1
|
Abé T, Yamazaki M, Nozumi M, Maruyama S, Takamura K, Ohashi R, Ajioka Y, Tanuma JI. Ladinin-1 in actin arcs of oral squamous cell carcinoma is involved in cell migration and epithelial phenotype. Sci Rep 2024; 14:22778. [PMID: 39354061 PMCID: PMC11445451 DOI: 10.1038/s41598-024-74041-z] [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: 10/22/2023] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
Histopathologically, oral squamous cell carcinoma (OSCC) consists of well-defined interfaces with adjacent non-cancerous epithelium. Previously, we found that SCC tissues expressed higher levels of specific proteins at this interface. Ladinin-1 (LAD1) is one of the specific molecules that has increased expressions in cancer fronts; however, its function in OSCC is unknown. Therefore, this study aimed to elucidate the function of LAD1 in human OSCC cells. LAD1 was localized on the actin arc at the distal periphery of cell clusters in the OSCC cell lines HSC-2, HSC-3, and HSC-4. When LAD1 was knocked down, cellular migration was repressed in wound scratch assays but was reversed in three-dimensional collagen gel invasion assays. Characteristic LAD1 localization along actin arcs forming the leading edge of migrating cells was diminished with loss of filopodia formation and ruffling in knockdown cells, in which the expression levels of cell motility-related genes-p21-activated kinase 1 (PAK1) and caveolin-1 (CAV1)-were upregulated and downregulated, respectively. LAD1 expression was also associated with the downregulation of vimentin and increased histological differentiation of OSCC. These results suggest that LAD1 is involved in actin dynamics during filopodia and lamellipodia formation, and in maintaining the epithelial phenotype of OSCC cells.
Collapse
Affiliation(s)
- Tatsuya Abé
- Division of Oral Pathology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan.
| | - Manabu Yamazaki
- Division of Oral Pathology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medicine, Niigata University, Niigata, Japan
| | - Satoshi Maruyama
- Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan
| | - Kaori Takamura
- Division of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Riuko Ohashi
- Division of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yoichi Ajioka
- Division of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Jun-Ichi Tanuma
- Division of Oral Pathology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| |
Collapse
|
2
|
Peng L, He Y, Wang W, Dai J, Li Q, Ju S. PAK1-Dependent Regulation of Microtubule Organization and Spindle Migration Is Essential for the Metaphase I-Metaphase II Transition in Porcine Oocytes. Biomolecules 2024; 14:237. [PMID: 38397472 PMCID: PMC10886677 DOI: 10.3390/biom14020237] [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: 12/26/2023] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
P21-activated kinase 1 (PAK1) is a critical downstream target that mediates the effect of small Rho GTPase on the regulation of cytoskeletal kinetics, cell proliferation, and cell migration. PAK1 has been identified as a crucial regulator of spindle assembly during the first meiotic division; however, its roles during the metaphase I (MI) to metaphase II (MII) transition in oocytes remain unclear. In the present study, the potential function of PAK1 in regulating microtubule organization and spindle positioning during the MI-MII transition was addressed in porcine oocytes. The results showed that activated PAK1 was co-localized with α-tubulin, and its expression was increased from the MI to MII stage (p < 0.001). However, inhibiting PAK1 activity with an inhibitor targeting PAK1 activation-3 (IPA-3) at the MI stage decreased the first polar body (PB1) extrusion rate (p < 0.05), with most oocytes arrested at the anaphase-telophase (ATI) stage. IPA-3-treated oocytes displayed a decrease in actin distribution in the plasma membrane (p < 0.001) and an increase in the rate of defects in MII spindle reassembly with abnormal spindle positioning (p < 0.001). Nevertheless, these adverse effects of IPA-3 on oocytes were reversed when the disulfide bond between PAK1 and IPA-3 was reduced by dithiothreitol (DTT). Co-immunoprecipitation revealed that PAK1 could recruit activated Aurora A and transform acidic coiled-coil 3 (TACC3) to regulate spindle assembly and interact with LIM kinase 1 (LIMK1) to facilitate actin filament-mediated spindle migration. Together, PAK1 is essential for microtubule organization and spindle migration during the MI-MII transition in porcine oocytes, which is associated with the activity of p-Aurora A, p-TACC3 and p-LIMK1.
Collapse
Affiliation(s)
- Lei Peng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (L.P.); (Y.H.); (W.W.)
| | - Yijing He
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (L.P.); (Y.H.); (W.W.)
| | - Weihan Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (L.P.); (Y.H.); (W.W.)
| | - Jianjun Dai
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China;
| | - Qiao Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (L.P.); (Y.H.); (W.W.)
| | - Shiqiang Ju
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (L.P.); (Y.H.); (W.W.)
| |
Collapse
|
3
|
Liu Q, Cheng C, Huang J, Yan W, Wen Y, Liu Z, Zhou B, Guo S, Fang W. MYH9: A key protein involved in tumor progression and virus-related diseases. Biomed Pharmacother 2024; 171:116118. [PMID: 38181716 DOI: 10.1016/j.biopha.2023.116118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of non-muscle myosin IIA (NMIIA), which belongs to the myosin II subfamily of actin-based molecular motors. Previous studies have demonstrated that abnormal expression and mutations of MYH9 were correlated with MYH9-related diseases and tumors. Furthermore, earlier investigations identified MYH9 as a tumor suppressor. However, subsequent research revealed that MYH9 promoted tumorigenesis, progression and chemoradiotherapy resistance. Note-worthily, MYH9 has also been linked to viral infections, like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Epstein-Barr virus, and hepatitis B virus, as a receptor or co-receptor. In addition, MYH9 promotes the development of hepatocellular carcinoma by interacting with the hepatitis B virus-encoding X protein. Finally, various findings highlighted the role of MYH9 in the development of these illnesses, especially in tumors. This review summarizes the involvement of the MYH9-regulated signaling network in tumors and virus-related diseases and presents possible drug interventions on MYH9, providing insights for the use of MYH9 as a therapeutic target for tumors and virus-mediated diseases.
Collapse
Affiliation(s)
- Qing Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Chao Cheng
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology hospital, Shenzhen 518000, China
| | - Jiyu Huang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Weiwei Yan
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Yinhao Wen
- Department of Oncology, Pingxiang People's Hospital, Pingxiang 337000, China
| | - Zhen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China; Key Laboratory of Protein Modification and Degradation, Basic School of Guangzhou Medical University, Guangzhou 510315, China.
| | - Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou 525200, China.
| | - Suiqun Guo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510315, China.
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China; The People's Hospital of Gaozhou, Gaozhou 525200, China; Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510315, China.
| |
Collapse
|
4
|
Lobos Patorniti N, Zulkefli KL, McAdam ME, Vargas P, Bakke O, Progida C. Rai14 is a novel interactor of Invariant chain that regulates macropinocytosis. Front Immunol 2023; 14:1182180. [PMID: 37545539 PMCID: PMC10401043 DOI: 10.3389/fimmu.2023.1182180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
Invariant chain (Ii, CD74) is a type II transmembrane glycoprotein that acts as a chaperone and facilitates the folding and transport of MHC II chains. By assisting the assembly and subcellular targeting of MHC II complexes, Ii has a wide impact on the functions of antigen-presenting cells such as antigen processing, endocytic maturation, signal transduction, cell migration, and macropinocytosis. Ii is a multifunctional molecule that can alter endocytic traffic and has several interacting molecules. To understand more about Ii's function and to identify further Ii interactors, a yeast two-hybrid screening was performed. Retinoic Acid-Induced 14 (Rai14) was detected as a putative interaction partner, and the interaction was confirmed by co-immunoprecipitation. Rai14 is a poorly characterized protein, which is believed to have a role in actin cytoskeleton and membrane remodeling. In line with this, we found that Rai14 localizes to membrane ruffles, where it forms macropinosomes. Depletion of Rai14 in antigen-presenting cells delays MHC II internalization, affecting macropinocytic activity. Intriguingly, we demonstrated that, similar to Ii, Rai14 is a positive regulator of macropinocytosis and a negative regulator of cell migration, two antagonistic processes in antigen-presenting cells. This antagonism is known to depend on the interaction between myosin II and Ii. Here, we show that Rai14 also binds to myosin II, suggesting that Ii, myosin II, and Rai14 work together to coordinate macropinocytosis and cell motility.
