grb2-depleted cells contain less paxillin, and paxillin overexpression rescues FAK-Tyr397 phosphorylation, suggesting that the FAK-activating action of Grb2 involves paxillin. A second distinct role for Grb2 in PTPα-Tyr789 phosphorylation involves Grb2-mediated coupling of Src-FAK and PTPα. This requires two phosphosites, FAK-Tyr925 and PTPα-Tyr789, for Grb2-Src homology 2 (SH2) binding. We propose that a Grb2 dimer links FAK and PTPα, and this positions active Src-FAK in proximity with other, perhaps integrin-clustered, molecules of PTPα to enable maximal PTPα-Tyr789 phosphorylation. These findings identify Grb2 as a new FAK activator and reveal its essential role in coordinating PTPα tyrosine phosphorylation to enable downstream integrin signaling and migration.
Collapse
|
30
|
Lee HR, Kim J, Park J, Ahn S, Jeong E, Park H. FERM domain promotes resveratrol-induced apoptosis in endothelial cells via inhibition of NO production. Biochem Biophys Res Commun 2013; 441:891-6. [PMID: 24211585 DOI: 10.1016/j.bbrc.2013.10.154] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 10/29/2013] [Indexed: 12/14/2022]
Abstract
Focal adhesion kinase (FAK) consists of an N-terminal band 4.1; ezrin, radixin, moesin (FERM) domain; tyrosine kinase domain; and C-terminal FA targeting domain. Here we show that ectopically expressed FERM is largely located in the cytosolic fraction under quiescent conditions. We further found that this ectopically expressed FERM domain aggravates endothelial cell apoptosis triggered by 100 μM resveratrol, whereas FERM had no effect on apoptosis induced by TNF-α. We determined that resveratrol at low doses (<20 μM) promotes phosphorylation (S1177) of eNOS via an AMPK-dependent pathway. The presence of the FERM domain blocked this resveratrol-stimulated eNOS phosphorylation and NO production. Thus, the pro-apoptotic activity of cytosolic FERM domain is at least partially mediated by down-regulation of NO, a critical cell survival factor. Consistently, we found that the apoptosis induced by cytosolic FERM in the presence of resveratrol was reversed by an NO donor, SNAP. In conclusion, FERM located in the cytosolic fraction plays a pivotal role in aggravating cell apoptosis through diminishing NO production.
Collapse
Affiliation(s)
- Hye-Rim Lee
- Department of Molecular Biology & Institute of Nanosensor and Biotechnology, Dankook Univiersity, 126, Jukjeon-dong, Suji-gu, Yongin-si, Gyeonggi-do 448-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
31
|
Reynolds AB, Kanner SB, Bouton AH, Schaller MD, Weed SA, Flynn DC, Parsons JT. SRChing for the substrates of Src. Oncogene 2013; 33:4537-47. [PMID: 24121272 DOI: 10.1038/onc.2013.416] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/16/2013] [Accepted: 08/17/2013] [Indexed: 12/12/2022]
Abstract
By the mid 1980's, it was clear that the transforming activity of oncogenic Src was linked to the activity of its tyrosine kinase domain and attention turned to identifying substrates, the putative next level of control in the pathway to transformation. Among the first to recognize the potential of phosphotyrosine-specific antibodies, Parsons and colleagues launched a risky shotgun-based approach that led ultimately to the cDNA cloning and functional characterization of many of today's best-known Src substrates (for example, p85-Cortactin, p110-AFAP1, p130Cas, p125FAK and p120-catenin). Two decades and over 6000 citations later, the original goals of the project may be seen as secondary to the enormous impact of these protein substrates in many areas of biology. At the request of the editors, this review is not restricted to the current status of the substrates, but reflects also on the anatomy of the project itself and some of the challenges and decisions encountered along the way.
