1
|
Brock K, Alpha KM, Brennan G, De Jong EP, Luke E, Turner CE. A comparative analysis of paxillin and Hic-5 proximity interactomes. Cytoskeleton (Hoboken) 2025; 82:12-31. [PMID: 38801098 PMCID: PMC11599474 DOI: 10.1002/cm.21878] [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: 02/14/2024] [Revised: 04/18/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Abstract
Focal adhesions serve as structural and signaling hubs, facilitating bidirectional communication at the cell-extracellular matrix interface. Paxillin and the related Hic-5 (TGFβ1i1) are adaptor/scaffold proteins that recruit numerous structural and regulatory proteins to focal adhesions, where they perform both overlapping and discrete functions. In this study, paxillin and Hic-5 were expressed in U2OS osteosarcoma cells as biotin ligase (BioID2) fusion proteins and used as bait proteins for proximity-dependent biotinylation in order to directly compare their respective interactomes. The fusion proteins localized to both focal adhesions and the centrosome, resulting in biotinylation of components of each of these structures. Biotinylated proteins were purified and analyzed by mass spectrometry. The list of proximity interactors for paxillin and Hic-5 comprised numerous shared core focal adhesion proteins that likely contribute to their similar functions in cell adhesion and migration, as well as proteins unique to paxillin and Hic-5 that have been previously localized to focal adhesions, the centrosome, or the nucleus. Western blotting confirmed biotinylation and enrichment of FAK and vinculin, known interactors of Hic-5 and paxillin, as well as several potentially unique proximity interactors of Hic-5 and paxillin, including septin 7 and ponsin, respectively. Further investigation into the functional relationship between the unique interactors and Hic-5 or paxillin may yield novel insights into their distinct roles in cell migration.
Collapse
Affiliation(s)
- Katia Brock
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Kyle M. Alpha
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Grant Brennan
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Ebbing P. De Jong
- Proteomics Core facility, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Elizabeth Luke
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Christopher E. Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
2
|
Zhao Y, Lykov N, Tzeng C. Talin‑1 interaction network in cellular mechanotransduction (Review). Int J Mol Med 2022; 49:60. [PMID: 35266014 PMCID: PMC8930095 DOI: 10.3892/ijmm.2022.5116] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
The mechanical signals within the extracellular matrix (ECM) regulate cell growth, proliferation and differentiation, and integrins function as the hub between the ECM and cellular actin. Focal adhesions (FAs) are multi‑protein, integrin‑containing complexes, acting as tension‑sensing anchoring points that bond cells to the extracellular microenvironment. Talin‑1 serves as the central protein of FAs that participates in the activation of integrins and connects them with the actin cytoskeleton. As a cytoplasmic protein, Talin‑1 consists of a globular head domain and a long rod comprised of a series of α‑helical bundles. The unique structure of the Talin‑1 rod domain permits folding and unfolding in response to the mechanical stress, revealing various binding sites. Thus, conformation changes of the Talin‑1 rod domain enable the cell to convert mechanical signals into chemical through multiple signaling pathways. The present review discusses the binding partners of Talin‑1, their interactions, effects on the cellular processes, and their possible roles in diseases.
Collapse
Affiliation(s)
- Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu 211800, P.R. China
| | - Nikita Lykov
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu 211800, P.R. China
| | - Chimeng Tzeng
- Translational Medicine Research Center-Key Laboratory for Cancer T-Cell Theragnostic and Clinical Translation, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361005, P.R. China
- Xiamen Chang Gung Hospital Medical Research Center, Xiamen, Fujian 361005, P.R. China
| |
Collapse
|
3
|
Nguyen HTT, Xu Z, Shi X, Liu S, Schulte ML, White GC, Ma YQ. Paxillin binding to the PH domain of kindlin-3 in platelets is required to support integrin αIIbβ3 outside-in signaling. J Thromb Haemost 2021; 19:3126-3138. [PMID: 34411430 PMCID: PMC9080902 DOI: 10.1111/jth.15505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/05/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Kindlin-3 is essential for supporting the bidirectional signaling of integrin αIIbβ3 in platelets by bridging the crosstalk between integrin αIIbβ3 and the cytoplasmic signaling adaptors. OBJECTIVE In this study, we identified a previously unrecognized paxillin binding site in the pleckstrin homology (PH) domain of kindlin-3 and verified its functional significance. METHODS Structure-based approaches were employed to identify the paxillin binding site in the PH domain of kindlin-3. In addition, the bidirectional signaling of integrin αIIbβ3 were evaluated in both human and mouse platelets. RESULTS In brief, we found that a β1-β2 loop in the PH domain of kindlin-3, an important part of the canonical membrane phospholipid binding pocket, was also involved in mediating paxillin interaction. Interestingly, the binding sites of paxillin and membrane phospholipids in the PH domain of kindlin-3 were mutually exclusive. Specific disruption of paxillin binding to the PH domain by point mutations inhibited platelet spreading on immobilized fibrinogen while having no inhibition on soluble fibrinogen binding to stimulated platelets. In addition, a membrane-permeable peptide derived from the β1-β2 loop in the PH domain of kindlin-3 was capable of inhibiting platelet spreading and clot retraction, but it had no effect on soluble fibrinogen binding to platelets and platelet aggregation. Treatment with this peptide significantly reduced thrombus formation in mice. CONCLUSION Taken together, these findings suggest that interaction between paxillin and the PH domain of kindlin-3 plays an important role in supporting integrin αIIbβ3 outside-in signaling in platelets, thus providing a novel antithrombotic target.
Collapse
Affiliation(s)
| | - Zhen Xu
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Xiaofeng Shi
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- The second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shuzhen Liu
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Gilbert C. White
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Department of Biochemistry, Medical College of Milwaukee, Milwaukee, Wisconsin, USA
| | - Yan-Qing Ma
- Versiti Blood Research Institute, Milwaukee, Wisconsin, USA
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
- Department of Biochemistry, Medical College of Milwaukee, Milwaukee, Wisconsin, USA
| |
Collapse
|
4
|
Bello IO, Alrabeeah MA, AlFouzan NF, Alabdulaali NA, Nieminen P. FAK, paxillin, and PI3K in ameloblastoma and adenomatoid odontogenic tumor. Clin Oral Investig 2020; 25:1559-1567. [PMID: 32681423 DOI: 10.1007/s00784-020-03465-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/14/2020] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Integrins function to bind cells to extracellular matrix in tissues, which triggers downstream signaling cascades that are important in cell survival, proliferation, cytokine activation, and cytoskeleton reorganization. These processes also play significant roles in neoplasms. This work aimed to investigate the pattern of expression of FAK, paxillin, and PI3K in ameloblastoma and adenomatoid odontogenic tumor (AOT). MATERIALS AND METHODS Immunohistochemistry was used to study FAK, paxillin, and PI3K in 45 ameloblastomas (32 conventional, 12 unicystic, and 1 peripheral types), 7 AOTs, and two developing human teeth. RESULTS Weak expression of FAK was seen in all AOT cases, while ameloblastoma had varying expression patterns, mostly strong to weak. The pattern of expression of paxillin and PI3K was relatively similar in both tumor types. In the dental germ, FAK and paxillin stained all the enamel organ components, while PI3K stained strongly the inner enamel epithelium. Stromal expression of FAK was not found to be useful in differentiating between tumors or tumor classes. CONCLUSION The expression of the proteins in the enamel organ suggests that their signaling may be important in odontogenesis. While some ameloblastomas strongly expressed FAK, all cases of AOT had weak signals suggesting low presence and phosphorylating activity of FAK in the latter. CLINICAL RELEVANCE A subset of FAK-positive ameloblastoma (as well as their malignant or metastasizing counterparts) which may have relatively aggressive behavior may be candidates for drug targeting of FAK as an additional management option.
Collapse
Affiliation(s)
- Ibrahim O Bello
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, King Saud University, Riyadh, 11545, Saudi Arabia.
- Department of Pathology, University of Helsinki, Helsinki, Finland.
| | | | | | | | - Pentti Nieminen
- Medical Informatics and Statistics Research Group, University of Oulu, Oulu, Finland
| |
Collapse
|
5
|
Gulvady AC, Forsythe IJ, Turner CE. Hic-5 regulates Src-induced invadopodia rosette formation and organization. Mol Biol Cell 2019; 30:1298-1313. [PMID: 30893012 PMCID: PMC6724605 DOI: 10.1091/mbc.e18-10-0629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fibroblasts transformed by the proto-oncogene Src form individual invadopodia that can spontaneously self-organize into large matrix-degrading superstructures called rosettes. However, the mechanisms by which the invadopodia can spatiotemporally reorganize their architecture is not well understood. Here, we show that Hic-5, a close relative of the scaffold protein paxillin, is essential for the formation and organization of rosettes in active Src-transfected NIH3T3 fibroblasts and cancer-associated fibroblasts. Live cell imaging, combined with domain-mapping analysis of Hic-5, identified critical motifs as well as phosphorylation sites that are required for the formation and dynamics of rosettes. Using pharmacological inhibition and mutant expression, we show that FAK kinase activity, along with its proximity to and potential interaction with the LD2,3 motifs of Hic-5, is necessary for rosette formation. Invadopodia dynamics and their coalescence into rosettes were also dependent on Rac1, formin, and myosin II activity. Superresolution microscopy revealed the presence of formin FHOD1 and INF2-mediated unbranched radial F-actin fibers emanating from invadopodia and rosettes, which may facilitate rosette formation. Collectively, our data highlight a novel role for Hic-5 in orchestrating the organization of invadopodia into higher-order rosettes, which may promote the localized matrix degradation necessary for tumor cell invasion.