Collapse
Affiliation(s)
| | | | | | - Pablo Vargas
- Inserm U1151, Institut Necker Enfants Malades, Paris, France
| | - Oddmund Bakke
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Zhao AJ, Montes-Laing J, Perry WMG, Shiratori M, Merfeld E, Rogers SL, Applewhite DA. The Drosophila spectraplakin Short stop regulates focal adhesion dynamics by crosslinking microtubules and actin. Mol Biol Cell 2022; 33:ar19. [PMID: 35235367 PMCID: PMC9282009 DOI: 10.1091/mbc.e21-09-0434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The spectraplakin family of proteins includes ACF7/MACF1 and BPAG1/dystonin in mammals, VAB-10 in Caenorhabditis elegans, Magellan in zebrafish, and Short stop (Shot), the sole Drosophila member. Spectraplakins are giant cytoskeletal proteins that cross-link actin, microtubules, and intermediate filaments, coordinating the activity of the entire cytoskeleton. We examined the role of Shot during cell migration using two systems: the in vitro migration of Drosophila tissue culture cells and in vivo through border cell migration. RNA interference (RNAi) depletion of Shot increases the rate of random cell migration in Drosophila tissue culture cells as well as the rate of wound closure during scratch-wound assays. This increase in cell migration prompted us to analyze focal adhesion dynamics. We found that the rates of focal adhesion assembly and disassembly were faster in Shot-depleted cells, leading to faster adhesion turnover that could underlie the increased migration speeds. This regulation of focal adhesion dynamics may be dependent on Shot being in an open confirmation. Using Drosophila border cells as an in vivo model for cell migration, we found that RNAi depletion led to precocious border cell migration. Collectively, these results suggest that spectraplakins not only function to cross-link the cytoskeleton but may regulate cell–matrix adhesion.
Collapse
Affiliation(s)
- Andrew J Zhao
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Julia Montes-Laing
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Wick M G Perry
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Mari Shiratori
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Emily Merfeld
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| | - Stephen L Rogers
- Department of Biology & Carolina Center for Genome Sciences, The University of North Carolina at Chapel Hill, Campus Box 3280, 422 Fordham Hall, Chapel Hill, NC 27599-3280, USA
| | - Derek A Applewhite
- Department of Biology, Reed College, 3203 SE Woodstock Boulevard, Portland, OR 97202, USA
| |
Collapse
|
6
|
Chandra A, Butler MT, Bear JE, Haugh JM. Modeling cell protrusion predicts how myosin II and actin turnover affect adhesion-based signaling. Biophys J 2022; 121:102-118. [PMID: 34861242 PMCID: PMC8758409 DOI: 10.1016/j.bpj.2021.11.2889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/03/2021] [Accepted: 11/29/2021] [Indexed: 01/07/2023] Open
Abstract
Orchestration of cell migration is essential for development, tissue regeneration, and the immune response. This dynamic process integrates adhesion, signaling, and cytoskeletal subprocesses across spatial and temporal scales. In mesenchymal cells, adhesion complexes bound to extracellular matrix mediate both biochemical signal transduction and physical interaction with the F-actin cytoskeleton. Here, we present a mathematical model that offers insight into both aspects, considering spatiotemporal dynamics of nascent adhesions, active signaling molecules, mechanical clutching, actin treadmilling, and nonmuscle myosin II contractility. At the core of the model is a positive feedback loop, whereby adhesion-based signaling promotes generation of barbed ends at, and protrusion of, the cell's leading edge, which in turn promotes formation and stabilization of nascent adhesions. The model predicts a switch-like transition and optimality of membrane protrusion, determined by the balance of actin polymerization and retrograde flow, with respect to extracellular matrix density. The model, together with new experimental measurements, explains how protrusion can be modulated by mechanical effects (nonmuscle myosin II contractility and adhesive bond stiffness) and F-actin turnover.
Collapse
Affiliation(s)
- Ankit Chandra
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - James E Bear
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina.
| |
Collapse
|
7
|
Wang Y, Guo F. Group I PAKs in myelin formation and repair of the central nervous system: what, when, and how. Biol Rev Camb Philos Soc 2021; 97:615-639. [PMID: 34811887 DOI: 10.1111/brv.12815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/20/2021] [Accepted: 11/04/2021] [Indexed: 11/30/2022]
Abstract
p21-activated kinases (PAKs) are a family of cell division control protein 42/ras-related C3 botulinum toxin substrate 1 (Cdc42/Rac1)-activated serine/threonine kinases. Group I PAKs (PAK1-3) have distinct activation mechanisms from group II PAKs (PAK4-6) and are the focus of this review. In transformed cancer cells, PAKs regulate a variety of cellular processes and molecular pathways which are also important for myelin formation and repair in the central nervous system (CNS). De novo mutations in group I PAKs are frequently seen in children with neurodevelopmental defects and white matter anomalies. Group I PAKs regulate virtually every aspect of neuronal development and function. Yet their functions in CNS myelination and remyelination remain incompletely defined. Herein, we highlight the current understanding of PAKs in regulating cellular and molecular pathways and discuss the status of PAK-regulated pathways in oligodendrocyte development. We point out outstanding questions and future directions in the research field of group I PAKs and oligodendrocyte development.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurology, Shriners Hospitals for Children/School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), University of California, Davis, 2425 Stockton Blvd, Sacramento, CA, 95817, U.S.A
| | - Fuzheng Guo
- Department of Neurology, Shriners Hospitals for Children/School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), University of California, Davis, 2425 Stockton Blvd, Sacramento, CA, 95817, U.S.A
| |
Collapse
|
8
|
PAK1 Silencing Attenuated Proinflammatory Macrophage Activation and Foam Cell Formation by Increasing PPAR γ Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6957900. [PMID: 34603600 PMCID: PMC8483905 DOI: 10.1155/2021/6957900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/28/2021] [Indexed: 12/24/2022]
Abstract
Macrophage polarization in response to environmental cues has emerged as an important event in the development of atherosclerosis. Compelling evidences suggest that P21-activated kinases 1 (PAK1) is involved in a wide variety of diseases. However, the potential role and mechanism of PAK1 in regulation of macrophage polarization remains to be elucidated. Here, we observed that PAK1 showed a dramatically increased expression in M1 macrophages but decreased expression in M2 macrophages by using a well-established in vitro model to study heterogeneity of macrophage polarization. Adenovirus-mediated loss-of-function approach demonstrated that PAK1 silencing induced an M2 macrophage phenotype-associated gene profiles but repressed the phenotypic markers related to M1 macrophage polarization. Additionally, dramatically decreased foam cell formation was found in PAK1 silencing-induced M2 macrophage activation which was accompanied with alternation of marker account for cholesterol efflux or influx from macrophage foam cells. Moderate results in lipid metabolism and foam cell formation were found in M1 macrophage activation mediated by AdshPAK1. Importantly, we presented mechanistic evidence that PAK1 knockdown promoted the expression of PPARγ, and the effect of macrophage activation regulated by PAK1 silencing was largely reversed when a PPARγ antagonist was utilized. Collectively, these findings reveal that PAK1 is an independent effector of macrophage polarization at least partially attributed to regulation of PPARγ expression, which suggested PAK1-PPARγ axis as a novel therapeutic strategy in atherosclerosis management.
Collapse
|
9
|
Ostrowska-Podhorodecka Z, Ding I, Lee W, Tanic J, Abbasi S, Arora PD, Liu RS, Patteson AE, Janmey PA, McCulloch CA. Vimentin tunes cell migration on collagen by controlling β1 integrin activation and clustering. J Cell Sci 2021; 134:jcs.254359. [PMID: 33558312 DOI: 10.1242/jcs.254359] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Vimentin is a structural protein that is required for mesenchymal cell migration and directly interacts with actin, β1 integrin and paxillin. We examined how these interactions enable vimentin to regulate cell migration on collagen. In fibroblasts, depletion of vimentin increased talin-dependent activation of β1 integrin by more than 2-fold. Loss of vimentin was associated with reduction of β1 integrin clustering by 50% and inhibition of paxillin recruitment to focal adhesions by more than 60%, which was restored by vimentin expression. This reduction of paxillin was associated with 65% lower Cdc42 activation, a 60% reduction of cell extension formation and a greater than 35% decrease in cell migration on collagen. The activation of PAK1, a downstream effector of Cdc42, was required for vimentin phosphorylation and filament maturation. We propose that vimentin tunes cell migration through collagen by acting as an adaptor protein for focal adhesion proteins, thereby regulating β1 integrin activation, resulting in well-organized, mature integrin clusters.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Isabel Ding
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Wilson Lee
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Jelena Tanic
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Sevil Abbasi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Richard S Liu
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Alison E Patteson
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA.,Physics Department, Syracuse University, Syracuse, NY 13244, USA
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA
| | | |
Collapse
|
10
|
Cell stretchers and the LINC complex in mechanotransduction. Arch Biochem Biophys 2021; 702:108829. [PMID: 33716002 DOI: 10.1016/j.abb.2021.108829] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023]
Abstract
How cells respond to mechanical forces from the surrounding environment is critical for cell survival and function. The LINC complex is a central component in the mechanotransduction pathway that transmits mechanical information from the cell surface to the nucleus. Through LINC complex functionality, the nucleus is able to respond to mechanical stress by altering nuclear structure, chromatin organization, and gene expression. The use of specialized devices that apply mechanical strain to cells have been central to investigating how mechanotransduction occurs, how cells respond to mechanical stress, and the role of the LINC complexes in these processes. A large variety of designs have been reported for these devices, with the most common type being cell stretchers. Here we highlight some of the salient features of cell stretchers and suggest some key parameters that should be considered when using these devices. We provide a brief overview of how the LINC complexes contribute to the cellular responses to mechanical strain. And finally, we suggest that stretchers may be a useful tool to study aging.