Collapse
Affiliation(s)
- A B Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - S B Kanner
- Arrowhead Research Corporation, Madison, WI, USA
| | - A H Bouton
- Departments of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - M D Schaller
- Department of Biochemistry, 3124 HSN, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - S A Weed
- Department of Neurobiology and Anatomy, 1833 Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - D C Flynn
- Department of Medical Lab Sciences, College of Health Sciences, University of Delaware, Newark, DE, USA
| | - J T Parsons
- Departments of Microbiology, Immunology and Cancer Biology, University of Virginia Cancer Center, Charlottesville, VA, USA
| |
Collapse
|
32
|
Petridou NI, Stylianou P, Skourides PA. A dominant-negative provides new insights into FAK regulation and function in early embryonic morphogenesis. Development 2013; 140:4266-76. [PMID: 24048589 DOI: 10.1242/dev.096073] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
FAK is a non-receptor tyrosine kinase involved in a wide variety of biological processes and crucial for embryonic development. In this manuscript, we report the generation of a new FAK dominant negative (FF), composed of the C terminus (FRNK) and the FERM domain of the protein. FF, unlike FRNK and FERM, mimics the localization of active FAK in the embryo, demonstrating that both domains are necessary to target FAK to its complexes in vivo. We show that the FERM domain has a role in the recruitment of FAK on focal adhesions and controls the dynamics of the protein on these complexes. Expression of FF blocks focal adhesion turnover and, unlike FRNK, acts as a dominant negative in vivo. FF expression in Xenopus results in an overall phenotype remarkably similar to the FAK knockout in mice, including loss of mesodermal tissues. Expression of FF in the animal cap revealed a previously unidentified role of FAK in early morphogenesis and specifically epiboly. We show that a fibronectin-derived signal transduced by FAK governs polarity and cell intercalation. Finally, failure of epiboly results in severe gastrulation problems that can be rescued by either mechanical or pharmacological relief of tension within the animal cap, demonstrating that epiboly is permissive for gastrulation. Overall, this work introduces a powerful new tool for the study of FAK, uncovers new roles for FAK in morphogenesis and reveals new mechanisms through which the FERM domain regulates the localization and dynamics of FAK.
Collapse
|
33
|
Dreier B, Gasiorowski JZ, Morgan JT, Nealey PF, Russell P, Murphy CJ. Early responses of vascular endothelial cells to topographic cues. Am J Physiol Cell Physiol 2013; 305:C290-8. [PMID: 23703527 DOI: 10.1152/ajpcell.00264.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular endothelial cells in vivo are exposed to multiple biophysical cues provided by the basement membrane, a specialized extracellular matrix through which vascular endothelial cells are attached to the underlying stroma. The importance of biophysical cues has been widely reported, but the signaling pathways that mediate cellular recognition and response to these cues remain poorly understood. Anisotropic topographically patterned substrates with nano- through microscale feature dimensions were fabricated to investigate cellular responses to topographic cues. The present study focuses on early events following exposure of human umbilical vein endothelial cells (HUVECs) to these patterned substrates. In serum-free medium and on substrates without protein coating, HUVECs oriented parallel to the long axis of underlying ridges in as little as 30 min. Immunocytochemistry showed clear differences in the localization of the focal adhesion proteins Src, p130Cas, and focal adhesion kinase (FAK) in HUVECs cultured on topographically patterned surfaces and on planar surfaces, suggesting involvement of these proteins in mediating the response to topographic features. Knockdown experiments demonstrated that FAK was not necessary for HUVEC alignment in response to topographic cues, although FAK knockdown did modulate HUVEC migration. These data identify key events early in the cellular response to biophysical stimuli.
Collapse
Affiliation(s)
- Britta Dreier
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | | | | | | | | | | |
Collapse
|
34
|
Chi C, Trinkaus-Randall V. New insights in wound response and repair of epithelium. J Cell Physiol 2013; 228:925-9. [PMID: 23129239 DOI: 10.1002/jcp.24268] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 10/18/2012] [Indexed: 01/26/2023]
Abstract
Epithelial wounds usually heal relatively quickly, but repair may be impaired by environmental stressors, such as hypoxic or diabetic states, rendering patients vulnerable to a number of corneal pathologies. Though this response appears simple, at first, years of research have uncovered the complicated biochemical pathways coordinating the wound healing response. Here, we investigate signaling cascades and individual proteins involved in the corneal epithelium's self-repair. We will explore how an epithelial cell migrates across the wound bed and attaches itself to its new post-injury surroundings, including its neighboring cells and the basement membrane, through focal adhesions and hemidesmosomes. We will also discuss how the cell coordinates this motion physiologically, through calcium signaling and protein phosphorylation, focusing on the communication through purinergic, glutamatergic, and growth factor receptors. Many of these aspects reflect and can be extended to similar epithelial surfaces, and can be used to facilitate wound healing in patients with various underlying pathologies. The collective library of laboratory and clinical research done around the world has demonstrated how important precise regulation of these processes is in order for the injured corneal epithelium to properly heal.