Collapse
Affiliation(s)
- Anushree C Gulvady
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Ian J Forsythe
- 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
|
6
|
Goreczny GJ, Forsythe IJ, Turner CE. Hic-5 regulates fibrillar adhesion formation to control tumor extracellular matrix remodeling through interaction with tensin1. Oncogene 2018; 37:1699-1713. [PMID: 29348458 PMCID: PMC5876083 DOI: 10.1038/s41388-017-0074-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/02/2017] [Accepted: 11/19/2017] [Indexed: 01/26/2023]
Abstract
The linearization of the stromal extracellular matrix (ECM) by cancer associated fibroblasts (CAFs) facilitates tumor cell growth and metastasis. However, the mechanism by which the ECM is remodeled is not fully understood. Hic-5 (TGFβ1i1), a focal adhesion scaffold protein, has previously been reported to be crucial for stromal ECM deposition and remodeling in vivo. Herein we show that CAFs lacking Hic-5 exhibit a significant reduction in the ability to form fibrillar adhesions, a specialized form of focal adhesion that promote fibronectin fibrillogenesis. Hic-5 was found to promote fibrillar adhesion formation through a newly characterized interaction with tensin1. Furthermore, Src dependent phosphorylation of Hic-5 facilitated the interaction with tensin1 to prevent β1 integrin internalization and trafficking to the lysosome. The interaction between Hic-5 and tensin1 was mechanosensitive, promoting fibrillar adhesion formation and fibronectin fibrillogenesis in a rigidity dependent fashion. Importantly, this Src dependent mechanism was conserved in three-dimensional (3D) ECM environments. Immunohistochemistry of tensin1 showed enrichment in CAFs in vivo, which was abrogated upon deletion of Hic-5. Interestingly, elevated Hic-5 expression correlates with reduced distant metastasis free survival in patients with basal-like, HER2+ and grade 3 tumors. Thus, we have identified Hic-5 as a crucial regulator of ECM remodeling in CAFs by promoting fibrillar adhesion formation through a novel interaction with tensin1.
Collapse
Affiliation(s)
- Gregory J Goreczny
- Department of Cell & Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ian J Forsythe
- Department of Cell & Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Christopher E Turner
- Department of Cell & Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
7
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
8
|
Sheta R, Wang ZQ, Bachvarova M, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Gobeil S, Morin C, Macdonald E, Vanderhyden B, Bachvarov D. Hic-5 regulates epithelial to mesenchymal transition in ovarian cancer cells in a TGFβ1-independent manner. Oncotarget 2017; 8:82506-82530. [PMID: 29137281 PMCID: PMC5669907 DOI: 10.18632/oncotarget.19714] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/17/2017] [Indexed: 01/01/2023] Open
Abstract
The molecular basis of epithelial ovarian cancer (EOC) dissemination is still poorly understood. We have previously identified the hydrogen peroxide-inducible clone-5 (Hic-5) gene as hypomethylated in high-grade (HG) serous EOC tumors, compared to normal ovarian tissues. Hic-5 is a focal adhesion scaffold protein and has been primarily studied for its role as a key mediator of TGF-β–induced epithelial-to-mesenchymal transition (EMT) in epithelial cells of both normal and malignant origin; however, its role in EOC has been never investigated. Here we demonstrate that Hic-5 is overexpressed in advanced EOC, and that Hic-5 is upregulated upon TGFβ1 treatment in the EOC cell line with epithelial morphology (A2780s), associated with EMT induction. However, ectopic expression of Hic-5 in A2780s cells induces EMT independently of TGFβ1, accompanied with enhancement of cellular proliferation rate and migratory/invasive capacity and increased resistance to chemotherapeutic drugs. Moreover, Hic-5 knockdown in the EOC cells with mesenchymal morphology (SKOV3) was accompanied by induction of mesenchymal-to-epithelial transition (MET), followed by a reduction of their proliferative, migratory/invasive capacity, and increased drugs sensitivity in vitro, as well as enhanced tumor cell colonization and metastatic growth in vivo. The modulation of Hic-5 expression in EOC cells resulted in altered regulation of numerous EMT-related canonical pathways and was indicative for a possible role of Hic-5 in controlling EMT through a RhoA/ROCK mediated mechanism. To our knowledge, this is the first report examining the role of Hic-5 in EOC, and its role in maintaining the mesenchymal phenotype of EOC cells independently of exogenous TGFβ1 treatment.
Collapse
Affiliation(s)
- Razan Sheta
- Department of Molecular Medicine, Université Laval, Québec, Québec, Canada.,Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada
| | - Zhi-Qiang Wang
- Department of Molecular Medicine, Université Laval, Québec, Québec, Canada.,Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada
| | - Magdalena Bachvarova
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada
| | - Marie Plante
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, Québec, Canada
| | - Jean Gregoire
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, Québec, Canada
| | - Marie-Claude Renaud
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, Québec, Canada
| | - Alexandra Sebastianelli
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, Québec, Canada
| | - Stephane Gobeil
- Department of Molecular Medicine, Université Laval, Québec, Québec, Canada.,Centre de recherche du CHU de Québec, CHUL, Québec, Québec, Canada
| | - Chantale Morin
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada
| | - Elizabeth Macdonald
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Barbara Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dimcho Bachvarov
- Department of Molecular Medicine, Université Laval, Québec, Québec, Canada.,Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec, Québec, Canada
| |
Collapse
|
9
|
Bengtsen M, Sørensen L, Aabel L, Ledsaak M, Matre V, Gabrielsen OS. The adaptor protein ARA55 and the nuclear kinase HIPK1 assist c-Myb in recruiting p300 to chromatin. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:751-760. [DOI: 10.1016/j.bbagrm.2017.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 02/01/2023]
|
10
|
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: 1.1] [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
|
11
|
Dave JM, Abbey CA, Duran CL, Seo H, Johnson GA, Bayless KJ. Hic-5 mediates the initiation of endothelial sprouting by regulating a key surface metalloproteinase. J Cell Sci 2016; 129:743-56. [PMID: 26769900 DOI: 10.1242/jcs.170571] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 01/05/2016] [Indexed: 12/16/2022] Open
Abstract
During angiogenesis, endothelial cells must coordinate matrix proteolysis with migration. Here, we tested whether the focal adhesion scaffold protein Hic-5 (also known as TGFB1I1) regulated endothelial sprouting in three dimensions. Hic-5 silencing reduced endothelial sprouting and lumen formation, and sprouting defects were rescued by the return of Hic-5 expression. Pro-angiogenic factors enhanced colocalization and complex formation between membrane type-1 matrix metalloproteinase (MT1-MMP, also known as MMP14) and Hic-5, but not between paxillin and MT1-MMP. The LIM2 and LIM3 domains of Hic-5 were necessary and sufficient for Hic-5 to form a complex with MT1-MMP. The degree of interaction between MT1-MMP and Hic-5 and the localization of the complex within detergent-resistant membrane fractions were enhanced during endothelial sprouting, and Hic-5 depletion lowered the surface levels of MT1-MMP. In addition, we observed that loss of Hic-5 partially reduced complex formation between MT1-MMP and focal adhesion kinase (FAK, also known as PTK2), suggesting that Hic-5 bridges MT1-MMP and FAK. Finally, Hic-5 LIM2-LIM3 deletion mutants reduced sprout initiation. Hic-5, MT1-MMP and FAK colocalized in angiogenic vessels during porcine pregnancy, supporting that this complex assembles during angiogenesis in vivo. Collectively, Hic-5 appears to enhance complex formation between MT1-MMP and FAK in activated endothelial cells, which likely coordinates matrix proteolysis and cell motility.
Collapse
Affiliation(s)
- Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Colette A Abbey
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Heewon Seo
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Gregory A Johnson
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
12
|
Pattabiraman PP, Rao PV. Hic-5 Regulates Actin Cytoskeletal Reorganization and Expression of Fibrogenic Markers and Myocilin in Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2015; 56:5656-69. [PMID: 26313302 DOI: 10.1167/iovs.15-17204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To explore the role of inducible focal adhesion (FA) protein Hic-5 in actin cytoskeletal reorganization, FA formation, fibrogenic activity, and expression of myocilin in trabecular meshwork (TM) cells. METHODS Using primary cultures of human TM (HTM) cells, the effects of various external factors on Hic-5 protein levels, as well as the effects of recombinant Hic-5 and Hic-5 small interfering RNA (siRNA) on actin cytoskeleton, FAs, myocilin, α-smooth muscle actin (αSMA), and collagen-1 were determined by immunofluorescence and immunoblot analyses. RESULTS Hic-5 distributes discretely to the FAs in HTM cells and throughout the TM and Schlemm's canal of the human aqueous humor (AH) outflow pathway. Transforming growth factor-β2 (TGF-β2), endothelin-1, lysophosphatidic acid, hydrogen peroxide, and RhoA significantly increased Hic-5 protein levels in HTM cells in association with reorganization of actin cytoskeleton and FAs. While recombinant Hic-5 induced actin stress fibers, FAs, αv integrin redistribution to the FAs, increased levels of αSMA, collagen-1, and myocilin, Hic-5 siRNA suppressed most of these responses in HTM cells. Hic-5 siRNA also suppressed TGF-β2-induced fibrogenic activity and dexamethasone-induced myocilin expression in HTM cells. CONCLUSIONS Taken together, these results reveal that Hic-5, whose levels were increased by various external factors implicated in elevated intraocular pressure, induces actin cytoskeletal reorganization, FAs, expression of fibrogenic markers, and myocilin in HTM cells. These characteristics of Hic-5 in TM cells indicate its importance in regulation of AH outflow through the TM in both normal and glaucomatous eyes.
Collapse
Affiliation(s)
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States 2Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
13
|
Walkiewicz KW, Girault JA, Arold ST. How to awaken your nanomachines: Site-specific activation of focal adhesion kinases through ligand interactions. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 119:60-71. [PMID: 26093249 DOI: 10.1016/j.pbiomolbio.2015.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 06/07/2015] [Accepted: 06/14/2015] [Indexed: 01/12/2023]
Abstract
The focal adhesion kinase (FAK) and the related protein-tyrosine kinase 2-beta (Pyk2) are highly versatile multidomain scaffolds central to cell adhesion, migration, and survival. Due to their key role in cancer metastasis, understanding and inhibiting their functions are important for the development of targeted therapy. Because FAK and Pyk2 are involved in many different cellular functions, designing drugs with partial and function-specific inhibitory effects would be desirable. Here, we summarise recent progress in understanding the structural mechanism of how the tug-of-war between intramolecular and intermolecular interactions allows these protein 'nanomachines' to become activated in a site-specific manner.