Collapse
|
11
|
Bilotta AJ, Ma C, Yang W, Yu Y, Yu Y, Zhao X, Zhou Z, Yao S, Dann SM, Cong Y. Propionate Enhances Cell Speed and Persistence to Promote Intestinal Epithelial Turnover and Repair. Cell Mol Gastroenterol Hepatol 2020; 11:1023-1044. [PMID: 33238220 PMCID: PMC7898181 DOI: 10.1016/j.jcmgh.2020.11.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Gut bacteria-derived short-chain fatty acids (SCFAs) play crucial roles in the maintenance of intestinal homeostasis. However, how SCFAs regulate epithelial turnover and tissue repair remain incompletely understood. In this study, we investigated how the SCFA propionate regulates cell migration to promote epithelial renewal and repair. METHODS Mouse small intestinal epithelial cells (MSIE) and human Caco-2 cells were used to determine the effects of SCFAs on gene expression, proliferation, migration, and cell spreading in vitro. Video microscopy and single cell tracking were used to assess cell migration kinetically. 5-bromo-2'-deoxyuridine (BrdU) and hydroxyurea were used to assess the effects of SCFAs on migration in vivo. Lastly, an acute colitis model using dextran sulfate sodium (DSS) was used to examine the effects of SCFAs in vivo. RESULTS Using video microscopy and single cell tracking, we found that propionate promoted intestinal epithelial cell migration by enhancing cell spreading and polarization, which led to increases in both cell speed and persistence. This novel function of propionate was dependent on inhibition of class I histone deacetylases (HDAC) and GPR43 and required signal transducer and activator of transcription 3 (STAT3). Furthermore, using 5-bromo-2'-deoxyuridine (BrdU) and hydroxyurea in vivo, we found that propionate enhanced cell migration up the crypt-villus axis under homeostatic conditions, while also protecting against ulcer formation in experimental colitis. CONCLUSION Our results demonstrate a mechanism by which propionate stimulates cell migration in an HDAC inhibition, GPR43, and STAT3 dependent manner, and suggest that propionate plays an important role in epithelial migration independent of proliferation.
Collapse
Affiliation(s)
- Anthony J. Bilotta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Chunyan Ma
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas,Department of Central Laboratory, Shandong Provincial Hospital Shandong First Medical University, Jinan, China
| | - Wenjing Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Yanbo Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Yu Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Xiaojing Zhao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Zheng Zhou
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Sara M. Dann
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas,Department of Pathology, University of Texas Medical Branch, Galveston, Texas,Correspondence Address correspondence to: Yingzi Cong, PhD, Department of Microbiology and Immunology, University of Texas Medical Branch, 4.142C Medical Research Building, 301 University Boulevard, Galveston, Texas 77555-1019. fax: (409) 772-5065.
| |
Collapse
|
12
|
Chang H, He KY, Li C, Ni YY, Li MN, Chen L, Hou M, Zhou Z, Xu ZP, Ji MJ. P21 activated kinase-1 (PAK1) in macrophages is required for promotion of Th17 cell response during helminth infection. J Cell Mol Med 2020; 24:14325-14338. [PMID: 33124146 PMCID: PMC7753984 DOI: 10.1111/jcmm.16050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
CD4+T cells differentiate into distinct functional effector and inhibitory subsets are facilitated by distinct cytokine cues present at the time of antigen recognition. Maintaining a balance between T helper 17 (Th17) and regulatory T (Treg) cells are critical for the control of the immunopathogenesis of liver diseases. Here, by using the mouse model of helminth Schistosoma japonicum (Sjaponicum) infection, we show that the hepatic mRNA levels of P21‐activated kinase 1 (PAK1), a key regulator of the actin cytoskeleton, adhesion and cell motility, are significantly increased and associated with the development of liver pathology during Sjaponicum infection. In addition, PAK1‐deficient mice are prone to suppression of Th17 cell responses but increased Treg cells. Furthermore, PAK1 enhances macrophage activation through promoting IRF1 nuclear translocation in an NF‐κB‐dependent pathway, resulting in promoting Th17 cell differentiation through inducing IL‐6 production. These findings highlight the importance of PAK1 in macrophages fate determination and suggest that PAK1/IRF1 axis‐dependent immunomodulation can ameliorate certain T cell–based immune pathologies.
Collapse
Affiliation(s)
- Hao Chang
- Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Kai-Yue He
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Chen Li
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Yang-Yue Ni
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Mai-Ning Li
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Lin Chen
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Min Hou
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Zikai Zhou
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Peng Xu
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Min-Jun Ji
- Center for Global Health, Nanjing Medical University, Nanjing, China.,Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Zhang M, Fang X, Wang C, Hua Y, Huang C, Wang M, Zhu L, Wang Z, Gao Y, Zhang T, Liu H, Zhang Y, Lu S, Lu T, Chen Y, Li H. Design and synthesis of 1H-indazole-3-carboxamide derivatives as potent and selective PAK1 inhibitors with anti-tumour migration and invasion activities. Eur J Med Chem 2020; 203:112517. [DOI: 10.1016/j.ejmech.2020.112517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/15/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022]
|
14
|
Merino-Cortés SV, Gardeta SR, Roman-Garcia S, Martínez-Riaño A, Pineau J, Liebana R, Merida I, Dumenil AML, Pierobon P, Husson J, Alarcon B, Carrasco YR. Diacylglycerol kinase ζ promotes actin cytoskeleton remodeling and mechanical forces at the B cell immune synapse. Sci Signal 2020; 13:13/627/eaaw8214. [PMID: 32291315 DOI: 10.1126/scisignal.aaw8214] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Diacylglycerol kinases (DGKs) limit antigen receptor signaling in immune cells by consuming the second messenger diacylglycerol (DAG) to generate phosphatidic acid (PA). Here, we showed that DGKζ promotes lymphocyte function-associated antigen 1 (LFA-1)-mediated adhesion and F-actin generation at the immune synapse of B cells with antigen-presenting cells (APCs), mostly in a PA-dependent manner. Measurement of single-cell mechanical force generation indicated that DGKζ-deficient B cells exerted lower forces at the immune synapse than did wild-type B cells. Nonmuscle myosin activation and translocation of the microtubule-organizing center (MTOC) to the immune synapse were also impaired in DGKζ-deficient B cells. These functional defects correlated with the decreased ability of B cells to present antigen and activate T cells in vitro. The in vivo germinal center response of DGKζ-deficient B cells was also reduced compared with that of wild-type B cells, indicating that loss of DGKζ in B cells impaired T cell help. Together, our data suggest that DGKζ shapes B cell responses by regulating actin remodeling, force generation, and antigen uptake-related events at the immune synapse. Hence, an appropriate balance in the amounts of DAG and PA is required for optimal B cell function.
Collapse
Affiliation(s)
- Sara V Merino-Cortés
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Sofia R Gardeta
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Sara Roman-Garcia
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Ana Martínez-Riaño
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC-UAM, Madrid, Spain
| | - Judith Pineau
- Institut Curie, PSL Research University, INSERM U932, Paris, France.,Université de Paris, 75006, Paris, France
| | - Rosa Liebana
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Isabel Merida
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | | | - Paolo Pierobon
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Julien Husson
- Laboratoire d'Hydrodynamique (LadHyx), Ecole polytechnique, CNRS, Institut Polytechnique de Paris, Paris, France
| | - Balbino Alarcon
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC-UAM, Madrid, Spain
| | - Yolanda R Carrasco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain.