Collapse
Affiliation(s)
- Cheryl Chi
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
35
|
Dreier B, Raghunathan VK, Russell P, Murphy CJ. Focal adhesion kinase knockdown modulates the response of human corneal epithelial cells to topographic cues. Acta Biomater 2012; 8:4285-94. [PMID: 22813850 DOI: 10.1016/j.actbio.2012.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 06/15/2012] [Accepted: 07/10/2012] [Indexed: 11/17/2022]
Abstract
A rapidly expanding literature broadly documents the impact of biophysical cues on cellular behaviors. In spite of increasing research efforts in this field, the underlying signaling processes are poorly understood. One of the candidate molecules for being involved in mechanotransduction is focal adhesion kinase (FAK). To examine the role of FAK in the response of immortalized human corneal epithelial (hTCEpi) cells to topographic cues, FAK was depleted by siRNA transfection. Contrary to expectations, FAK knockdown resulted in an enhanced response with a greater number of hTCEpi cells aligned to the long axis of anisotropically ordered surface ridges and grooves. Both underlying topographic features and FAK depletion modulated the migration of corneal epithelial cells. The impact of FAK knockdown on both migration and alignment varied depending on the topographic cues to which the cells were exposed, with the most significant change observed on the biologically relevant size scale (400nm). Additionally, a change in expression of genes encoding perinuclear Nesprins 1 and 2 (SYNE1, 2) was observed in response to topographic cues. SYNE1/2 expression was also altered by FAK depletion, suggesting that these proteins might represent a link between cytosolic and nuclear signaling processes. The data presented here have relevance to our understanding of the fundamental processes involved in corneal cell behavior to topographic cues. These results highlight the importance of incorporating biophysical cues in the conduction of in vitro studies and into the design and fabrication of implantable prosthetics.
Collapse
Affiliation(s)
- Britta Dreier
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, One Shields Avenue, University of California Davis, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
36
|
Roca-Cusachs P, Iskratsch T, Sheetz MP. Finding the weakest link: exploring integrin-mediated mechanical molecular pathways. J Cell Sci 2012; 125:3025-38. [PMID: 22797926 DOI: 10.1242/jcs.095794] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
From the extracellular matrix to the cytoskeleton, a network of molecular links connects cells to their environment. Molecules in this network transmit and detect mechanical forces, which subsequently determine cell behavior and fate. Here, we reconstruct the mechanical pathway followed by these forces. From matrix proteins to actin through integrins and adaptor proteins, we review how forces affect the lifetime of bonds and stretch or alter the conformation of proteins, and how these mechanical changes are converted into biochemical signals in mechanotransduction events. We evaluate which of the proteins in the network can participate in mechanotransduction and which are simply responsible for transmitting forces in a dynamic network. Besides their individual properties, we also analyze how the mechanical responses of a protein are determined by their serial connections from the matrix to actin, their parallel connections in integrin clusters and by the rate at which force is applied to them. All these define mechanical molecular pathways in cells, which are emerging as key regulators of cell function alongside better studied biochemical pathways.
Collapse
Affiliation(s)
- Pere Roca-Cusachs
- University of Barcelona and Institute for Bioengineering of Catalonia, Barcelona, Spain.