Collapse
Affiliation(s)
- Katarzyna W Walkiewicz
- King Abdullah University of Science and Technology (KAUST), Division of Biological and Environmental Sciences and Engineering, Computational Bioscience Research Center (CBRC), Thuwal, Saudi Arabia
| | - Jean-Antoine Girault
- Inserm, UMR-S 839, F-75005 Paris, France; Université Pierre & Marie Curie (UPMC), Sorbonne Universités, F-75005 Paris, France; Institut du Fer à Moulin, F-75005 Paris, France
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Division of Biological and Environmental Sciences and Engineering, Computational Bioscience Research Center (CBRC), Thuwal, Saudi Arabia.
| |
Collapse
|
14
|
Chodankar R, Wu DY, Gerke DS, Stallcup MR. Selective coregulator function and restriction of steroid receptor chromatin occupancy by Hic-5. Mol Endocrinol 2015; 29:716-29. [PMID: 25763609 DOI: 10.1210/me.2014-1403] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Steroid receptors (SRs) bind specific DNA regulatory sequences, thereby activating and repressing gene expression. We previously showed that transcriptional coregulator Hic-5 facilitates glucocorticoid regulation of some genes but blocks glucocorticoid regulation of others. Here, in a genome-wide analysis, Hic-5 depletion dramatically increased the global number of sites occupied by glucocorticoid receptor (GR) α (the major GR isoform), and many binding sites blocked by Hic-5 were associated with genes for which Hic-5 also blocked glucocorticoid-regulated expression. Hic-5 had similar effects on GRγ (a splice variant of GRα) and estrogen receptor α (ERα), facilitating hormonal regulation of some genes and blocking hormonal regulation of others. As with GRα, Hic-5 blocking of hormonal gene regulation mediated by GRγ and ERα was associated with blocking of GRγ and ERα occupancy at nearby sites. Hic-5 supported hormonal regulation of many more genes for GRα than for GRγ or ERα and thus exhibited selective coregulator functions for different SRs. In contrast, the number of Hic-5-blocked genes was similar for all 3 SRs. In addition to classic coregulator activity, Hic-5 influences the genomic occupancy of multiple SRs and thereby blocks some aspects of hormonal regulation. Thus, Hic-5, because of its tissue-specific expression, could contribute to tissue-specific genomic occupancy and gene regulation by SRs.
Collapse
Affiliation(s)
- Rajas Chodankar
- Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089-9176
| | | | | | | |
Collapse
|
15
|
Ohanian J, Pieri M, Ohanian V. Non-receptor tyrosine kinases and the actin cytoskeleton in contractile vascular smooth muscle. J Physiol 2014; 593:3807-14. [PMID: 25433074 DOI: 10.1113/jphysiol.2014.284174] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/14/2014] [Indexed: 01/01/2023] Open
Abstract
The contractility of vascular smooth muscle cells within the walls of arteries is regulated by mechanical stresses and vasoactive signals. Transduction of these diverse stimuli into a cellular response occurs through many different mechanisms, one being reorganisation of the actin cytoskeleton. In addition to a structural role in maintaining cellular architecture it is now clear that the actin cytoskeleton of contractile vascular smooth muscle cells is a dynamic structure reacting to changes in the cellular environment. Equally clear is that disrupting the cytoskeleton or interfering with its rearrangement, has profound effects on artery contractility. The actin cytoskeleton associates with dense plaques, also called focal adhesions, at the plasma membrane of smooth muscle cells. Vasoconstrictors and mechanical stress induce remodelling of the focal adhesions, concomitant with cytoskeletal reorganisation. Recent work has shown that non-receptor tyrosine kinases and tyrosine phosphorylation of focal adhesion proteins such as paxillin and Hic-5 are important for actin cytoskeleton and focal adhesion remodelling and contraction.
Collapse
Affiliation(s)
- Jacqueline Ohanian
- Institute of Cardiovascular Sciences, Manchester Academic Health Services Centre, University of Manchester, Manchester, UK
| | - Maria Pieri
- Institute of Cardiovascular Sciences, Manchester Academic Health Services Centre, University of Manchester, Manchester, UK
| | - Vasken Ohanian
- Institute of Cardiovascular Sciences, Manchester Academic Health Services Centre, University of Manchester, Manchester, UK
| |
Collapse
|
16
|
Leach DA, Need EF, Trotta AP, Grubisha MJ, DeFranco DB, Buchanan G. Hic-5 influences genomic and non-genomic actions of the androgen receptor in prostate myofibroblasts. Mol Cell Endocrinol 2014; 384:185-99. [PMID: 24440747 DOI: 10.1016/j.mce.2014.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/27/2013] [Accepted: 01/03/2014] [Indexed: 01/31/2023]
Abstract
There is extensive knowledge of androgen receptor (AR) signaling in cancer cells, but less regarding androgen action in stromal cells of the tumor microenvironment. We report here the genome-wide effects of a stromal cell specific molecular adapter and AR coregulator, hydrogen peroxide-inducible gene 5 (Hic-5/TGFB1I1), on AR function in prostate myofibroblasts. Following androgen stimulation, Hic-5 rapidly translocates to the nucleus, coincident with increased phosphorylation of focal adhesion kinase. As a coregulator, Hic-5 acted to amplify or inhibit regulation of approximately 50% of AR target genes, affected androgen regulation of growth, cell adhesion, motility and invasion. These data suggest Hic-5 as a transferable adaptor between focal adhesions and the nucleus of prostate myofibroblasts, where it acts a key mediator of the specificity and sensitivity of AR signaling. We propose a model in which Hic-5 coordinates AR signaling with adhesion and extracellular matrix contacts to regulate cell behavior in the tumor microenvironment.
Collapse
Affiliation(s)
- Damien A Leach
- Cancer Biology Group, The Basil Hetzel Institute for Translational Health Research, School of Medicine, University of Adelaide, SA, Australia
| | - Eleanor F Need
- Cancer Biology Group, The Basil Hetzel Institute for Translational Health Research, School of Medicine, University of Adelaide, SA, Australia
| | - Andrew P Trotta
- Cancer Biology Group, The Basil Hetzel Institute for Translational Health Research, School of Medicine, University of Adelaide, SA, Australia
| | - Melanie J Grubisha
- School of Medicine, Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA, USA
| | - Donald B DeFranco
- School of Medicine, Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA, USA
| | - Grant Buchanan
- Cancer Biology Group, The Basil Hetzel Institute for Translational Health Research, School of Medicine, University of Adelaide, SA, Australia.
| |
Collapse
|
17
|
Han SJ, O'Malley BW. The dynamics of nuclear receptors and nuclear receptor coregulators in the pathogenesis of endometriosis. Hum Reprod Update 2014; 20:467-84. [PMID: 24634322 DOI: 10.1093/humupd/dmu002] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Endometriosis is defined as the colonization and growth of endometrial tissue at anatomic sites outside the uterine cavity. Up to 15% of reproductive-aged women in the USA suffer from painful symptoms of endometriosis, such as infertility, pelvic pain, menstrual cycle abnormalities and increased risk of certain cancers. However, many of the current clinical treatments for endometriosis are not sufficiently effective and yield unacceptable side effects. There is clearly an urgent need to identify new molecular mechanisms that critically underpin the initiation and progression of endometriosis in order to develop more specific and effective therapeutics which lack the side effects of current therapies. The aim of this review is to discuss how nuclear receptors (NRs) and their coregulators promote the progression of endometriosis. Understanding the pathogenic molecular mechanisms for the genesis and maintenance of endometriosis as modulated by NRs and coregulators can reveal new therapeutic targets for alternative endometriosis treatments. METHODS This review was prepared using published gene expression microarray data sets obtained from patients with endometriosis and published literature on NRs and their coregulators that deal with endometriosis progression. Using the above observations, our current understanding of how NRs and NR coregulators are involved in the progression of endometriosis is summarized. RESULTS Aberrant levels of NRs and NR coregulators in ectopic endometriosis lesions are associated with the progression of endometriosis. As an example, endometriotic cell-specific alterations in gene expression are correlated with a differential methylation status of the genome compared with the normal endometrium. These differential epigenetic regulations can generate favorable cell-specific NR and coregulator milieus for endometriosis progression. Genetic alterations, such as single nucleotide polymorphisms and insertion/deletion polymorphisms of NR and coregulator genes, are frequently detected in ectopic lesions compared with the normal endometrium. These genetic variations impart new molecular properties to NRs and coregulators to increase their capacity to stimulate progression of endometriosis. Finally, post-translational modifications of NR coregulators, such as proteolytic processing, generate endometriosis-specific isoforms. Compared with the unmodified coregulators, these coregulator isoforms have unique functions that enhance the pathogenesis of endometriosis. CONCLUSIONS Epigenetic/genetic variations and posttranslational modifications of NRs and coregulators alter their original function so that they become potent 'drivers' of endometriosis progression.
Collapse
Affiliation(s)
- Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
18
|
LIM domains target actin regulators paxillin and zyxin to sites of stress fiber strain. PLoS One 2013; 8:e69378. [PMID: 23990882 PMCID: PMC3749209 DOI: 10.1371/journal.pone.0069378] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/08/2013] [Indexed: 01/25/2023] Open
Abstract
Contractile actomyosin stress fibers are critical for maintaining the force balance between the interior of the cell and its environment. Consequently, the actin cytoskeleton undergoes dynamic mechanical loading. This results in spontaneous, stochastic, highly localized strain events, characterized by thinning and elongation within a discrete region of stress fiber. Previous work showed the LIM-domain adaptor protein, zyxin, is essential for repair and stabilization of these sites. Using live imaging, we show paxillin, another LIM-domain adaptor protein, is also recruited to stress fiber strain sites. Paxillin recruitment to stress fiber strain sites precedes zyxin recruitment. Zyxin and paxillin are each recruited independently of the other. In cells lacking paxillin, actin recovery is abrogated, resulting in slowed actin recovery and increased incidence of catastrophic stress fiber breaks. For both paxillin and zyxin, the LIM domains are necessary and sufficient for recruitment. This work provides further evidence of the critical role of LIM-domain proteins in responding to mechanical stress in the actin cytoskeleton.