| |
Collapse
|
15
|
Kiepas A, Voorand E, Mubaid F, Siegel PM, Brown CM. Optimizing live-cell fluorescence imaging conditions to minimize phototoxicity. J Cell Sci 2020; 133:jcs242834. [PMID: 31988150 DOI: 10.1242/jcs.242834] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/09/2020] [Indexed: 08/31/2023] Open
Abstract
Fluorescence illumination can cause phototoxicity that negatively affects living samples. This study demonstrates that much of the phototoxicity and photobleaching experienced with live-cell fluorescence imaging occurs as a result of 'illumination overhead' (IO). This occurs when a sample is illuminated but fluorescence emission is not being captured by the microscope camera. Several technological advancements have been developed, including fast-switching LED lamps and transistor-transistor logic (TTL) circuits, to diminish phototoxicity caused by IO. These advancements are not standard features on most microscopes and many biologists are unaware of their necessity for live-cell imaging. IO is particularly problematic when imaging rapid processes that require short exposure times. This study presents a workflow to optimize imaging conditions for measuring both slow and dynamic processes while minimizing phototoxicity on any standard microscope. The workflow includes a guide on how to (1) determine the maximum image exposure time for a dynamic process, (2) optimize excitation light intensity and (3) assess cell health with mitochondrial markers.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Alex Kiepas
- Department of Physiology, McGill University, Montreal, Canada, H3G 1Y6
- Goodman Cancer Research Centre, McGill University, Canada, H3G 1A1
| | - Elena Voorand
- Goodman Cancer Research Centre, McGill University, Canada, H3G 1A1
- Department of Biochemistry, McGill University, Montreal, Canada, H3G 1Y6
| | - Firas Mubaid
- Department of Physiology, McGill University, Montreal, Canada, H3G 1Y6
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, Canada, H3G 1A1
- Department of Biochemistry, McGill University, Montreal, Canada, H3G 1Y6
- Department of Medicine, McGill University, Montreal, Canada, H4A 3J1
- Department of Anatomy & Cell Biology, McGill University, Canada, H3G 0B1
| | - Claire M Brown
- Department of Physiology, McGill University, Montreal, Canada, H3G 1Y6
- Department of Anatomy & Cell Biology, McGill University, Canada, H3G 0B1
- Advanced BioImaging Facility (ABIF), McGill University, Montreal, Canada, H3A 0C7
- Cell Information Systems, McGill University, Montreal, Canada, H3G 0B1
- Centre for Applied Mathematics in Bioscience and Medicine (CAMBAM), McGill University, Montreal, Canada, H3G 1Y6
| |
Collapse
|
16
|
Chen KJ, Chiang TC, Yu CJ, Lee FJS. Cooperative recruitment of Arl4A and Pak1 to the plasma membrane contributes to sustained Pak1 activation for cell migration. J Cell Sci 2020; 133:jcs233361. [PMID: 31932503 DOI: 10.1242/jcs.233361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/17/2019] [Indexed: 01/27/2023] Open
Abstract
Cell migration requires the coordination of multiple signaling pathways involved in membrane dynamics and cytoskeletal rearrangement. The Arf-like small GTPase Arl4A has been shown to modulate actin cytoskeleton remodeling. However, evidence of the function of Arl4A in cell migration is insufficient. Here, we report that Arl4A acts with the serine/threonine protein kinase Pak1 to modulate cell migration through their cooperative recruitment to the plasma membrane. We first observed that Arl4A and its isoform Arl4D interact with Pak1 and Pak2 and showed that Arl4A recruits Pak1 and Pak2 to the plasma membrane. The fibronectin-induced Pak1 localization at the plasma membrane is reduced in Arl4A-depleted cells. Unexpectedly, we found that Pak1, but not Arl4A-binding-defective Pak1, can recruit a cytoplasmic myristoylation-deficient Arl4A-G2A mutant to the plasma membrane. Furthermore, we found that the Arl4A-Pak1 interaction, which is independent of Rac1 binding to Pak1, is required for Arl4A-induced cell migration. Thus, we infer that there is feedback regulation between Arl4A and Pak1, in which they mutually recruit each other to the plasma membrane for Pak1 activation, thereby modulating cell migration through direct interaction.
Collapse
Affiliation(s)
- Kuan-Jung Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Tsai-Chen Chiang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Chia-Jung Yu
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Fang-Jen S Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| |
Collapse
|
17
|
Mierke CT, Puder S, Aermes C, Fischer T, Kunschmann T. Effect of PAK Inhibition on Cell Mechanics Depends on Rac1. Front Cell Dev Biol 2020; 8:13. [PMID: 32047750 PMCID: PMC6997127 DOI: 10.3389/fcell.2020.00013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
Besides biochemical and molecular regulation, the migration and invasion of cells is controlled by the environmental mechanics and cellular mechanics. Hence, the mechanical phenotype of cells, such as fibroblasts, seems to be crucial for the migratory capacity in confined 3D extracellular matrices. Recently, we have shown that the migratory and invasive capacity of mouse embryonic fibroblasts depends on the expression of the Rho-GTPase Rac1, similarly it has been demonstrated that the Rho-GTPase Cdc42 affects cell motility. The p21-activated kinase (PAK) is an effector down-stream target of both Rho-GTPases Rac1 and Cdc42, and it can activate via the LIM kinase-1 its down-stream target cofilin and subsequently support the cell migration and invasion through the polymerization of actin filaments. Since Rac1 deficient cells become mechanically softer than controls, we investigated the effect of group I PAKs and PAK1 inhibition on cell mechanics in the presence and absence of Rac1. Therefore, we determined whether mouse embryonic fibroblasts, in which Rac1 was knocked-out, and control cells, displayed cell mechanical alterations after treatment with group I PAKs or PAK1 inhibitors using a magnetic tweezer (adhesive cell state) and an optical cell stretcher (non-adhesive cell state). In fact, we found that group I PAKs and Pak1 inhibition decreased the stiffness and the Young’s modulus of fibroblasts in the presence of Rac1 independent of their adhesive state. However, in the absence of Rac1 the effect was abolished in the adhesive cell state for both inhibitors and in their non-adhesive state, the effect was abolished for the FRAX597 inhibitor, but not for the IPA3 inhibitor. The migration and invasion were additionally reduced by both PAK inhibitors in the presence of Rac1. In the absence of Rac1, only FRAX597 inhibitor reduced their invasiveness, whereas IPA3 had no effect. These findings indicate that group I PAKs and PAK1 inhibition is solely possible in the presence of Rac1 highlighting Rac1/PAK I (PAK1, 2, and 3) as major players in cell mechanics.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| | - Stefanie Puder
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| | - Christian Aermes
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| | - Tony Fischer
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| | - Tom Kunschmann
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
18
|
Paxillin S273 Phosphorylation Regulates Adhesion Dynamics and Cell Migration through a Common Protein Complex with PAK1 and βPIX. Sci Rep 2019; 9:11430. [PMID: 31391572 PMCID: PMC6686007 DOI: 10.1038/s41598-019-47722-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Cell migration is an important biological phenomenon involved in many homeostatic and aberrant physiological processes. Phosphorylation of the focal adhesion adaptor protein, paxillin, on serine 273 (S273) has been implicated as a key regulator of cell migration. Here, it is shown that phosphorylation on paxillin S273 leads to highly migratory cells with small dynamic adhesions. Adhesions at protrusive edges of the cell were more dynamic than adhesions at retracting edges. Temporal image correlation microscopy revealed that these dynamic adhesions undergo rapid binding of paxillin, PAK1 and βPIX. We identified membrane proximal adhesion subdomains in protrusive regions of the cell that show rapid protein binding that is dependent on paxillin S273 phosphorylation, PAK1 kinase activity and phosphatases. These dynamic adhesion subdomains corresponded to regions of the adhesion that also show co-binding of paxillin/PAK1 and paxillin/βPIX complexes. It is likely that parts of individual adhesions are more dynamic while others are less dynamic due to their association with the actin cytoskeleton. Variable adhesion and binding dynamics are regulated via differential paxillin S273 phosphorylation across the cell and within adhesions and are required for regulated cell migration. Dysregulation through phosphomutants, PAK1-KD or βPIX mutants resulted in large stable adhesions, long protein binding times and slow cell migration. Dysregulation through phosphomimics or PAK1-CA led to small dynamic adhesions and rapid cell migration reminiscent of highly migratory cancer cells. Thus, phosphorylation of paxillin S273 is a key regulator of cell migration through recruitment of βPIX and PAK1 to sites of adhesion.
Collapse
|
19
|
Campbell HK, Salvi AM, O'Brien T, Superfine R, DeMali KA. PAK2 links cell survival to mechanotransduction and metabolism. J Cell Biol 2019; 218:1958-1971. [PMID: 30940647 PMCID: PMC6548143 DOI: 10.1083/jcb.201807152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/29/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022] Open
Abstract
Campbell et al. show that force stimulates PAK2 activation at cell–cell junctions, where it protects cells under force from death and plays a key role in linking force-induced mechanotransduction, metabolism, and cell survival. Too little or too much force can trigger cell death, yet factors that ensure the survival of cells remain largely unknown. Here, we demonstrate that E-cadherin responds to force by recruiting and activating p21-activated protein kinase 2 (PAK2) to allow cells to stiffen, metabolize, and survive. Interestingly, PAK2 activation and its control of the apoptotic response are specific for the amplitude of force applied. Specifically, under low amplitudes of physiological force, PAK2 is protected from proteolysis, thereby ensuring cell survival. In contrast, under higher amplitudes of physiological force, PAK2 is left unprotected and stimulates apoptosis, an effect that is prevented by cleavage-resistant forms of the protein. Finally, we demonstrate that PAK2 protection is conferred by direct binding of AMPK. Thus, PAK2 mediates the survival of cells under force. These findings reveal an unexpected paradigm for how mechanotransduction, metabolism, and cell survival are linked.