| | | | | |
Collapse
|
37
|
XIAP reverses various functional activities of FRNK in endothelial cells. Biochem Biophys Res Commun 2012; 419:419-24. [DOI: 10.1016/j.bbrc.2012.02.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 02/07/2012] [Indexed: 11/22/2022]
|
38
|
Koshman YE, Chu M, Engman SJ, Kim T, Iyengar R, Robia SL, Samarel AM. Focal adhesion kinase-related nonkinase inhibits vascular smooth muscle cell invasion by focal adhesion targeting, tyrosine 168 phosphorylation, and competition for p130(Cas) binding. Arterioscler Thromb Vasc Biol 2012; 31:2432-40. [PMID: 21852560 DOI: 10.1161/atvbaha.111.235549] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Focal adhesion kinase-related nonkinase (FRNK), the C-terminal domain of focal adhesion kinase (FAK), is a tyrosine-phosphorylated, vascular smooth muscle cell (VSMC)-specific inhibitor of cell migration. FRNK inhibits both FAK and proline-rich tyrosine kinase 2 (PYK2) in cultured VSMCs, and both kinases may be involved in VSMC invasion during vascular remodeling. METHODS AND RESULTS Adenovirally mediated gene transfer of green fluorescent protein-tagged, wild-type (wt) FRNK into balloon-injured rat carotid arteries confirmed that FRNK overexpression inhibited both FAK and PYK2 phosphorylation and downstream signaling in vivo. To identify which kinase was involved in regulating VSMC invasion, adenovirally mediated expression of specific short hairpin RNAs was used to knock down FAK versus PYK2 in cultured VSMCs, but only FAK short hairpin RNA was effective in reducing VSMC invasion. The role of FRNK tyrosine phosphorylation was then examined using adenoviruses expressing nonphosphorylatable (Tyr168Phe-, Tyr232Phe-, and Tyr168,232Phe-) green fluorescent protein-FRNK mutants. wtFRNK and all FRNK mutants localized to FAs, but only Tyr168 phosphorylation was required for FRNK to inhibit invasion. Preventing Tyr168 phosphorylation also increased FRNK-paxillin interaction, as determined by coimmunoprecipitation, total internal reflection fluorescence microscopy, and fluorescence recovery after photobleaching. Furthermore, wtFRNK competed with FAK for binding to p130(Cas) (a critically important regulator of cell migration) and prevented its phosphorylation. However, Tyr168Phe-FRNK was unable to bind p130(Cas). CONCLUSION We propose a 3-stage mechanism for FRNK inhibition: focal adhesion targeting, Tyr168 phosphorylation, and competition with FAK for p130 binding and phosphorylation, which are all required for FRNK to inhibit VSMC invasion.
Collapse
Affiliation(s)
- Yevgeniya E Koshman
- Cardiovascular Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Sero JE, Thodeti CK, Mammoto A, Bakal C, Thomas S, Ingber DE. Paxillin mediates sensing of physical cues and regulates directional cell motility by controlling lamellipodia positioning. PLoS One 2011; 6:e28303. [PMID: 22194823 PMCID: PMC3237434 DOI: 10.1371/journal.pone.0028303] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 11/05/2011] [Indexed: 12/17/2022] Open
Abstract
Physical interactions between cells and the extracellular matrix (ECM) guide directional migration by spatially controlling where cells form focal adhesions (FAs), which in turn regulate the extension of motile processes. Here we show that physical control of directional migration requires the FA scaffold protein paxillin. Using single-cell sized ECM islands to constrain cell shape, we found that fibroblasts cultured on square islands preferentially activated Rac and extended lamellipodia from corner, rather than side regions after 30 min stimulation with PDGF, but that cells lacking paxillin failed to restrict Rac activity to corners and formed small lamellipodia along their entire peripheries. This spatial preference was preceded by non-spatially constrained formation of both dorsal and lateral membrane ruffles from 5-10 min. Expression of paxillin N-terminal (paxN) or C-terminal (paxC) truncation mutants produced opposite, but complementary, effects on lamellipodia formation. Surprisingly, pax-/- and paxN cells also formed more circular dorsal ruffles (CDRs) than pax+ cells, while paxC cells formed fewer CDRs and extended larger lamellipodia even in the absence of PDGF. In a two-dimensional (2D) wound assay, pax-/- cells migrated at similar speeds to controls but lost directional persistence. Directional motility was rescued by expressing full-length paxillin or the N-terminus alone, but paxN cells migrated more slowly. In contrast, pax-/- and paxN cells exhibited increased migration in a three-dimensional (3D) invasion assay, with paxN cells invading Matrigel even in the absence of PDGF. These studies indicate that paxillin integrates physical and chemical motility signals by spatially constraining where cells will form motile processes, and thereby regulates directional migration both in 2D and 3D. These findings also suggest that CDRs may correspond to invasive protrusions that drive cell migration through 3D extracellular matrices.