Collapse
|
19
|
Deakin NO, Pignatelli J, Turner CE. Diverse roles for the paxillin family of proteins in cancer. Genes Cancer 2012; 3:362-70. [PMID: 23226574 DOI: 10.1177/1947601912458582] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The paxillin family of intracellular scaffold proteins includes paxillin, Hic-5, and leupaxin, and all have been identified as key regulators of the cellular migration machinery in both 2- and 3-dimensional microenvironments. Herein, we provide insight into the roles of these proteins during tumorigenesis and metastasis, highlighting their functions in cancer initiation as well as tumor cell dissemination and survival. Furthermore, we speculate on the potential of paxillin family proteins as both future prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Nicholas O Deakin
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | | | | |
Collapse
|
20
|
Cbl-c ubiquitin ligase activity is increased via the interaction of its RING finger domain with a LIM domain of the paxillin homolog, Hic 5. PLoS One 2012; 7:e49428. [PMID: 23145173 PMCID: PMC3492284 DOI: 10.1371/journal.pone.0049428] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 10/08/2012] [Indexed: 12/31/2022] Open
Abstract
Cbl proteins (Cbl, Cbl-b and Cbl-c) are ubiquitin ligases that are critical regulators of tyrosine kinase signaling. In this study we identify a new Cbl-c interacting protein, Hydrogen peroxide Induced Construct 5 (Hic-5). The two proteins interact through a novel interaction mediated by the RING finger of Cbl-c and the LIM2 domain of Hic-5. Further, this interaction is mediated and dependent on specific zinc coordinating complexes within the RING finger and LIM domain. Binding of Hic-5 to Cbl-c leads to an increase in the ubiquitin ligase activity of Cbl-c once Cbl-c has been activated by Src phosphorylation or through an activating phosphomimetic mutation. In addition, co-transfection of Hic-5 with Cbl-c leads to an increase in Cbl-c mediated ubiquitination of the EGFR. These data suggest that Hic-5 enhances Cbl-c ubiquitin ligase activity once Cbl-c has been phosphorylated and activated. Interactions between heterologous RING fingers have been shown to activate E3s. This is the first demonstration of enhancement of ubiquitin ligase activity of a RING finger ubiquitin ligase by the direct interaction of a LIM zinc coordinating domain.
Collapse
|
21
|
Abstract
PURPOSE The 27-kDa heat shock protein (HSP27) has been implicated in wound healing in multiple tissues. We investigated the expression and localization of phosphorylated HSP27 during epithelial wound healing in the murine cornea. METHODS Corneas of 8- to 10-week-old C57BL6 mice were wounded by epithelial debridement (n = 40). Unwounded corneas served as controls (n = 3). After 3, 7, and 14 days, phosphorylated HSP27 localization in wounded corneas was observed by confocal immunohistochemistry and double immunogold labeling transmission immunoelectron microscopy. Western blot analysis was performed to determine expression levels of phosphorylated HSP27 in scraped epithelia. Phosphorylated HSP27 localization was also separately performed with confocal immunohistochemistry 8 hours after epithelial debridement to investigate the early epithelial wound-healing process. RESULTS In unwounded corneas, phosphorylated HSP27 was localized only to the superficial epithelium. In contrast, phosphorylated HSP27 was localized in the basal and superficial epithelia 3 days after corneal epithelial wounding. After 7 and 14 days, HSP27 localization was similar to that in unwounded controls. Expression levels of phosphorylated HSP27 were greater in wounded corneal epithelia on day 3 than in unwounded controls and on day 14. After 8 hours, phosphorylated HSP27 expression was prominent in the leading edge of migrating corneal epithelium. CONCLUSIONS Constitutive expression of phosphorylated HSP27 is limited to the superficial corneal epithelium in unwounded murine corneas. Changes in HSP27 epithelial distribution and expression levels after corneal epithelial wounding suggest that phosphorylated HSP27 plays a role in early phase of corneal epithelial wound healing.
Collapse
|
22
|
Kim-Kaneyama JR, Miyauchi A, Lei XF, Arita S, Mino T, Takeda N, Kou K, Eto K, Yoshida T, Miyazaki T, Shioda S, Miyazaki A. Identification of Hic-5 as a novel regulatory factor for integrin αIIbβ3 activation and platelet aggregation in mice. J Thromb Haemost 2012; 10:1867-74. [PMID: 22812543 DOI: 10.1111/j.1538-7836.2012.04856.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Integrin αIIbβ3 plays key roles in platelet aggregation and subsequent thrombus formation. Hydrogen peroxide-inducible clone-5 (Hic-5), a member of the paxillin family, serves as a focal adhesion adaptor protein associated with αIIbβ3 at its cytoplasmic strand. OBJECTIVES Hic-5 function in αIIbβ3 activation and subsequent platelet aggregation remains unknown. To address this question, platelets from Hic-5(-/-) mice were analyzed. METHODS AND RESULTS Hic-5(-/-) mice displayed a significant hemostatic defect and resistance to thromboembolism, which were explained in part by weaker thrombin-induced aggregation in Hic-5(-/-) platelets. Mechanistically, Hic-5(-/-) platelets showed limited activation of αIIbβ3 upon thrombin treatment. Morphological alteration in Hic-5(-/-) platelets after thrombin stimulation on fibrinogen plates was also limited. As a direct consequence, the quantity of actin co-immunoprecipitating with the activated αIIbβ3 was smaller in Hic-5(-/-) platelets than in wild-type platelets. CONCLUSION We identified Hic-5 as a novel and specific regulatory factor for thrombin-induced αIIbβ3 activation and subsequent platelet aggregation in mice.
Collapse
Affiliation(s)
- J R Kim-Kaneyama
- Department of Biochemistry, Showa University School of Medicine, Tokyo Department of Clinical Toxicology, Showa University School of Pharmacy, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kim-Kaneyama JR, Lei XF, Arita S, Miyauchi A, Miyazaki T, Miyazaki A. Hydrogen peroxide-inducible clone 5 (Hic-5) as a potential therapeutic target for vascular and other disorders. J Atheroscler Thromb 2012; 19:601-7. [PMID: 22472216 DOI: 10.5551/jat.10736] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Hydrogen peroxide-inducible clone-5 (Hic-5) is a focal adhesion scaffold protein primarily expressed in vascular and visceral smooth muscle cells. We recently generated mice lacking Hic-5, which grew with no apparent abnormality (Kim-Kaneyama J, et al. J Mol Cell Cardiol. 2011;50(1):77-86). However, we discovered that recovery of arterial media following vascular injury is delayed significantly in Hic-5 knockout mice consequent to enhanced apoptosis of cultured vascular smooth muscle cells after mechanical stress; thus, Hic-5 is regarded as a novel factor in vascular remodeling. The Hic-5 gene is also induced by transforming growth factor-β, a well-known accelerator in fibrosis. Hic-5 involvement in various fibrotic disorders, e.g., scar formation, keloid formation and glomerulosclerosis, has been proposed. siRNA silencing of Hic-5 in a breast cancer cell line reduces its invasiveness; moreover, Hic-5 serves as a steroid hormone co-activator and likely participates in endometriosis and prostate cancer. Thus, functional characterization of Hic-5 in various pathophysiological conditions may afford novel mechanistic insights into a wide variety of diseases.
Collapse
Affiliation(s)
- Joo-ri Kim-Kaneyama
- Department of Biochemistry, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
24
|
Pignatelli J, Tumbarello DA, Schmidt RP, Turner CE. Hic-5 promotes invadopodia formation and invasion during TGF-β-induced epithelial-mesenchymal transition. ACTA ACUST UNITED AC 2012; 197:421-37. [PMID: 22529104 PMCID: PMC3341156 DOI: 10.1083/jcb.201108143] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The focal adhesion protein Hic-5 acts through RhoC to promote TGF-β–stimulated invadopodia formation, cell migration, and invasion. Transforming growth factor β (TGF-β)–stimulated epithelial–mesenchymal transition (EMT) is an important developmental process that has also been implicated in increased cell invasion and metastatic potential of cancer cells. Expression of the focal adhesion protein Hic-5 has been shown to be up-regulated in epithelial cells in response to TGF-β. Herein, we demonstrate that TGF-β–induced Hic-5 up-regulation or ectopic expression of Hic-5 in normal MCF10A cells promoted increased extracellular matrix degradation and invasion through the formation of invadopodia. Hic-5 was tyrosine phosphorylated in an Src-dependent manner after TGF-β stimulation, and inhibition of Src activity or overexpression of a Y38/60F nonphosphorylatable mutant of Hic-5 inhibited matrix degradation and invasion. RhoC, but not RhoA, was also required for TGF-β– and Hic-5–induced matrix degradation. Hic-5 also induced matrix degradation, cell migration, and invasion in the absence of TGF-β via Rac1 regulation of p38 MAPK. These data identify Hic-5 as a critical mediator of TGF-β–stimulated invadopodia formation, cell migration, and invasion.
Collapse
Affiliation(s)
- Jeanine Pignatelli
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | | | | | | |
Collapse
|
25
|
Deakin NO, Turner CE. Distinct roles for paxillin and Hic-5 in regulating breast cancer cell morphology, invasion, and metastasis. Mol Biol Cell 2011; 22:327-41. [PMID: 21148292 PMCID: PMC3031464 DOI: 10.1091/mbc.e10-09-0790] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This study reveals novel roles for the focal adhesion proteins paxillin and Hic-5 in regulating breast cancer invasion strategies and metastasis. Depletion of paxillin promotes a hypermesenchymal phenotype while dysregulating 3D adhesion dynamics. In contrast, RNAi of Hic-5 induces a hyperamoeboid phenotype with dysregulated RhoA/pMLC signaling. Individual metastatic tumor cells exhibit two interconvertible modes of cell motility during tissue invasion that are classified as either mesenchymal or amoeboid. The molecular mechanisms by which invasive breast cancer cells regulate this migratory plasticity have yet to be fully elucidated. Herein we show that the focal adhesion adaptor protein, paxillin, and the closely related Hic-5 have distinct and unique roles in the regulation of breast cancer cell lung metastasis by modulating cell morphology and cell invasion through three-dimensional extracellular matrices (3D ECMs). Cells depleted of paxillin by RNA interference displayed a highly elongated mesenchymal morphology, whereas Hic-5 knockdown induced an amoeboid phenotype with both cell populations exhibiting reduced plasticity, migration persistence, and velocity through 3D ECM environments. In evaluating associated signaling pathways, we determined that Rac1 activity was increased in cells devoid of paxillin whereas Hic-5 silencing resulted in elevated RhoA activity and associated Rho kinase–induced nonmuscle myosin II activity. Hic-5 was essential for adhesion formation in 3D ECMs, and analysis of adhesion dynamics and lifetime identified paxillin as a key regulator of 3D adhesion assembly, stabilization, and disassembly.