Collapse
Affiliation(s)
- Hannah K Campbell
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Alicia M Salvi
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Timothy O'Brien
- Department of Physics, University of North Carolina, Chapel Hill, NC
| | - Richard Superfine
- Department of Physics, University of North Carolina, Chapel Hill, NC
| | - Kris A DeMali
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
20
|
Modulation of store-operated calcium entry and nascent adhesion by p21-activated kinase 1. Exp Mol Med 2018; 50:1-10. [PMID: 29780159 PMCID: PMC5960643 DOI: 10.1038/s12276-018-0093-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/13/2018] [Accepted: 03/13/2018] [Indexed: 01/30/2023] Open
Abstract
Calcium mobilization is necessary for cell movement during embryonic development, lymphocyte synapse formation, wound healing, and cancer cell metastasis. Depletion of calcium in the lumen of the endoplasmic reticulum using inositol triphosphate (IP3) or thapsigargin (TG) is known to induce oligomerization and cytoskeleton-mediated translocation of stromal interaction molecule 1 (STIM1) to the plasma membrane, where it interacts with the calcium release-activated calcium channel Orai1 to mediate calcium influx; this process is referred to as store-operated calcium entry (SOCE). Furthermore, aberrant STIM1 or SOCE regulation is associated with cancer cell motility and metastasis. The p21-activated kinases (PAKs), which are downstream effectors of GTPases, reportedly regulate cytoskeletal organization, protrusive activity, and cell migration. Although cytoskeletal remodeling apparently contributes to calcium mobilization via SOCE, and vice versa, the mechanisms by which they regulate each other remain unclear. In this study, we aimed to characterize whether PAK1 modulates calcium mobilization and STIM1 localization. Our data demonstrate that PAK1 interacts with STIM1 in vitro and that this interaction was enhanced by treatment with a nascent adhesion inducer, such as phorbol 12,13-dibutyrate (PDBu). Under basal conditions, both proteins appeared to primarily colocalize in the cytosol, whereas treatment with PDBu induced their colocalization to vinculin-positive peripheral adhesions. Downregulation of PAK1 activity via chemical inhibitors or by PAK1 shDNA expression impaired STIM1-mediated calcium mobilization via SOCE. Based on these findings, we propose that PAK1 interacts with STIM1 to regulate calcium mobilization and the formation of cellular adhesions. A molecular mechanism underlying cell movement may contribute to the aggressive migration of metastatic tumor cells. A team led by Ki-Duk Song at Chonbuk National University, Jeonju-si, and Joong-Kook Choi at Chungbuk National University, Cheongju in South Korea investigated the function of a protein called p21-activated kinase 1 (PAK1). PAK1 is known to contribute to the reorganization of cellular structure. The researchers determined that it directly interacts with molecular machinery that controls the storage and release of stockpiled calcium ions at the periphery of the cell where migration takes place. These ions play an important role in enabling cell movement and attachment, and the researchers showed that they could disrupt cellular calcium ion accumulation by switching off the gene encoding PAK1. They now aim to investigate how this mechanism contributes to cancer cell migration.
Collapse
|
21
|
Hyperactivity of Rac1-GTPase pathway impairs neuritogenesis of cortical neurons by altering actin dynamics. Sci Rep 2018; 8:7254. [PMID: 29740022 PMCID: PMC5940682 DOI: 10.1038/s41598-018-25354-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/18/2018] [Indexed: 01/10/2023] Open
Abstract
The small-GTPase Rac1 is a key molecular regulator linking extracellular signals to actin cytoskeleton dynamics. Loss-of-function mutations in RAC1 and other genes of the Rac signaling pathway have been implicated in the pathogenesis of Intellectual Disability (ID). The Rac1 activity is negatively controlled by GAP proteins, however the effect of Rac1 hyperactivity on neuronal networking in vivo has been poorly studied. ArhGAP15 is a Rac-specific negative regulator, expressed in the main subtypes of pyramidal cortical neurons. In the absence of ArhGAP15, cortical pyramidal neurons show defective neuritogenesis, delayed axonal elongation, reduced dendritic branching, both in vitro and in vivo. These phenotypes are associated with altered actin dynamics at the growth cone due to increased activity of the PAK-LIMK pathway and hyperphosphorylation of ADF/cofilin. These results can be explained by shootin1 hypo-phosphorylation and uncoupling with the adhesion system. Functionally, ArhGAP15−/− mice exhibit decreased synaptic density, altered electroencephalographic rhythms and cognitive deficits. These data suggest that both hypo- and hyperactivation of the Rac pathway due to mutations in Rac1 regulators can result in conditions of ID, and that a tight regulation of Rac1 activity is required to attain the full complexity of the cortical networks.
Collapse
|
22
|
Meyer zum Büschenfelde U, Brandenstein LI, von Elsner L, Flato K, Holling T, Zenker M, Rosenberger G, Kutsche K. RIT1 controls actin dynamics via complex formation with RAC1/CDC42 and PAK1. PLoS Genet 2018; 14:e1007370. [PMID: 29734338 PMCID: PMC5937737 DOI: 10.1371/journal.pgen.1007370] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
Abstract
RIT1 belongs to the RAS family of small GTPases. Germline and somatic RIT1 mutations have been identified in Noonan syndrome (NS) and cancer, respectively. By using heterologous expression systems and purified recombinant proteins, we identified the p21-activated kinase 1 (PAK1) as novel direct effector of RIT1. We found RIT1 also to directly interact with the RHO GTPases CDC42 and RAC1, both of which are crucial regulators of actin dynamics upstream of PAK1. These interactions are independent of the guanine nucleotide bound to RIT1. Disease-causing RIT1 mutations enhance protein-protein interaction between RIT1 and PAK1, CDC42 or RAC1 and uncouple complex formation from serum and growth factors. We show that the RIT1-PAK1 complex regulates cytoskeletal rearrangements as expression of wild-type RIT1 and its mutant forms resulted in dissolution of stress fibers and reduction of mature paxillin-containing focal adhesions in COS7 cells. This effect was prevented by co-expression of RIT1 with dominant-negative CDC42 or RAC1 and kinase-dead PAK1. By using a transwell migration assay, we show that RIT1 wildtype and the disease-associated variants enhance cell motility. Our work demonstrates a new function for RIT1 in controlling actin dynamics via acting in a signaling module containing PAK1 and RAC1/CDC42, and highlights defects in cell adhesion and migration as possible disease mechanism underlying NS. Noonan syndrome (NS) belongs to the RASopathies, a group of developmental diseases caused by mutations in genes encoding RAS-MAPK pathway components. Germline mutations in RIT1 have been identified in NS. RIT1 belongs to the RAS superfamily, however, the cellular function of RIT1 remains elusive. We show that RIT1 binds p21-activated kinase 1 (PAK1), an effector of the RHO GTPases RAC1 and CDC42, which are important regulators of cytoskeletal dynamics. NS-associated RIT1 mutants enhance complex formation between RIT1, RAC1/CDC42 and PAK1. Expression of wild-type or mutant forms of RIT1 caused loss of stress fibers and mature focal adhesions and enhanced cell motility. Our data suggest that dysfunction in actin dynamics is a novel aspect in the pathophysiology of RASopathies.
Collapse
Affiliation(s)
| | | | - Leonie von Elsner
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Flato
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tess Holling
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Georg Rosenberger
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail: (KK); (GR)
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail: (KK); (GR)
| |
Collapse
|
23
|
Diggins NL, Kang H, Weaver A, Webb DJ. α5β1 integrin trafficking and Rac activation are regulated by APPL1 in a Rab5-dependent manner to inhibit cell migration. J Cell Sci 2018; 131:jcs.207019. [PMID: 29361527 DOI: 10.1242/jcs.207019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/09/2018] [Indexed: 01/04/2023] Open
Abstract
Cell migration is a tightly coordinated process that requires the spatiotemporal regulation of many molecular components. Because adaptor proteins can serve as integrators of cellular events, they are being increasingly studied as regulators of cell migration. The adaptor protein containing a pleckstrin-homology (PH) domain, phosphotyrosine binding (PTB) domain, and leucine zipper motif 1 (APPL1) is a 709 amino acid endosomal protein that plays a role in cell proliferation and survival as well as endosomal trafficking and signaling. However, its function in regulating cell migration is poorly understood. Here, we show that APPL1 hinders cell migration by modulating both trafficking and signaling events controlled by Rab5 in cancer cells. APPL1 decreases internalization and increases recycling of α5β1 integrin, leading to higher levels of α5β1 integrin at the cell surface that hinder adhesion dynamics. Furthermore, APPL1 decreases the activity of the GTPase Rac and its effector PAK, which in turn regulate cell migration. Thus, we demonstrate a novel role for the interaction between APPL1 and Rab5 in governing crosstalk between signaling and trafficking pathways on endosomes to affect cancer cell migration.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Nicole L Diggins
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Hakmook Kang
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alissa Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Donna J Webb
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA.,Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
24
|
Pi J, Liu J, Zhuang T, Zhang L, Sun H, Chen X, Zhao Q, Kuang Y, Peng S, Zhou X, Yu Z, Tao T, Tomlinson B, Chan P, Tian Y, Fan H, Liu Z, Zheng X, Morrisey E, Zhang Y. Elevated Expression of miR302-367 in Endothelial Cells Inhibits Developmental Angiogenesis via CDC42/CCND1 Mediated Signaling Pathways. Theranostics 2018; 8:1511-1526. [PMID: 29556338 PMCID: PMC5858164 DOI: 10.7150/thno.21986] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/14/2017] [Indexed: 12/22/2022] Open
Abstract
Rationale: Angiogenesis is critical for embryonic development and microRNAs fine-tune this process, but the underlying mechanisms remain incompletely understood. Methods: Endothelial cell (EC) specific miR302-367 line was used as gain-of-function and anti-miRs as loss-of-function models to investigate the effects of miR302-367 on developmental angiogenesis with embryonic hindbrain vasculature as an in vivo model and fibrin gel beads and tube formation assay as in vitro models. Cell migration was evaluated by Boyden chamber and scratch wound healing assay and cell proliferation by cell count, MTT assay, Ki67 immunostaining and PI cell cycle analysis. RNA high-throughput sequencing identified miR-target genes confirmed by chromatin immunoprecipitation and 3'-UTR luciferase reporter assay, and finally target site blocker determined the pathway contributing significantly to the phenotype observed upon microRNA expression. Results: Elevated EC miR302-367 expression reduced developmental angiogenesis, whereas it was enhanced by inhibition of miR302-367, possibly due to the intrinsic inhibitory effects on EC migration and proliferation. We identified Cdc42 as a direct target gene and elevated EC miR302-367 decreased total and active Cdc42, and further inhibited F-actin formation via the WASP and Klf2/Grb2/Pak1/LIM-kinase/Cofilin pathways. MiR302-367-mediated-Klf2 regulation of Grb2 for fine-tuning Pak1 activation contributing to the inhibited F-actin formation, and then the attenuation of EC migration. Moreover, miR302-367 directly down-regulated EC Ccnd1 and impaired cell proliferation via the Rb/E2F pathway. Conclusion: miR302-367 regulation of endothelial Cdc42 and Ccnd1 signal pathways for EC migration and proliferation advances our understanding of developmental angiogenesis, and meanwhile provides a rationale for future interventions of pathological angiogenesis that shares many common features of physiological angiogenesis.