Collapse
Affiliation(s)
- Julia E. Sero
- Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Charles K. Thodeti
- Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Akiko Mammoto
- Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Chris Bakal
- Dynamical Cell Systems Team, Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Sheila Thomas
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Donald E. Ingber
- Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts, United States of America
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, United States of America
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
40
|
Zebda N, Dubrovskyi O, Birukov KG. Focal adhesion kinase regulation of mechanotransduction and its impact on endothelial cell functions. Microvasc Res 2011; 83:71-81. [PMID: 21741394 DOI: 10.1016/j.mvr.2011.06.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 06/19/2011] [Accepted: 06/20/2011] [Indexed: 01/06/2023]
Abstract
Vascular endothelial cells lining the blood vessels form the interface between the bloodstream and the vessel wall and as such they are continuously subjected to shear and cyclic stress from the flowing blood in the lumen. Additional mechanical stimuli are also imposed on these cells in the form of substrate stiffness transmitted from the extracellular matrix components in the basement membrane, and additional mechanical loads imposed on the lung endothelium as the result of respiration or mechanical ventilation in clinical settings. Focal adhesions (FAs) are complex structures assembled at the abluminal endothelial plasma membrane which connect the extracellular filamentous meshwork to the intracellular cytoskeleton and hence constitute the ideal checkpoint capable of controlling or mediating transduction of bidirectional mechanical signals. In this review we focus on focal adhesion kinase (FAK), a component of FAs, which has been studied for a number of years with regards to its involvement in mechanotransduction. We analyzed the recent advances in the understanding of the role of FAK in the signaling cascade(s) initiated by various mechanical stimuli with particular emphasis on potential implications on endothelial cell functions.
Collapse
Affiliation(s)
- Noureddine Zebda
- Section of Pulmonary and Critical Care, Lung Injury Center, Department of Medicine, The University of Chicago, IL 60637, USA
| | | | | |
Collapse
|
41
|
St-Pierre J, Lysechko TL, Ostergaard HL. Hypophosphorylated and inactive Pyk2 associates with paxillin at the microtubule organizing center in hematopoietic cells. Cell Signal 2011; 23:718-30. [DOI: 10.1016/j.cellsig.2010.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/13/2010] [Accepted: 12/21/2010] [Indexed: 10/18/2022]
|
42
|
Abstract
As materials technology and the field of tissue engineering advance, the role of cellular adhesive mechanisms, in particular, interactions with implantable devices, becomes more relevant in both research and clinical practice. A key tenet of medical device technology is to use the exquisite ability of biological systems to respond to the material surface or chemical stimuli in order to help to develop next-generation biomaterials. The focus of this review is on recent studies and developments concerning focal adhesion formation in osteoneogenesis, with an emphasis on the influence of synthetic constructs on integrin-mediated cellular adhesion and function.
Collapse
Affiliation(s)
- M J P Biggs
- Nanotechnology Center for Mechanics in Regenerative Medicine, Department of Applied Physics and Applied Mathematics, Columbia University, New York 10027, USA.
| | | |
Collapse
|
43
|
March ME, Long EO. β2 integrin induces TCRζ-Syk-phospholipase C-γ phosphorylation and paxillin-dependent granule polarization in human NK cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:2998-3005. [PMID: 21270398 DOI: 10.4049/jimmunol.1002438] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cytotoxic lymphocytes kill target cells through polarized release of the content of lytic granules at the immunological synapse. In human NK cells, signals for granule polarization and for degranulation can be uncoupled: Binding of β(2) integrin LFA-1 to ICAM is sufficient to induce polarization but not degranulation, whereas CD16 binding to IgG triggers unpolarized degranulation. In this study, we investigated the basis for this difference. IL-2-expanded human NK cells were stimulated by incubation with plate-bound ligands of LFA-1 (ICAM-1) and CD16 (human IgG). Surprisingly, LFA-1 elicited signals similar to those induced by CD16, including tyrosine phosphorylation of the TCR ζ-chain, tyrosine kinase Syk, and phospholipase C-γ. Whereas CD16 activated Ca(2+) mobilization and LAT phosphorylation, LFA-1 did not, but induced strong Pyk2 and paxillin phosphorylation. LFA-1-dependent granule polarization was blocked by inhibition of Syk, phospholipase C-γ, and protein kinase C, as well as by paxillin knockdown. Therefore, common signals triggered by CD16 and LFA-1 bifurcate to provide independent control of Ca(2+)-dependent degranulation and paxillin-dependent granule polarization.