Collapse
Affiliation(s)
- Nicholas O Deakin
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | | |
Collapse
|
26
|
Chen PW, Kroog GS. Leupaxin is similar to paxillin in focal adhesion targeting and tyrosine phosphorylation but has distinct roles in cell adhesion and spreading. Cell Adh Migr 2011; 4:527-40. [PMID: 20543562 DOI: 10.4161/cam.4.4.12399] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Focal adhesion (FA) formation is induced by extracellular matrix-stimulated integrin clustering and activation of receptors for diffusible factors. Leupaxin (LPXN) is a member of the paxillin family of FA proteins expressed in many cancer cell lines. We found activation of gastrin-releasing peptide receptor (GRPr) by bombesin (BN) stimulated LPXN translocation from cytoplasm to FAs. Using mutagenesis, we identified LIM3 as the primary FA targeting domain for LPXN and showed BN-induced LPXN tyrosine phosphorylation on residues 22, 62 and 72. A LIM3 point mutant of LPXN failed to target to FAs and had no BN-stimulated tyrosine phosphorylation. Conversely, a non-phosphorylatable mutant (Y22/62/72F) translocated to FAs after BN addition. Stimulation of FA formation using vinblastine also induced LPXN translocation and tyrosine phosphorylation. Therefore, dynamic LPXN tyrosine phosphorylation requires translocation to FAs. LPXN and paxillin had opposite roles in adhesion to collagen I (CNI) in MDA-MB-231 breast cancer cells. LPXN siRNA stimulated whereas paxillin siRNA inhibited cell adhesion. Knockdown of both LPXN and paxillin behaved similarly to paxillin knockdown alone, suggesting LPXN’s function in adhesion might depend on paxillin. Additionally, LPXN regulated cell spreading on CNI but not on fibronectin whereas paxillin knockdown suppressed spreading on both substrates. These results demonstrate that although LPXN and paxillin’s FA targeting and tyrosine phosphorylation are similar, each protein has distinct functions.
Collapse
Affiliation(s)
- Pei-Wen Chen
- Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | |
Collapse
|
27
|
Protein Quality Control, Retention, and Degradation at the Endoplasmic Reticulum. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 292:197-280. [DOI: 10.1016/b978-0-12-386033-0.00005-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Tanaka T, Moriwaki K, Murata S, Miyasaka M. LIM domain-containing adaptor, leupaxin, localizes in focal adhesion and suppresses the integrin-induced tyrosine phosphorylation of paxillin. Cancer Sci 2010; 101:363-8. [PMID: 19917054 PMCID: PMC11158308 DOI: 10.1111/j.1349-7006.2009.01398.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Focal adhesion (FA) consists of multiple cellular proteins including paxillin and serves as a center for adhesion-mediated signaling. The assembly and disassembly of FAs is regulated by locally produced intracellular signals, and tyrosine phosphorylation of paxillin has been implicated in this process. A Lin-11 Isl-1 Mec-3 (LIM) domain-containing adaptor protein, leupaxin, a member of the paxillin family, is expressed in leukocytes as well as in certain cancer cells, and shares overall structural characteristics with paxillin. However, it remains unknown whether leupaxin and paxillin cooperate with or antagonize each other in integrin signaling. Here we show that leupaxin potently represses the tyrosine phosphorylation of paxillin. When expressed in mouse thymoma BW5147 cells bound to ICAM-1, leupaxin accumulated in FA-like patches in the cell periphery. When expressed in NIH3T3 and HEK293T cells, leupaxin localized to FAs upon cell adhesion to fibronectin and strongly suppressed the integrin-induced tyrosine phosphorylation of paxillin. In integrin-stimulated HEK293T cells, leupaxin's LIM3 domain appeared essential for selective FA localization and the suppression of paxillin tyrosine phosphorylation. Leupaxin's LD3 motif, which is critical for stable association with FAK, was dispensable for leupaxin's suppressive ability. In addition, leupaxin reduced the spreading of NIH3T3 cells on fibronectin, which required both the LD3 motif and LIM3 domain. When expressed in human leukocytic K562 cells, leupaxin significantly suppressed integrin alpha5beta1-mediated cell adhesion to fibronectin and the tyrosine phosphorylation of paxillin. These findings indicate that leupaxin functions as a paxillin counterpart that potently suppresses the tyrosine phosphorylation of paxillin during integrin signaling.
Collapse
Affiliation(s)
- Toshiyuki Tanaka
- Laboratory of Immunodynamics, Department of Microbiology and Immunology, Osaka University, Graduate School of Medicine, Osaka, Japan.
| | | | | | | |
Collapse
|
29
|
Aghajanova L, Velarde MC, Giudice LC. The progesterone receptor coactivator Hic-5 is involved in the pathophysiology of endometriosis. Endocrinology 2009; 150:3863-70. [PMID: 19389829 PMCID: PMC2717860 DOI: 10.1210/en.2009-0008] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 04/15/2009] [Indexed: 11/19/2022]
Abstract
Endometriosis is an estrogen-dependent disorder primarily associated with pelvic pain and infertility in up to 10% of women of reproductive age. Recent studies suggest that resistance to progesterone action may contribute to the development and pathophysiology of this disorder. In this study we examined the in vivo and in vitro expression and function of one progesterone receptor (PR) coactivator, Hic-5, in human endometrium and endometrial stromal fibroblasts (hESFs) from 29 women with and 30 (control) women without endometriosis. Hic-5 was highly expressed in stromal, but not epithelial, cells in women without endometriosis, in a cycle-dependent manner. In contrast, Hic-5 expression was not regulated during the menstrual cycle in hESFs from women with endometriosis and was significantly reduced in hESFs from women with vs. without disease. Hic-5 mRNA expression throughout the cycle in endometrium from control women, but not those with endometriosis, correlated with expression of PR. Hic-5 mRNA in hESFs was significantly up-regulated in control but not endometriosis hESFs after treatment in vitro with 8-bromoadenosine-cAMP for 96 h but only modestly after 14 d of progesterone treatment. Hic-5 silencing did not influence cAMP-regulated gene expression but affected genes regulated solely by progesterone (e.g. DKK1 and calcitonin). Together the data suggest that the proposed progesterone resistance in endometrium from women with endometriosis derives, in part, from impaired expression of the PR coactivator, Hic-5, in endometrial tissue and cultured endometrial stromal fibroblasts.
Collapse
Affiliation(s)
- Lusine Aghajanova
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | |
Collapse
|
30
|
Hic-5, an adaptor protein expressed in vascular smooth muscle cells, modulates the arterial response to injury in vivo. Biochem Biophys Res Commun 2008; 376:682-7. [PMID: 18812162 DOI: 10.1016/j.bbrc.2008.09.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Accepted: 09/11/2008] [Indexed: 11/22/2022]
Abstract
Focal adhesion components are targets for biochemical and mechanical stimuli that evoke crucial injury. Hic-5 (hydrogen peroxide-inducible clone 5) is a multidomain adaptor protein which is implicated in the regulation of integrin signaling in focal adhesion. The aim of this research was to test the hypothesis that Hic-5, a focal adhesion LIM protein expressed in smooth muscle cells, is involved in dynamic processes by pathological stimuli in the vessel wall. Here, we describe the analysis of the function of Hic-5 using a mouse model of vascular injury that may mimic balloon angioplasty. At 4 days after vascular injury, marked down-regulation of the Hic-5 expression was observed in the smooth muscle layer, and local delivery of the Hic-5 using adenovirus vectors repressed injury-induced neointimal expansion. In addition, Hic-5 reduced cells migration into three-dimensional collagen gels, and the forced expression of Hic-5 in cells embedded in the collagen gel matrix repressed the expression of uPA that participates in smooth muscle cell migration. These results suggest that Hic-5 modulates cellular responses to pathological stimuli in the vessel wall.
Collapse
|
31
|
Smad7 is inactivated through a direct physical interaction with the LIM protein Hic-5/ARA55. Oncogene 2008; 27:6791-805. [PMID: 18762808 DOI: 10.1038/onc.2008.291] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We recently reported that hydrogen peroxide-inducible clone-5 (Hic-5, also named androgen receptor-associated protein 55) can bind to the transforming growth factor-beta (TGF-beta)-signaling regulator Smad3, thereby inhibiting certain Smad3-dependent TGF-beta responses. We now show that Hic-5 can also control TGF-beta responses through an alternative mechanism involving Smad7, a key negative regulator of TGF-beta signaling. Hic-5 binds directly to Smad7. This interaction requires the LIM3 domain of Hic-5, and enhances TGF-beta signaling through causing loss of Smad7 protein but not mRNA. Enforced expression of Hic-5 reverses the ability of Smad7 to suppress TGF-beta-induced phosphorylation of Smads 2 and 3 and activation of the plasminogen activator inhibitor-1 promoter (in NRP-154 and PC3 prostate carcinoma and WPMY-1 prostate myofibroblast cell lines). Lentiviral-mediated small-hairpin RNA silencing of endogenous Hic-5 reduced TGF-beta responses in PC3 and WPMY-1 cells. Further work suggests that the level of Smad7 is modulated by its physical interaction with Hic-5 and targeted to a degradation pathway not likely to be proteasomal. Our findings support that Hic-5 functions as a cell-type-specific activator of TGF-beta signaling through its ability to physically interact with and neutralize Smad7.
Collapse
|
32
|
Kaulfuss S, Grzmil M, Hemmerlein B, Thelen P, Schweyer S, Neesen J, Bubendorf L, Glass AG, Jarry H, Auber B, Burfeind P. Leupaxin, a novel coactivator of the androgen receptor, is expressed in prostate cancer and plays a role in adhesion and invasion of prostate carcinoma cells. Mol Endocrinol 2008; 22:1606-21. [PMID: 18451096 DOI: 10.1210/me.2006-0546] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
In the present study, we demonstrate that leupaxin mRNA is overexpressed in prostate cancer (PCa) as compared with normal prostate tissue by using cDNA arrays and quantitative RT-PCR analyses. Moderate to strong expression of leupaxin protein was detected in approximately 22% of the PCa tissue sections analyzed, and leupaxin expression intensities were found to be significantly correlated with Gleason patterns/scores. In addition, different leupaxin expression levels were observed in PCa cell lines, and at the subcellular level, leupaxin was usually localized in focal adhesion sites. Furthermore, mutational analysis and transfection experiments of LNCaP cells using different green fluorescent protein-leupaxin constructs demonstrated that leupaxin contains functional nuclear export signals in its LD3 and LD4 motifs, thus shuttling between the cytoplasm and the nucleus. We could also demonstrate for the first time that leupaxin interacts with the androgen receptor in a ligand-dependent manner and serves as a transcriptional activator of this hormone receptor in PCa cells. Down-regulation of leupaxin expression using RNA interference in LNCaP cells resulted in a high rate of morphological changes, detachment, spontaneous apoptosis, and a reduction of prostate-specific antigen secretion. In contrast, knockdown of leupaxin expression in androgen-independent PC-3 and DU 145 cells induced a significant decrease of both the invasive capacity and motility. Our results therefore indicate that leupaxin could serve as a potential progression marker for a subset of PCa and may represent a novel coactivator of the androgen receptor. Leupaxin could function as a putative target for therapeutic interventions of a subset of advanced PCa.