Collapse
|
25
|
Osmani N, Pontabry J, Comelles J, Fekonja N, Goetz JG, Riveline D, Georges-Labouesse E, Labouesse M. An Arf6- and caveolae-dependent pathway links hemidesmosome remodeling and mechanoresponse. Mol Biol Cell 2017; 29:435-451. [PMID: 29237817 PMCID: PMC6014169 DOI: 10.1091/mbc.e17-06-0356] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/06/2017] [Accepted: 12/08/2017] [Indexed: 01/08/2023] Open
Abstract
Hemidesmosomes are epithelial-specific cell-matrix adhesions stably anchoring the intracellular keratin network to the extracellular matrix and providing mechanical resilience to epithelia. The small GTPase Arf6 and caveolae are essential for their remodeling, notably in response to external mechanical cues. Hemidesmosomes (HDs) are epithelial-specific cell–matrix adhesions that stably anchor the intracellular keratin network to the extracellular matrix. Although their main role is to protect the epithelial sheet from external mechanical strain, how HDs respond to mechanical stress remains poorly understood. Here we identify a pathway essential for HD remodeling and outline its role with respect to α6β4 integrin recycling. We find that α6β4 integrin chains localize to the plasma membrane, caveolae, and ADP-ribosylation factor-6+ (Arf6+) endocytic compartments. Based on fluorescence recovery after photobleaching and endocytosis assays, integrin recycling between both sites requires the small GTPase Arf6 but neither caveolin1 (Cav1) nor Cavin1. Strikingly, when keratinocytes are stretched or hypo-osmotically shocked, α6β4 integrin accumulates at cell edges, whereas Cav1 disappears from it. This process, which is isotropic relative to the orientation of stretch, depends on Arf6, Cav1, and Cavin1. We propose that mechanically induced HD growth involves the isotropic flattening of caveolae (known for their mechanical buffering role) associated with integrin diffusion and turnover.
Collapse
Affiliation(s)
- Naël Osmani
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Inserm U1109, MN3T, 67200 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Julien Pontabry
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Université de Strasbourg, 67000 Strasbourg, France
| | - Jordi Comelles
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Université de Strasbourg, 67000 Strasbourg, France.,Laboratory of Cell Physics, ISIS/IGBMC, CNRS UMR 7006, 67000 Strasbourg, France
| | - Nina Fekonja
- Inserm U1109, MN3T, 67200 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Jacky G Goetz
- Inserm U1109, MN3T, 67200 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Daniel Riveline
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France .,Université de Strasbourg, 67000 Strasbourg, France.,Laboratory of Cell Physics, ISIS/IGBMC, CNRS UMR 7006, 67000 Strasbourg, France
| | - Elisabeth Georges-Labouesse
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Université de Strasbourg, 67000 Strasbourg, France
| | - Michel Labouesse
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France .,Université de Strasbourg, 67000 Strasbourg, France.,UMR7622-CNRS, IBPS, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
26
|
McGonigle TA, Dwyer AR, Greenland EL, Scott NM, Carter KW, Keane KN, Newsholme P, Goodridge HS, Pixley FJ, Hart PH. Reticulon-1 and Reduced Migration toward Chemoattractants by Macrophages Differentiated from the Bone Marrow of Ultraviolet-Irradiated and Ultraviolet-Chimeric Mice. THE JOURNAL OF IMMUNOLOGY 2017; 200:260-270. [DOI: 10.4049/jimmunol.1700760] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 10/18/2017] [Indexed: 01/12/2023]
|
27
|
Abstract
p21-Activated kinase 1 (PAK1) has attracted much attention as a potential therapeutic target due to its central role in many oncogenic signaling pathways, its frequent dysregulation in cancers and neurological disorders, and its tractability as a target for small-molecule inhibition. To date, several PAK1-targeting compounds have been developed as preclinical agents, including one that has been evaluated in a clinical trial. A series of ATP-competitive inhibitors, allosteric inhibitors and peptide inhibitors with distinct biochemical and pharmacokinetic properties represent useful laboratory tools for studies on the role of PAK1 in biology and in disease contexts, and could lead to promising therapeutic agents. Given the central role of PAK1 in vital signaling pathways, future clinical development of PAK1 inhibitors will require careful investigation of their safety and efficacy.
Collapse
|
28
|
Cai YD, Zhang Q, Zhang YH, Chen L, Huang T. Identification of Genes Associated with Breast Cancer Metastasis to Bone on a Protein–Protein Interaction Network with a Shortest Path Algorithm. J Proteome Res 2017; 16:1027-1038. [DOI: 10.1021/acs.jproteome.6b00950] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yu-Dong Cai
- School
of Life Sciences, Shanghai University, Shanghai 200444 People’s Republic of China
| | - Qing Zhang
- School
of Life Sciences, Shanghai University, Shanghai 200444 People’s Republic of China
| | - Yu-Hang Zhang
- Institute
of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
| | - Lei Chen
- College
of Information Engineering, Shanghai Maritime University, Shanghai 201306, People’s Republic of China
| | - Tao Huang
- Institute
of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
| |
Collapse
|
29
|
MicroRNA-494 inhibits breast cancer progression by directly targeting PAK1. Cell Death Dis 2017; 8:e2529. [PMID: 28055013 PMCID: PMC5386359 DOI: 10.1038/cddis.2016.440] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 11/03/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022]
Abstract
MicroRNA (miRNA) is involved in the progression and metastasis of diverse human cancers, including breast cancer, as strong evidence has been found that miRNAs can act as oncogenes or tumor suppressor genes. Here, we show that miR-494 is decreased in human breast cancer specimens and breast cancer cell lines. Ectopic expression of miR-494 in basal-like breast cancer cell lines MDA-MB-231-LUC-D2H3LN and BT-549 inhibits clonogenic ability and metastasis-relevant traits in vitro. Moreover, ectopic expression of miR-494 suppresses neoplasm initiation as well as pulmonary metastasis in vivo. Further studies have identified PAK1, as a direct target gene of miR-494, contributes to the functions of miR-494. Remarkably, the expression of PAK1 is inversely correlated with the level of miR-494 in human breast cancer samples. Furthermore, re-expression of PAK1 partially reverses miR-494-mediated proliferative and clonogenic inhibition as well as migration and invasion suppression in breast cancer cells. Taken together, these findings highlight an important role for miR-494 in the regulation of progression and metastatic potential of breast cancer and suggest a potential application of miR-494 in breast cancer treatment.
Collapse
|
30
|
Su B, Su J, Zeng Y, Liu F, Xia H, Ma YH, Zhou ZG, Zhang S, Yang BM, Wu YH, Zeng X, Ai XH, Ling H, Jiang H, Su Q. Diallyl disulfide suppresses epithelial-mesenchymal transition, invasion and proliferation by downregulation of LIMK1 in gastric cancer. Oncotarget 2016; 7:10498-512. [PMID: 26871290 PMCID: PMC4891135 DOI: 10.18632/oncotarget.7252] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/24/2016] [Indexed: 11/25/2022] Open
Abstract
Diallyl disulfide (DADS) has been shown to have multi-targeted antitumor activities. We have previously discovered that it has a repressive effect on LIM kinase-1 (LIMK1) expression in gastric cancer MGC803 cells. This suggests that DADS may inhibit epithelial-mesenchymal transition (EMT) by downregulating LIMK1, resulting in the inhibition of invasion and growth in gastric cancer. In this study, we reveal that LIMK1 expression is correlated with tumor differentiation, invasion depth, clinical stage, lymph node metastasis, and poor prognosis. DADS downregulated the Rac1-Pak1/Rock1-LIMK1 pathway in MGC803 cells, as shown by decreased p-LIMK1 and p-cofilin1 levels, and suppressed cell migration and invasion. Knockdown and overexpression experiments performed in vitro demonstrated that downregulating LIMK1 with DADS resulted in restrained EMT that was coupled with decreased matrix metalloproteinase-9 (MMP-9) and increased tissue inhibitor of metalloproteinase-3 (TIMP-3) expression. In in vitro and in vivo experiments, the DADS-induced suppression of cell proliferation was enhanced and antagonized by the knockdown and overexpression of LIMK1, respectively. Similar results were observed for DADS-induced changes in the expression of vimentin, CD34, Ki-67, and E-cadherin in xenografted tumors. These results indicate that downregulation of LIMK1 by DADS could explain the inhibition of EMT, invasion and proliferation in gastric cancer cells.