Collapse
Affiliation(s)
- Michael E March
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | | |
Collapse
|
44
|
Cheng R, Shao MY, Yang H, Cheng L, Wang FM, Zhou XD, Hu T. The effect of lysophosphatidic acid and Rho-associated kinase patterning on adhesion of dental pulp cells. Int Endod J 2010; 44:2-8. [PMID: 21073482 DOI: 10.1111/j.1365-2591.2010.01773.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AIM To investigate the effects of lysophosphatidic acid (LPA) and the Rho/Rho-associated kinase (ROCK) pathway on adhesion of dental pulp cells (DPCs). METHODOLOGY Human DPCs were cultured ex vivo. After treatment of LPA and Y-27632, a specific ROCK inhibitor, changes in focal contacts (FCs) were examined by immunofluorescent staining. Activation of FCs proteins was examined by measuring tyrosine 397 phosphorylation of focal adhesion kinase (FAK) and paxillin using immunoblotting. The data were analysed by Student's t-test. RESULTS The immunofluorescent staining indicated LPA stimulation induced larger focal adhesion in the cell periphery, compared with the control. Inhibition of ROCK by Y-27632 decreased the formation of FCs markedly, even in the LPA-stimulated cells. LPA also increased the level of tyrosine phosphorylation of paxillin at 30min (P<0.05) and FAK at 5 and 30min (P<0.05). Furthermore, p-paxillin levels declined immediately after Y-27632 treatment and remained low at 5, 30, 60min. Y-27632 also suppressed the effects of LPA on p-paxillin and p-FAK at 5 and 30min (P<0.05). CONCLUSION LPA activated Rho and then subsequently activated ROCK, suggesting that LPA influences the FCs of DPCs by modulating tyrosine phosphorylation of FAK and paxillin via the Rho/ROCK pathway.
Collapse
Affiliation(s)
- R Cheng
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Kim MO, Lee YJ, Han HJ. Involvement of Cx43 phosphorylation in 5'-N-ethylcarboxamide-induced migration and proliferation of mouse embryonic stem cells. J Cell Physiol 2010; 224:187-94. [PMID: 20232318 DOI: 10.1002/jcp.22117] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite a lot of gap junction research, the complex connection between gap junction and cell proliferation remains an exciting area of investigation. Thus, we examined the effect of connexin 43 (Cx43) on the migration and proliferation of embryonic stem (ES) cells and its related signaling pathways following stimulation with the adenosine analogue 5'-N-ethylcarboxamide (NECA). NECA increased phosphorylation of Cx43 which was blocked by caffeine, a non-selective adenosine receptor antagonist. In experiment to measure the gap junctional intercellular communication, NECA blocked transfer of Lucifer yellow to neighboring cells in a scrape loading/dye transfer (SL/DT) assay. In addition, NECA-induced phosphorylation of phosphoinositide 3-kinase (PI3K)/Akt, protein kinase C (PKC), mitogen-activated protein kinases (MAPKs), and nuclear factor-kappa B (NF-kappaB) signal pathways. Inhibition of these signaling pathways reduced NECA-induced phosphorylation of Cx43. Moreover, NECA-treated cells demonstrated phosphorylation of Src, which was blocked by caffeine. In this experiment, a disruption of Cx43 using Cx43-specific small interfering RNA (siRNA) also enhanced Src phosphorylation. In a further study, phosphorylations of integrin beta1, focal adhesion kinase (FAK), and paxillin by NECA were restrained by caffeine as well as the Src blocker, PP2. Finally, we identified that NECA-stimulated cell migration and expressions of cell-cycle regulatory proteins [cyclin D1, cyclin-dependent kinase (CDK) 4, cyclin E, and CDK2]; these increases were inhibited by caffeine, or PP2. We conclude that NECA-stimulated Cx43 phosphorylation mediated by PI3K/Akt, PKC, MAPKs, and NF-kappaB, which subsequently stimulated cell migration and proliferation through Src, integrin beta1, FAK, and paxillin signal pathways.