Collapse
Affiliation(s)
- Silke Kaulfuss
- Institute of Human Genetics, University of Göttingen, Heinrich-Düker Weg 12, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Srinivasan R, Forman S, Quinlan RA, Ohanian J, Ohanian V. Regulation of contractility by Hsp27 and Hic-5 in rat mesenteric small arteries. Am J Physiol Heart Circ Physiol 2007; 294:H961-9. [PMID: 18083901 DOI: 10.1152/ajpheart.00939.2007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The regulation of small artery contractility by vasoconstrictors is important for vascular function, and actin cytoskeleton remodeling is required for contraction. p38 MAPK and tyrosine kinases are implicated in actin polymerization and contraction through heat shock protein 27 (Hsp27) and the cytoskeletal protein paxillin, respectively. We evaluated the roles of downstream targets of p38 MAPK and tyrosine kinases in cytoskeletal reorganization and contraction and whether the two signaling pathways regulate contraction independent of each other. We identified the expression of the paxillin homologue hydrogen peroxide-inducible clone-5 (Hic-5) and showed its activation by norepinephrine (NE) in a Src-dependent manner. Furthermore, we demonstrated a NE-induced interaction of proline-rich tyrosine kinase-2 (PYK2) but not Src or p125 focal adhesion kinase with Hic-5. This interaction was Src dependent, suggesting that Hic-5 was a substrate for PYK2 downstream from Src. The activation of Hic-5 induced its relocalization to the cytosol. The parallel activation of Hsp27 by NE was p38 MAPK dependent and led to its dissociation from actin filaments and translocation from membrane to cytosol and increased actin polymerization. Both Hsp27 and Hic-5 activation resulted in their association within the same time frame as NE-induced contraction, and the inhibition of either p38 MAPK or Src inhibited the interaction between Hsp27 and Hic-5 and the contractile response. Furthermore, combined p38 MAPK and Src inhibition had no greater effect on contraction than individual inhibition, suggesting that the two pathways act through a common mechanism. These data show that NE-induced activation and the association of Hsp27 and Hic-5 are required for the reorganization of the actin cytoskeleton and force development in small arteries.
Collapse
Affiliation(s)
- R Srinivasan
- Cardiovascular Research Group, University of Manchester, UK
| | | | | | | | | |
Collapse
|
34
|
Avraamides C, Bromberg ME, Gaughan JP, Thomas SM, Tsygankov AY, Panetti TS. Hic-5 promotes endothelial cell migration to lysophosphatidic acid. Am J Physiol Heart Circ Physiol 2007; 293:H193-203. [PMID: 17337598 DOI: 10.1152/ajpheart.00728.2006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cell migration is critical for proper blood vessel development. Signals from growth factors and matrix proteins are integrated through focal adhesion proteins to alter cell migration. Hydrogen peroxide-inducible clone 5 (Hic-5), a paxillin family member, is enriched in the focal adhesions in bovine pulmonary artery endothelial (BPAE) cells, which migrate to lysophosphatidic acid (LPA) on denatured collagen. In this study, we investigate the role of Hic-5 in LPA-stimulated endothelial cell migration. LPA recruits Hic-5 to the focal adhesions and to the pseudopodia in BPAE cells plated on collagen, suggesting that recruitment of Hic-5 to focal adhesions is associated with endothelial cell migration. Knockdown of endogenous Hic-5 significantly decreases migration toward LPA, confirming involvement of Hic-5 in migration. To address the role of Hic-5 in endothelial cell migration, we exogenously expressed wild-type (WT) Hic-5 and green fluorescent protein Hic-5 C369A/C372A (LIM3 mutant) constructs in BPAE cells. WT Hic-5 expression increases chemotaxis of BPAE cells to LPA, whereas migration toward LPA of the green fluorescent protein Hic-5 C369A/C372A-expressing cells is similar to that shown in vector control cells. Additionally, ERK phosphorylation is enhanced in the presence of LPA in WT Hic-5 cells. A pharmacological inhibitor of MEK activity inhibits LPA-stimulated WT Hic-5 cell migration and ERK phosphorylation, suggesting Hic-5 enhances migration via MEK activation of ERK. Together, these studies indicate that Hic-5, a focal adhesion protein in endothelial cells, is recruited to the pseudopodia in the presence of LPA and enhances migration.
Collapse
Affiliation(s)
- C Avraamides
- Department of Microbiology and Immunology, and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 N. Broad Street, Philadelphia, PA 19140, USA
| | | | | | | | | | | |
Collapse
|
35
|
Croke JM, Pike LRG, MacPhee DJ. The focal adhesion protein Hic-5 is highly expressed in the rat myometrium during late pregnancy and labour and co-localizes with FAK. Reprod Biol Endocrinol 2007; 5:22. [PMID: 17550607 PMCID: PMC1892559 DOI: 10.1186/1477-7827-5-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Accepted: 06/05/2007] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Myometrial growth and remodeling of the cytoskeleton and focal adhesions during late pregnancy may be critical aspects of myometrial activation and thus labour. Yet our understanding of these aspects is inhibited by the paucity of information concerning the components of focal adhesions in the myometrium. The focal adhesion protein hydrogen peroxide-inducible clone-5 (Hic-5) has recently been found in mononuclear smooth muscle but was not examined in the myometrium during pregnancy. Thus, the goal of this study was to characterize Hic-5 mRNA and protein expression in the rat myometrium during pregnancy and labour. METHODS Rat myometrium samples were obtained from non-pregnant animals, pregnant animals on days (d) 6, 12, 15, 17, 19, 21, 22, 23 (active labour) and 1 day postpartum (PP). In addition, myometrium samples were collected from rats within a progesterone-delayed labour paradigm. Hic-5 mRNA expression was analyzed by Northern blot analysis while Hic-5 protein expression was examined by immunoblot and immunofluorescence analysis. RESULTS Hic-5 mRNA expression on d15, d19 and d21 was found to be significantly elevated compared to d6 and d12 of pregnancy and expression on d23 was significantly elevated over d6 (p < 0.05). Immunofluorescence analysis demonstrated that detection of Hic-5 protein in the circular muscle layer appeared to increase from d17 onwards, except PP, and Hic-5 was detectable in the cell cytoplasm and more continuously associated with myometrial cell membranes. In the longitudinal muscle layer Hic-5 was readily detectable by d15 and thereafter and primarily associated at myometrial cell membranes. Co-immunofluorescence analysis of potential Hic-5 and focal adhesion kinase (FAK) association in situ demonstrated a limited level of co-localization on d19, d23 and PP in the circular muscle layer while in the longitudinal muscle layer Hic-5 and FAK were readily co-localized at myometrial cell membranes. CONCLUSION Hic-5 is highly expressed in the rat myometrium during late pregnancy and labour and co-localizes with FAK in situ. Our results are consistent with a potential role for Hic-5 in focal adhesion remodeling in the rat myometrium during late pregnancy.
Collapse
Affiliation(s)
- Jenn M Croke
- Division of Basic Medical Sciences, Health Sciences Centre, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Luke RG Pike
- Division of Basic Medical Sciences, Health Sciences Centre, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Daniel J MacPhee
- Division of Basic Medical Sciences, Health Sciences Centre, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| |
Collapse
|
36
|
Differential roles of HIC-5 isoforms in the regulation of cell death and myotube formation during myogenesis. Exp Cell Res 2007; 313:4000-14. [PMID: 17935713 DOI: 10.1016/j.yexcr.2007.05.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 05/16/2007] [Accepted: 05/16/2007] [Indexed: 11/21/2022]
Abstract
Hic-5 is a LIM-Only member of the paxillin superfamily of focal adhesion proteins. It has been shown to regulate a range of biological processes including: senescence, tumorigenesis, steroid hormone action, integrin signaling, differentiation, and apoptosis. To better understand the roles of Hic-5 during development, we initiated a detailed analysis of Hic-5 expression and function in C(2)C(12) myoblasts, a well-established model for myogenesis. We have found that: (1) myoblasts express at least 6 distinct Hic-5 isoforms; (2) the two predominant isoforms, Hic-5alpha and Hic-5beta, are differentially expressed during myogenesis; (3) any experimentally induced change in Hic-5 expression results in a substantial increase in apoptosis during differentiation; (4) ectopic expression of Hic-5alpha is permissive to differentiation while expression of either Hic-5beta or antisense Hic-5 blocks myoblast fusion but not chemodifferentiation; (5) Hic-5 localizes to focal adhesions in C(2)C(12) myoblasts and perturbation of Hic-5 leads to defects in cell spreading; (6) alterations in Hic-5 expression interfere with the normal dynamics of laminin expression; and (7) ectopic laminin, but not fibronectin, can rescue the Hic-5-induced blockade of myoblast survival and differentiation. Our data demonstrate differential roles for individual Hic-5 isoforms during myogenesis and support the hypothesis that Hic-5 mediates these effects via integrin signaling.
Collapse
|
37
|
Golubovskaya VM, Cance WG. Focal adhesion kinase and p53 signaling in cancer cells. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 263:103-53. [PMID: 17725966 DOI: 10.1016/s0074-7696(07)63003-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The progression of human cancer is characterized by a process of tumor cell motility, invasion, and metastasis to distant sites, requiring the cancer cells to be able to survive the apoptotic pressures of anchorage-independent conditions. One of the critical tyrosine kinases linked to these processes of tumor invasion and survival is the focal adhesion kinase (FAK). FAK was first isolated from human tumors, and FAK mRNA was found to be upregulated in invasive and metastatic human breast and colon cancer samples. Recently, the FAK promoter was cloned, and it has been found to contain p53-binding sites. p53 inhibits FAK transcription, and recent data show direct binding of FAK and p53 proteins in vitro and in vivo. The structure of FAK and p53, proteins interacting with FAK, and the role of FAK in tumorigenesis and FAK-p53-related therapy are reviewed. This review focuses on FAK signal transduction pathways, particularly on FAK and p53 signaling, revealing a new paradigm in cell biology, linking signaling from the extracellular matrix to the nucleus.