Collapse
Affiliation(s)
- Bo Su
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory for Pharmacoproteomics of Hunan Provincial University, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, 421001 Hunan, China
| | - Jian Su
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China.,Department of Pathology, Second Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China
| | - Ying Zeng
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Fang Liu
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Hong Xia
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Yan-Hua Ma
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Zhi-Gang Zhou
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Shuo Zhang
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Bang-Min Yang
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - You-Hua Wu
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China
| | - Xi Zeng
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Xiao-Hong Ai
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China
| | - Hui Ling
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| | - Hao Jiang
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China
| | - Qi Su
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, 421001 Hunan, China.,Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial University, Cancer Research Institute, University of South China, Hengyang, 421001 Hunan, China
| |
Collapse
|
31
|
Significance of kinase activity in the dynamic invadosome. Eur J Cell Biol 2016; 95:483-492. [PMID: 27465307 DOI: 10.1016/j.ejcb.2016.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 12/19/2022] Open
Abstract
Invadosomes are actin rich protrusive structures that facilitate invasive migration in multiple cell types. Comprised of invadopodia and podosomes, these highly dynamic structures adhere to and degrade the extracellular matrix, and are also thought to play a role in mechanosensing. Many extracellular signals have been implicated in invadosome stimulation, activating complex signalling cascades to drive the formation, activity and turnover of invadosomes. While the structural components of invadosomes have been well studied, the regulation of invadosome dynamics is still poorly understood. Protein kinases are essential to this regulation, affecting all stages of invadosome dynamics and allowing tight spatiotemporal control of their activity. Invadosome organisation and function have been linked to pathophysiological states such as cancer invasion and metastasis; therapeutic targeting of invadosome regulatory components is thus warranted. In this review, we discuss the involvement of kinase signalling in every stage of the invadosome life cycle and evaluate its significance.
Collapse
|
32
|
Zhang W, Huang Y, Gunst SJ. p21-Activated kinase (Pak) regulates airway smooth muscle contraction by regulating paxillin complexes that mediate actin polymerization. J Physiol 2016; 594:4879-900. [PMID: 27038336 DOI: 10.1113/jp272132] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/28/2016] [Indexed: 01/01/2023] Open
Abstract
KEY POINTS In airway smooth muscle, tension development caused by a contractile stimulus requires phosphorylation of the 20 kDa myosin light chain (MLC), which activates crossbridge cycling and the polymerization of a pool of submembraneous actin. The p21-activated kinases (Paks) can regulate the contractility of smooth muscle and non-muscle cells, and there is evidence that this occurs through the regulation of MLC phosphorylation. We show that Pak has no effect on MLC phosphorylation during the contraction of airway smooth muscle, and that it regulates contraction by mediating actin polymerization. We find that Pak phosphorylates the adhesion junction protein, paxillin, on Ser273, which promotes the formation of a signalling complex that activates the small GTPase, cdc42, and the actin polymerization catalyst, neuronal Wiskott-Aldrich syndrome protein (N-WASP). These studies demonstrate a novel role for Pak in regulating the contractility of smooth muscle by regulating actin polymerization. ABSTRACT The p21-activated kinases (Pak) can regulate contractility in smooth muscle and other cell and tissue types, but the mechanisms by which Paks regulate cell contractility are unclear. In airway smooth muscle, stimulus-induced contraction requires phosphorylation of the 20 kDa light chain of myosin, which activates crossbridge cycling, as well as the polymerization of a small pool of actin. The role of Pak in airway smooth muscle contraction was evaluated by inhibiting acetylcholine (ACh)-induced Pak activation through the expression of a kinase inactive mutant, Pak1 K299R, or by treating tissues with the Pak inhibitor, IPA3. Pak inhibition suppressed actin polymerization and contraction in response to ACh, but it did not affect myosin light chain phosphorylation. Pak activation induced paxillin phosphorylation on Ser273; the paxillin mutant, paxillin S273A, inhibited paxillin Ser273 phosphorylation and inhibited actin polymerization and contraction. Immunoprecipitation analysis of tissue extracts and proximity ligation assays in dissociated cells showed that Pak activation and paxillin Ser273 phosphorylation triggered the formation of an adhesion junction signalling complex with paxillin that included G-protein-coupled receptor kinase-interacting protein (GIT1) and the cdc42 guanine exchange factor, βPIX (Pak interactive exchange factor). Assembly of the Pak-GIT1-βPIX-paxillin complex was necessary for cdc42 and neuronal Wiskott-Aldrich syndrome protein (N-WASP) activation, actin polymerization and contraction in response to ACh. RhoA activation was also required for the recruitment of Pak to adhesion junctions, Pak activation, paxillin Ser273 phosphorylation and paxillin complex assembly. These studies demonstrate a novel role for Pak in the regulation of N-WASP activation, actin dynamics and cell contractility.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Cellular and Integrative Physiology, Indiana University School Medicine, Indianapolis, IN, 46202-5120, USA
| | - Youliang Huang
- Department of Cellular and Integrative Physiology, Indiana University School Medicine, Indianapolis, IN, 46202-5120, USA
| | - Susan J Gunst
- Department of Cellular and Integrative Physiology, Indiana University School Medicine, Indianapolis, IN, 46202-5120, USA
| |
Collapse
|
33
|
P21-Activated Kinase Inhibitors FRAX486 and IPA3: Inhibition of Prostate Stromal Cell Growth and Effects on Smooth Muscle Contraction in the Human Prostate. PLoS One 2016; 11:e0153312. [PMID: 27071060 PMCID: PMC4829229 DOI: 10.1371/journal.pone.0153312] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 03/28/2016] [Indexed: 12/25/2022] Open
Abstract
Prostate smooth muscle tone and hyperplastic growth are involved in the pathophysiology and treatment of male lower urinary tract symptoms (LUTS). Available drugs are characterized by limited efficacy. Patients' adherence is particularly low to combination therapies of 5α-reductase inhibitors and α1-adrenoceptor antagonists, which are supposed to target contraction and growth simultaneously. Consequently, molecular etiology of benign prostatic hyperplasia (BPH) and new compounds interfering with smooth muscle contraction or growth in the prostate are of high interest. Here, we studied effects of p21-activated kinase (PAK) inhibitors (FRAX486, IPA3) in hyperplastic human prostate tissues, and in stromal cells (WPMY-1). In hyperplastic prostate tissues, PAK1, -2, -4, and -6 may be constitutively expressed in catecholaminergic neurons, while PAK1 was detected in smooth muscle and WPMY-1 cells. Neurogenic contractions of prostate strips by electric field stimulation were significantly inhibited by high concentrations of FRAX486 (30 μM) or IPA3 (300 μM), while noradrenaline- and phenylephrine-induced contractions were not affected. FRAX486 (30 μM) inhibited endothelin-1- and -2-induced contractions. In WPMY-1 cells, FRAX486 or IPA3 (24 h) induced concentration-dependent (1-10 μM) degeneration of actin filaments. This was paralleled by attenuation of proliferation rate, being observed from 1 to 10 μM FRAX486 or IPA3. Cytotoxicity of FRAX486 and IPA3 in WPMY-1 cells was time- and concentration-dependent. Stimulation of WPMY-1 cells with endothelin-1 or dihydrotestosterone, but not noradrenaline induced PAK phosphorylation, indicating PAK activation by endothelin-1. Thus, PAK inhibitors may inhibit neurogenic and endothelin-induced smooth muscle contractions in the hyperplastic human prostate, and growth of stromal cells. Targeting prostate smooth muscle contraction and stromal growth at once by a single compound is principally possible, at least under experimental conditions.
Collapse
|
34
|
Osma-Garcia IC, Punzón C, Fresno M, Díaz-Muñoz MD. Dose-dependent effects of prostaglandin E2 in macrophage adhesion and migration. Eur J Immunol 2015; 46:677-88. [PMID: 26631603 DOI: 10.1002/eji.201545629] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 10/08/2015] [Accepted: 11/26/2015] [Indexed: 12/15/2022]
Abstract
Macrophage migration to the focus of infection is a hallmark of the innate immune response. Macrophage spreading, adhesion, and migration through the extracellular matrix require dynamic remodeling of the actin cytoskeleton associated to integrin clustering in podosomes and focal adhesions. Here, we show that prostaglandin E2 (PGE2 ), the main prostaglandin produced by macrophages during inflammation, promote the distinctive dose-dependent formation of podosomes or focal adhesions in macrophages. Low concentrations of PGE2 increased p110γ PI3K expression, phosphorylation of actin-related protein 2, and formation of podosomes, which enhanced macrophage migration in response to chemokines. However, high doses of PGE2 increased phosphorylation of paxillin and focal adhesion kinase, the expression of serine/threonine protein kinase 1, and promoted focal adhesion formation and macrophage adhesion, reducing macrophage chemotaxis. In summary, we describe the dual role of PGE2 as a promoter of macrophage chemotaxis and adhesion, proposing a new model of macrophage migration to the inflammatory focus in the presence of a gradient of PGE2 .