Collapse
Affiliation(s)
- Mi Ok Kim
- Department of Veterinary Physiology, Biotherapy Human Resources Center (BK 21), College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | | | | |
Collapse
|
46
|
Hassona MDH, Abouelnaga ZA, Elnakish MT, Awad MM, Alhaj M, Goldschmidt-Clermont PJ, Hassanain H. Vascular hypertrophy-associated hypertension of profilin1 transgenic mouse model leads to functional remodeling of peripheral arteries. Am J Physiol Heart Circ Physiol 2010; 298:H2112-20. [PMID: 20400688 DOI: 10.1152/ajpheart.00016.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increased mechanical stress/hypertension in the vessel wall triggers the hypertrophic signaling pathway, resulting in structural remodeling of vasculature. Vascular hypertrophy of resistance vessels leads to reduced compliance and elevation of blood pressure. We showed before that increased expression of profilin1 protein in the medial layer of the aorta induces stress fiber formation, triggering the hypertrophic signaling resulting in vascular hypertrophy and, ultimately, hypertension in older mice. Our hypothesis is that profilin1 induced vascular hypertrophy in resistance vessels, which led to elevation of blood pressure, both of which contributed to the modulation of vascular function. Our results showed significant increases in the expression of alpha(1)- and beta(1)-integrins (280 + or - 6.3 and 325 + or - 7.4%, respectively) and the activation of the Rho/Rho-associated kinase (ROCK) II pathway (260 and 350%, respectively, P < 0.05) in profilin1 mesenteric arteries. The activation of Rho/ROCK led to the inhibition of endothelial nitric oxide synthase expression (39 + or - 5.4%; P < 0.05) and phosphorylation (35 + or - 4.5%; P < 0.05) but also an increase in myosin light chain 20 phosphorylation (372%, P < 0.05). There were also increases in hypertrophic signaling pathways in the mesenteric arteries of profilin1 mice such as phospho-extracellular signal-regulated kinase 1/2 and phospho-c-Jun NH(2)-terminal kinase (312.15 and 232.5%, respectively, P < 0.05). Functional analyses of mesenteric arteries toward the vasoactive drugs were assessed using wire-myograph and showed significant increases in the vascular responses of profilin1 mesenteric arteries toward phenylephrine, but significant decreases in response toward ROCK inhibitor Y-27632, ACh, sodium nitrite, and cytochalasin D. The changes in vascular responses in the mesenteric arteries of profilin1 mice are due to vascular hypertrophy and the elevation of blood pressure in the profilin1 transgenic mice.
Collapse
Affiliation(s)
- Mohamed D H Hassona
- Dept. of Anesthesiology and Dorothy M. Davis Heart & Lung Research Institute, The Ohio State Univ., 460 West 12th Ave., Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Chen HH, Chen CW, Chang YY, Shen TL, Hsu CH. Preliminary crystallographic characterization of the Grb2 SH2 domain in complex with a FAK-derived phosphotyrosyl peptide. Acta Crystallogr Sect F Struct Biol Cryst Commun 2010; 66:195-7. [PMID: 20124721 DOI: 10.1107/s1744309109053184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 12/10/2009] [Indexed: 11/10/2022]
Abstract
Growth factor receptor-bound protein 2 (Grb2) is an adaptor protein with a single SH2 domain that specifically binds to focal adhesion kinase (FAK) when residue Tyr925 of FAK is phosphorylated. The Grb2-FAK interaction is associated with cellular integrin-activated signal transduction events leading to the activation of the Ras-MAPK pathway. Crystals of the Grb2 SH2 domain in complex with a phosphopeptide corresponding to residues 921-930 of FAK have been obtained using the sitting-drop vapour-diffusion technique. The crystals belonged to space group P3(1)21, with unit-cell parameters a = b = 102.7, c = 127.6 A, alpha = beta = 90.0, gamma = 120.0 degrees . A diffraction data set was collected from a flash-cooled crystal at 100 K to 2.49 A resolution using synchrotron radiation. Structure determination by molecular replacement and analysis of the detailed structure of the complex are currently in progress.