Collapse
Affiliation(s)
- Vita M Golubovskaya
- Department of Surgery, University of Florida School of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | | |
Collapse
|
38
|
Saelzler MP, Spackman CC, Liu Y, Martinez LC, Harris JP, Abe MK. ERK8 down-regulates transactivation of the glucocorticoid receptor through Hic-5. J Biol Chem 2006; 281:16821-32. [PMID: 16624805 DOI: 10.1074/jbc.m512418200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Extracellular signal-regulated kinase 8 (ERK8) is the most recently identified member of the ERK subfamily of MAPKs. Although other members of the ERK subfamily are established regulators of signaling pathways involved in cell growth and/or differentiation, less is known about ERK8. To understand the cellular function of ERK8, a yeast two-hybrid screen of a human lung library was performed to identify binding partners. One binding partner identified was Hic-5 (also known as ARA55), a multiple LIM domain containing protein implicated in focal adhesion signaling and the regulation of specific nuclear receptors, including the androgen receptor and the glucocorticoid receptor (GR). Co-immunoprecipitation experiments in mammalian cells confirmed the interaction between Hic-5 and both ERK8 and its rodent ortholog ERK7. The C-terminal region of ERK8 was not required for the interaction. Although the LIM3 and LIM4 domains of Hic-5 were sufficient and required for this interaction, the specific zinc finger motifs in these domains were not. Transcriptional activation reporter assays revealed that ERK8 can negatively regulate transcriptional co-activation of androgen receptor and GRalpha by Hic-5 in a kinase-independent manner. Knockdown of endogenous ERK8 in human airway epithelial cells enhanced dexamethasone-stimulated transcriptional activity of endogenous GR. Transcriptional regulation of GRalpha and interaction with its ligand binding domain by ERK8 were dependent on the presence of Hic-5. These results provide the first physiological function for human ERK8 as a negative regulator of human GRalpha, acting through Hic-5, and suggest a broader role for ERK8 in the regulation of nuclear receptors beyond estrogen receptor alpha.
Collapse
Affiliation(s)
- Matthew P Saelzler
- Department of Pediatrics, University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637-1470, USA
| | | | | | | | | | | |
Collapse
|
39
|
Ghogomu SM, van Venrooy S, Ritthaler M, Wedlich D, Gradl D. HIC-5 is a novel repressor of lymphoid enhancer factor/T-cell factor-driven transcription. J Biol Chem 2005; 281:1755-64. [PMID: 16291758 DOI: 10.1074/jbc.m505869200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of Wnt/beta-catenin target genes is regulated by a heterodimer of beta-catenin and the high mobility group box transcription factors of the lymphoid enhancer factor (LEF)/T-cell factor (TCF) family. In vertebrates, four LEF/TCF family members have been identified. They all contain a conserved beta-catenin-binding motif at the N terminus and a highly conserved high mobility group box for DNA binding. The core sequence between these motifs is less conserved and contributes to the specific properties of the individual family members. To identify interacting proteins that allocate specific functions to the individual LEF/TCF transcription factors, we performed a yeast two-hybrid screen using the less conserved core sequence as bait. We isolated the murine LIM protein HIC-5 (hydrogen peroxide-induced clone 5; also termed ARA-55 (androgen receptor activator of 55 kDa)) and cloned the highly conserved Xenopus homolog. In addition, we report that the LIM domain-containing C-terminal half of HIC-5 binds to a conserved alternatively spliced exon in LEF/TCF transcription factors. Our functional analyses revealed that HIC-5 acts as negative regulator of a subset of LEF/TCF family members, which have been characterized as activators in reporter gene analyses and in the Xenopus axis induction assay. In addition, we observed a repressive interference of LEF/TCF family members with HIC-5-mediated activation of glucocorticoid-driven transcription, which again could be allocated to specific LEF/TCF subtypes. With the characterization of HIC-5 as a binding partner of the alternatively spliced exon in LEF/TCF transcription factors, we identified a novel molecular mechanism in the dialog of steroid and canonical Wnt signaling that is LEF/TCF subtype-dependent.
Collapse
Affiliation(s)
- Stephen Mbigha Ghogomu
- Zoologisches Institut II, Universität Karlsruhe (Technische Hochschule), Kaiserstrasse 12, 76131 Karlsruhe, Germany
| | | | | | | | | |
Collapse
|
40
|
Gao Z, Schwartz LM. Identification and analysis of Hic-5/ARA55 isoforms: Implications for integrin signaling and steroid hormone action. FEBS Lett 2005; 579:5651-7. [PMID: 16219310 DOI: 10.1016/j.febslet.2005.08.086] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 08/30/2005] [Accepted: 08/30/2005] [Indexed: 11/27/2022]
Abstract
Hic-5/ARA55 is a LIM-only member of the paxillin superfamily. Conflicting reports have suggested that Hic-5/ARA55 can both repress and enhance a number of biological processes, including myogenesis and tumorigenesis. With two Hic-5 isoforms documented, we hypothesized that multiple Hic-5 isoforms may exist that have both overlapping and isoform-specific functions. To test this hypothesis, we performed an extensive analysis of Hic-5 transcripts in both cell lines and mouse tissues and found 12 distinct isoforms that fall into two sub-families. These isoforms are derived from both alternative splicing and alternative transcriptional start sites (TSS). Hic-5 expression is regulated in a temporally and spatially controlled manner in vivo. The identification of numerous Hic-5 isoforms suggests that Hic-5 subsumes a number of distinct roles in cells and may explain the range of biological responses attributed to Hic-5.
Collapse
Affiliation(s)
- Zhengliang Gao
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, 01003, USA
| | | |
Collapse
|
41
|
Hetey SE, Lalonde DP, Turner CE. Tyrosine-phosphorylated Hic-5 inhibits epidermal growth factor-induced lamellipodia formation. Exp Cell Res 2005; 311:147-56. [PMID: 16183059 DOI: 10.1016/j.yexcr.2005.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 08/24/2005] [Accepted: 08/25/2005] [Indexed: 11/29/2022]
Abstract
The focal adhesion protein Hic-5, a homologue to paxillin, has been shown to be tyrosine-phosphorylated in fibroblasts in response to stimuli such as osmotic stress, serum, LPA and endothelin. However, the function of this modification to Hic-5 is unclear. Herein, we show that Hic-5 is tyrosine-phosphorylated on residues 38 and 60 following epidermal growth factor (EGF) treatment of COS-7 cells, coincident with an increase in peripheral actin reorganization. To explore the role of Hic-5 phosphorylation in this process, we introduced wild-type (WT) and mutant Hic-5 constructs into COS-7 cells and determined that EGF-induced lamellipodia formation was suppressed by WT Hic-5. This effect required localization to focal adhesions as well as phosphorylation of Hic-5 as overexpression of both a non-targeting and a non-phosphorylatable Hic-5 failed to inhibit peripheral actin reorganization. Interestingly, overexpression of non-phosphorylatable Y31/118F or WT paxillin did not affect lamellipodia formation, indicating that this effect is specific to Hic-5. The EGF-induced lamellipodia were Rac-dependent and overexpressed WT Hic-5, but not non-phosphorylatable Hic-5 inhibited Rac activation. Our data suggest that Hic-5 tyrosine phosphorylation functions to regulate signaling associated with lamellipodia formation, a process fundamental to cell motility.
Collapse
Affiliation(s)
- Sara E Hetey
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
42
|
Drori S, Girnun GD, Tou L, Szwaya JD, Mueller E, Xia K, Kia X, Shivdasani RA, Spiegelman BM. Hic-5 regulates an epithelial program mediated by PPARgamma. Genes Dev 2005; 19:362-75. [PMID: 15687259 PMCID: PMC546514 DOI: 10.1101/gad.1240705] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PPARgamma is a dominant regulator of fat cell differentiation. However, this nuclear receptor also plays an important role in the differentiation of intestinal and other epithelial cell types. The mechanism by which PPARgamma can influence the differentiation of such diverse cell lineages is unknown. We show here that PPARgamma interacts with Hic-5, a coactivator protein expressed in gut epithelial cells. Hic-5 and PPARgamma colocalize to the villus epithelium of the small intestine, and their expression during embryonic gut development correlates with the transition from endoderm to a specialized epithelium; expression of both these factors is reduced in tumors. Forced expression of Hic-5 in colon cancer cells enhances the PPARgamma-mediated induction of several gut epithelial differentiation/maturation markers such as L-FABP, kruppel-like factor 4 (KLF4), and keratin 20. siRNA directed against Hic-5 specifically reduces PPARgamma-mediated induction of gut epithelial genes in colon cells and in an ex vivo model of embryonic gut differentiation. Finally, forced expression of Hic-5 during 3T3-L1 preadipocyte differentiation inhibits adipogenesis while inducing inappropriate expression of several mRNAs characteristic of gut epithelium in these mesenchymal cells. These results indicate that Hic5 is an important component in determining an epithelial differentiation program induced by PPARgamma.
Collapse
Affiliation(s)
- Stavit Drori
- Dana-Farber Cancer Institute and the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Shibanuma M, Mori K, Kim-Kaneyama JR, Nose K. Involvement of FAK and PTP-PEST in the regulation of redox-sensitive nuclear-cytoplasmic shuttling of a LIM protein, Hic-5. Antioxid Redox Signal 2005; 7:335-47. [PMID: 15706082 DOI: 10.1089/ars.2005.7.335] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The LIM protein Hic-5 is a focal adhesion protein shuttling in and out of the nucleus through the redox-sensitive nuclear export signal, and unlike other focal adhesion proteins including paxillin, the protein most homologous to Hic-5, it accumulates in the nucleus under oxidative conditions and participates in the transcription of c-fos and p21(Cip1) genes. Here, we examined the roles of the interacting partners of Hic-5, focal adhesion kinase (FAK) and protein tyrosine phosphatase PEST (PTP-PEST), in the nuclear translocation of Hic-5 and found that they were inhibitory. Interestingly, the interaction of Hic-5 with FAK was regulated by specific cysteines near the binding site and decreased in cells under oxidative conditions. Its interaction with PTP-PEST was also sensitive to the oxidant. These results suggest that the nuclear-cytoplasmic shuttling of Hic-5 is regulated by its interacting partners at focal adhesions or in the cytoplasm in a redox-sensitive manner, coordinating its role at focal adhesions with that in the nucleus, depending on the redox state of cells. Cytochalasin D or a phorbol ester also induced nuclear accumulation of Hic-5, which was inhibited by scavengers of reactive oxygen species (ROS), suggesting that besides oxidants, endogenously produced ROS induced the nuclear accumulation of Hic-5.