Collapse
Affiliation(s)
- Hsiao Hsin Chen
- Department of Agricultural Chemistry, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
48
|
Cai GQ, Zheng A, Tang Q, White ES, Chou CF, Gladson CL, Olman MA, Ding Q. Downregulation of FAK-related non-kinase mediates the migratory phenotype of human fibrotic lung fibroblasts. Exp Cell Res 2010; 316:1600-9. [PMID: 20109444 DOI: 10.1016/j.yexcr.2010.01.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 01/11/2023]
Abstract
Fibroblast migration plays an important role in the normal wound healing process; however, dysregulated cell migration may contribute to the progressive formation of fibrotic lesions in the diseased condition. To examine the role of focal-adhesion-kinase (FAK)-related non-kinase (FRNK) in regulation of fibrotic lung fibroblast migration, we examined cell migration, FRNK expression, and activation of focal adhesion kinase (FAK) and Rho GTPase (Rho and Rac) in primary lung fibroblasts derived from both idiopathic pulmonary fibrosis (IPF) patients and normal human controls. Fibrotic (IPF) lung fibroblasts have increased cell migration when compared to control human lung fibroblasts. FRNK expression is significantly reduced in IPF lung fibroblasts, while activation of FAK, Rho and Rac is increased in IPF lung fibroblasts. Endogenous FRNK expression is inversely correlated with FAK activation and cell migration rate in IPF lung fibroblasts. Forced exogenous FRNK expression abrogates the increased cell migration, and blocked the activation of FAK and Rho GTPase (Rho and Rac), in IPF lung fibroblasts. These data for the first time provide evidence that downregulation of endogenous FRNK plays a role in promoting cell migration through FAK and Rho GTPase in fibrotic IPF lung fibroblasts.
Collapse
Affiliation(s)
- Guo-qiang Cai
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Identification and functional characterization of paxillin as a target of protein tyrosine phosphatase receptor T. Proc Natl Acad Sci U S A 2010; 107:2592-7. [PMID: 20133777 DOI: 10.1073/pnas.0914884107] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein tyrosine phosphatase receptor-type T (PTPRT) is the most frequently mutated tyrosine phosphatase in human cancers. However, the cell signaling pathways regulated by PTPRT largely remain to be elucidated. Here, we show that paxillin is a direct substrate of PTPRT and that PTPRT specifically regulates paxillin phosphorylation at tyrosine residue 88 (Y88) in colorectal cancer (CRC) cells. We engineered CRC cells homozygous for a paxillin Y88F knock-in mutant and found that these cells exhibit significantly reduced cell migration and impaired anchorage-independent growth, fail to form xenograft tumors in nude mice, and have decreased phosphorylation of p130CAS, SHP2, and AKT. PTPRT knockout mice that we generated exhibit increased levels of colonic paxillin phosphorylation at residue Y88 and are highly susceptible to carcinogen azoxymethane-induced colon tumor, providing critical in vivo evidence that PTPRT normally functions as a tumor suppressor. Moreover, similarly increased paxillin pY88 is also found as a common feature of human colon cancers. These studies reveal an important signaling pathway that plays a critical role in colorectal tumorigenesis.
Collapse
|
50
|
Mei L, Xiong WC. FAK interaction with MBD2: A link from cell adhesion to nuclear chromatin remodeling? Cell Adh Migr 2010; 4:77-80. [PMID: 19949307 DOI: 10.4161/cam.4.1.10343] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cell adhesion, migration, proliferation and differentiation are tightly linked and coordinated cellular processes. Cell adhesion dependent gene expression is believed to contribute to such coordination. Focal adhesion kinase (FAK) and its related protein, PYK2 (proline rich tyrosine kinase 2), are a major family of cell adhesion activated tyrosine kinases that play important roles in these cellular processes. Whereas FAK or PYK2 is known to be a scaffold protein, recruiting many cytoplasmic proteins into the focal adhesion complex and regulating focal adhesion turnover and cell migration, how FAK or PYK2 links to the nuclei and regulates gene expression remain largely unclear. We recently report a new signaling of FAK in regulating heterochromatin remodeling by its interaction with MBD2 (Methyl CpG binding domain protein 2), which may underlie FAK regulation of myogenin expression and muscle differentiation. Two insights have been obtained through the analysis of FAK-MBD2 interaction. The interaction appears to be sufficient, but not necessary, for FAK translocation into or maintaining in the nucleus. The nuclear FAK-MBD2 complexes cause altered heterochromatin organization and decreased MBD2 association with HDAC1 (histone deacetylase complex 1) and methyl CpG site in the myogenin promoter, thus, inducing myogenin expression. These results demonstrate a new mechanism underlying FAK regulation of gene expression, and suggest a potential link between cell adhesion and cell differentiation.
Collapse
Affiliation(s)
- Lin Mei
- Institute of Molecular Medicine & Genetics and Department of Neurology, Medical College of Georgia, Augusta, GA, USA
| | | |
Collapse
|