Collapse
Affiliation(s)
- Motoko Shibanuma
- Department of Microbiology, Showa University School of Pharmaceutical Sciences, Hatanodai 1-5-8, Shinagawa-ku, Tokyo 142-8555, Japan.
| | | | | | | |
Collapse
|
44
|
Kim-Kaneyama JR, Suzuki W, Ichikawa K, Ohki T, Kohno Y, Sata M, Nose K, Shibanuma M. Uni-axial stretching regulates intracellular localization of Hic-5 expressed in smooth-muscle cells in vivo. J Cell Sci 2005; 118:937-49. [PMID: 15713747 DOI: 10.1242/jcs.01683] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hic-5 is a focal adhesion protein belonging to the paxillin LIM family that shuttles in and out of the nucleus. In the present study, we examined the expression of Hic-5 among mouse tissues by immunohistochemistry and found its expression only in smooth-muscle cells in several tissues. This result is consistent with a previous report on adult human tissues and contradicts the relatively ubiquitous expression of paxillin, the protein most homologous to Hic-5. One factor characterizing smooth-muscle cells in vivo is a continuous exposure to mechanical stretching in the organs. To study the involvement of Hic-5 in cellular responses to mechanical stress, we exposed mouse embryo fibroblasts to a uni-axial cyclic stretching and found that Hic-5 was relocalized from focal adhesions to stress fibers through its C-terminal LIM domains during the stress. In sharp contrast to this, paxillin did not change its focal-adhesion-based localization. Of the factors tested, which included interacting partners of Hic-5, only CRP2 (an only-LIM protein expressed in vascular smooth-muscle cells) and GIT1 were, like Hic-5, localized to stress fibers during the cyclic stretching. Interestingly, Hic-5 showed a suppressive effect on the contractile capability of cells embedded in three-dimensional collagen gels, and the effect was further augmented when CRP2 co-localized with Hic-5 to fiber structures of those cells. These results suggested that Hic-5 was a mediator of tensional force, translocating directly from focal adhesions to actin stress fibers upon mechanical stress and regulating the contractile capability of cells in the stress fibers.
Collapse
Affiliation(s)
- Joo-ri Kim-Kaneyama
- Department of Microbiology, Showa University School of Pharmaceutical Sciences, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang H, Song K, Sponseller TL, Danielpour D. Novel Function of Androgen Receptor-associated Protein 55/Hic-5 as a Negative Regulator of Smad3 Signaling. J Biol Chem 2005; 280:5154-62. [PMID: 15561701 DOI: 10.1074/jbc.m411575200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Androgen receptor-associated protein 55 (ARA55/Hic-5) belongs to the LIM protein superfamily and is featured by three or four N-terminal LD motifs and four C-terminal zinc finger-like LIM domains. Both LD motifs and LIM domains can serve as protein-protein interaction interfaces. Recently, we found that enforced expression of ARA55 inhibits transforming growth factor-beta-mediated up-regulation of Smad binding element-luciferase reporter activity in NRP-154 and NRP-152 rat prostate and LNCaP human prostate cell lines. Moreover, ARA55 also inhibits the induction of Smad-binding element 4-luciferase and 3TP-luciferase (a plasminogen activator inhibitor-1 (PAI-1) promoter construct) reporters by constitutively active (CA)-Smad3 in these cell lines. Co-immunoprecipitation studies suggest an interaction between ARA55 and either CA-Smad3 or wild-type Smad3 in HEK293 cells that occurs through the MH2 domain of Smad3 and the C terminus of ARA55 with wild-type Smad3 having stronger affinity than CA-Smad3 to ARA55. Glutathione S-transferase pull-down assays demonstrate that this interaction can occur in a cell-free system. These results are consistent with the luciferase data showing that the C terminus of ARA55 is critical for suppression of Smad3 activity. Furthermore, using a mammalian two-hybrid system, we confirmed that ARA55 interacts with the MH2 domain of Smad3 and suppresses CA-Smad3-induced transcriptional responses. In conclusion, these results support that ARA55 selectively intercepts transforming growth factor-beta signaling through an interaction of the LIM domain of ARA55 with the MH2 domain of Smad3.
Collapse
Affiliation(s)
- Hui Wang
- Ireland Cancer Center Research Laboratories and Department of Pharmacology, Case Western Reserve University/University Hospitals, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
46
|
Hirano S, Shelden EA, Gilmont RR. HSP27 regulates fibroblast adhesion, motility, and matrix contraction. Cell Stress Chaperones 2005; 9:29-37. [PMID: 15270075 PMCID: PMC1065303 DOI: 10.1379/471.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Heat shock protein 27 (HSP27) modulates actin-dependent cell functions in several systems. We hypothesized that HSP27 modulates wound contraction. Stably transfected fibroblast cell lines that overexpress HSP27 (SS12) or underexpress HSP27 (AS10) were established, and cell behaviors related to wound contraction were examined. First, fibroblast-populated collagen lattice (FPCL) contraction was examined because it has been studied as a wound-healing model. In floating FPCL contraction assays, SS12 cells caused increased contraction, whereas AS10 cells caused reduced contraction. Because floating matrix contraction is thought to be mediated by the tractional force of the cells, cell behaviors related to tractional force were examined. In collagen matrix, SS12 cells elongated faster and to a greater extent and contained longer stress fibers than control cells, whereas AS10 cells were slower to elongate than control cells. SS12 cells attached to the dishes more efficiently than the control, whereas AS10 cells attached less efficiently. Migration of SS12 cells on collagen-coated dishes was also enhanced, although AS10 cells did not differ from the control cells. In summary, HSP27 regulates fibroblast adhesion, elongation, and migration and the contraction of the floating matrix in a manner dependent on the level of its expression.
Collapse
Affiliation(s)
- Sahoko Hirano
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0592, USA
| | | | | |
Collapse
|
47
|
Sharp TV, Munoz F, Bourboulia D, Presneau N, Darai E, Wang HW, Cannon M, Butcher DN, Nicholson AG, Klein G, Imreh S, Boshoff C. LIM domains-containing protein 1 (LIMD1), a tumor suppressor encoded at chromosome 3p21.3, binds pRB and represses E2F-driven transcription. Proc Natl Acad Sci U S A 2004; 101:16531-6. [PMID: 15542589 PMCID: PMC534532 DOI: 10.1073/pnas.0407123101] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
LIM domains-containing protein 1 (LIMD1) is encoded at chromosome 3p21.3, a region commonly deleted in many solid malignancies. However, the function of LIMD1 is unknown. Here we show that LIMD1 specifically interacts with retinoblastoma protein (pRB), inhibits E2F-mediated transcription, and suppresses the expression of the majority of genes with E2F1-responsive elements. LIMD1 blocks tumor growth in vitro and in vivo and is down-regulated in the majority of human lung cancer samples tested. Our data indicate that LIMD1 is a tumor-suppressor gene, the protein product of which functionally interacts with pRB and the loss of which promotes lung carcinogenesis.
Collapse
MESH Headings
- Animals
- Base Sequence
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/secondary
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- Chromosomes, Human, Pair 3/genetics
- DNA/genetics
- DNA-Binding Proteins/metabolism
- E2F Transcription Factors
- E2F1 Transcription Factor
- Fibrosarcoma/genetics
- Fibrosarcoma/metabolism
- Fibrosarcoma/pathology
- Genes, Tumor Suppressor
- Humans
- Intracellular Signaling Peptides and Proteins
- LIM Domain Proteins
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Mice
- Mice, Nude
- Neoplasm Transplantation
- Protein Binding
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Retinoblastoma Protein/metabolism
- Subcellular Fractions/metabolism
- Transcription Factors/metabolism
- Transcription, Genetic
- Transplantation, Heterologous
- Tumor Stem Cell Assay
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Tyson V Sharp
- Cancer Research UK, Viral Oncology Group, Wolfson Institute for Biomedical Research, Cruciform Building, University College London, London WC1E 6BT, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Molecular scaffold or adaptor proteins facilitate precise spatiotemporal regulation and integration of multiple signaling pathways to effect the optimal cellular response to changes in the immediate environment. Paxillin is a multidomain adaptor that recruits both structural and signaling molecules to focal adhesions, sites of integrin engagement with the extracellular matrix, where it performs a critical role in transducing adhesion and growth factor signals to elicit changes in cell migration and gene expression.
Collapse
Affiliation(s)
- Michael C Brown
- Dept. of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | | |
Collapse
|
49
|
Guerrero-Santoro J, Yang L, Stallcup MR, DeFranco DB. Distinct LIM domains of Hic-5/ARA55 are required for nuclear matrix targeting and glucocorticoid receptor binding and coactivation. J Cell Biochem 2004; 92:810-9. [PMID: 15211577 DOI: 10.1002/jcb.20109] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Hydrogen peroxide-inducible clone-5 (Hic-5), belongs to the group III LIM domain protein family and contains four carboxyl-terminal LIM domains (LIM1-LIM4). In addition to its role in focal adhesion signaling, Hic-5 acts in the nucleus as a coactivator for some steroid hormone receptors such as the glucocorticoid receptor (GR) and androgen receptor (AR). Based upon its effect on AR transactivation, Hic-5 has also been designated as ARA55. Here, we report mapping studies of Hic-5/ARA55 functional domains and establish that LIM3 and LIM4 are necessary for maximal effects on GR transactivation. However, results from yeast two-hybrid assays demonstrated that these two LIM domains together, while necessary, are not sufficient to interact with the tau2 transactivation domain of GR. LIM4 also functions as a nuclear matrix targeting sequence (NMTS) for Hic-5/ARA55, as it is both necessary and sufficient to target a heterologous protein to the nuclear matrix. Thus, as suggested from previous analysis of LIM domain-containing proteins, separate but highly related LIM domains serve distinct functions.
Collapse
|
50
|
Hirano S, Shelden EA, Gilmont RR. HSP27 regulates fibroblast adhesion, motility, and matrix contraction. Cell Stress Chaperones 2004. [DOI: 10.1379/1466-1268(2004)009<0029:hrfama>2.0.co;2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|