Collapse
Affiliation(s)
- Inés C Osma-Garcia
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Punzón
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Fresno
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel D Díaz-Muñoz
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
35
|
Pinto V, Mohammadi H, Lee W, Cheung A, McCulloch C. PAK1 is involved in sensing the orientation of collagen stiffness gradients in mouse fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2526-38. [DOI: 10.1016/j.bbamcr.2015.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 05/01/2015] [Accepted: 05/19/2015] [Indexed: 01/13/2023]
|
36
|
Kubo Y, Baba K, Toriyama M, Minegishi T, Sugiura T, Kozawa S, Ikeda K, Inagaki N. Shootin1-cortactin interaction mediates signal-force transduction for axon outgrowth. J Cell Biol 2015; 210:663-76. [PMID: 26261183 PMCID: PMC4539990 DOI: 10.1083/jcb.201505011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/26/2015] [Indexed: 11/22/2022] Open
Abstract
The shootin1–cortactin interaction participates in netrin-1–induced F-actin–adhesion coupling and in the promotion of traction forces for axon outgrowth. Motile cells transduce environmental chemical signals into mechanical forces to achieve properly controlled migration. This signal–force transduction is thought to require regulated mechanical coupling between actin filaments (F-actins), which undergo retrograde flow at the cellular leading edge, and cell adhesions via linker “clutch” molecules. However, the molecular machinery mediating this regulatory coupling remains unclear. Here we show that the F-actin binding molecule cortactin directly interacts with a clutch molecule, shootin1, in axonal growth cones, thereby mediating the linkage between F-actin retrograde flow and cell adhesions through L1-CAM. Shootin1–cortactin interaction was enhanced by shootin1 phosphorylation by Pak1, which is activated by the axonal chemoattractant netrin-1. We provide evidence that shootin1–cortactin interaction participates in netrin-1–induced F-actin adhesion coupling and in the promotion of traction forces for axon outgrowth. Under cell signaling, this regulatory F-actin adhesion coupling in growth cones cooperates with actin polymerization for efficient cellular motility.
Collapse
Affiliation(s)
- Yusuke Kubo
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Kentarou Baba
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Michinori Toriyama
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takunori Minegishi
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Tadao Sugiura
- Laboratory of Biomedical Imaging, Graduate School of Information Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Satoshi Kozawa
- Mathematical Informatics, Graduate School of Information Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Kazushi Ikeda
- Mathematical Informatics, Graduate School of Information Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
37
|
Regulation of sarcoma cell migration, invasion and invadopodia formation by AFAP1L1 through a phosphotyrosine-dependent pathway. Oncogene 2015. [DOI: 10.1038/onc.2015.272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
38
|
Abstract
INTRODUCTION Rho GTPases are master regulators of actomyosin structure and dynamics and play pivotal roles in a variety of cellular processes including cell morphology, gene transcription, cell cycle progression, and cell adhesion. Because aberrant Rho GTPase signaling activities are widely associated with human cancer, key components of Rho GTPase signaling pathways have attracted increasing interest as potential therapeutic targets. Similar to Ras, Rho GTPases themselves were, until recently, deemed "undruggable" because of structure-function considerations. Several approaches to interfere with Rho GTPase signaling have been explored and show promise as new ways for tackling cancer cells. AREAS COVERED This review focuses on the recent progress in targeting the signaling activities of three prototypical Rho GTPases, that is, RhoA, Rac1, and Cdc42. The authors describe the involvement of these Rho GTPases, their key regulators and effectors in cancer. Furthermore, the authors discuss the current approaches for rationally targeting aberrant Rho GTPases along their signaling cascades, upstream and downstream of Rho GTPases, and posttranslational modifications at a molecular level. EXPERT OPINION To date, while no clinically effective drugs targeting Rho GTPase signaling for cancer treatment are available, tool compounds and lead drugs that pharmacologically inhibit Rho GTPase pathways have shown promise. Small-molecule inhibitors targeting Rho GTPase signaling may add new treatment options for future precision cancer therapy, particularly in combination with other anti-cancer agents.
Collapse
Affiliation(s)
- Yuan Lin
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio 45229, USA
| |
Collapse
|
39
|
Gan J, Zhang Y, Ke X, Tan C, Ren H, Dong H, Jiang J, Chen S, Zhuang Y, Zhang H. Dysregulation of PAK1 Is Associated with DNA Damage and Is of Prognostic Importance in Primary Esophageal Small Cell Carcinoma. Int J Mol Sci 2015; 16:12035-50. [PMID: 26023713 PMCID: PMC4490427 DOI: 10.3390/ijms160612035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/12/2015] [Indexed: 02/05/2023] Open
Abstract
Primary esophageal small cell carcinoma (PESCC) is a rare, but fatal subtype of esophageal carcinoma. No effective therapeutic regimen for it. P21-activated kinase 1 (PAK1) is known to function as an integrator and an indispensable node of major growth factor signaling and the molecular therapy targeting PAK1 has been clinical in pipeline. We thus set to examine the expression and clinical impact of PAK1 in PESCC. The expression of PAK1 was detected in a semi-quantitative manner by performing immunohistochemistry. PAK1 was overexpressed in 22 of 34 PESCC tumors, but in only 2 of 18 adjacent non-cancerous tissues. Overexpression of PAK1 was significantly associated with tumor location (p = 0.011), lymph node metastasis (p = 0.026) and patient survival (p = 0.032). We also investigated the association of PAK1 with DNA damage, a driven cause for malignancy progression. γH2AX, a DNA damage marker, was detectable in 18 of 24 (75.0%) cases, and PAK1 expression was associated with γH2AX (p = 0.027). Together, PAK1 is important in metastasis and progression of PESCC. The contribution of PAK1 to clinical outcomes may be involved in its regulating DNA damage pathway. Further studies are worth determining the potentials of PAK1 as prognostic indicator and therapeutic target for PESCC.
Collapse
Affiliation(s)
- Jinfeng Gan
- Cancer Research Centre, Shantou University Medical College, Shantou 515063, China.
| | - Yuling Zhang
- Department of Information, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515031, China.
| | - Xiurong Ke
- Cancer Research Centre, Shantou University Medical College, Shantou 515063, China.
- Department of Biotherapy, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515031, China.
| | - Chong Tan
- Department of General Surgery, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China.
| | - Hongzheng Ren
- Department of Pathology, Central Hospital of Kaifeng, Kaifeng 475000, China.
| | - Hongmei Dong
- Cancer Research Centre, Shantou University Medical College, Shantou 515063, China.
| | - Jiali Jiang
- Cancer Research Centre, Shantou University Medical College, Shantou 515063, China.
- Department of Biotherapy, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515031, China.
| | - Shaobin Chen
- Thoracic Surgery, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515031, China.
| | - Yixuan Zhuang
- Tumor Tissue Bank, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515031, China.
| | - Hao Zhang
- Cancer Research Centre, Shantou University Medical College, Shantou 515063, China.
- Department of Biotherapy, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515031, China.
- Tumor Tissue Bank, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515031, China.
| |
Collapse
|
40
|
Howell M, Brickner H, Delorme-Walker VD, Choi J, Saffin JM, Miller D, Panopoulos A, DerMardirossian C, Fotedar A, Margolis RL, Fotedar R. WISp39 binds phosphorylated Coronin 1B to regulate Arp2/3 localization and Cofilin-dependent motility. ACTA ACUST UNITED AC 2015; 208:961-74. [PMID: 25800056 PMCID: PMC4384738 DOI: 10.1083/jcb.201410095] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously identified Waf1 Cip1 stabilizing protein 39 (WISp39) as a binding partner for heat shock protein 90 (Hsp90). We now report that WISp39 has an essential function in the control of directed cell migration, which requires WISp39 interaction with Hsp90. WISp39 knockdown (KD) resulted in the loss of directional motility of mammalian cells and profound changes in cell morphology, including the loss of a single leading edge. WISp39 binds Coronin 1B, known to regulate the Arp2/3 complex and Cofilin at the leading edge. WISp39 preferentially interacts with phosphorylated Coronin 1B, allowing it to complex with Slingshot phosphatase (SSH) to dephosphorylate and activate Cofilin. WISp39 also regulates Arp2/3 complex localization at the leading edge. WISp39 KD-induced morphological changes could be rescued by overexpression of Coronin 1B together with a constitutively active Cofilin mutant. We conclude that WISp39 associates with Hsp90, Coronin 1B, and SSH to regulate Cofilin activation and Arp2/3 complex localization at the leading edge.
Collapse
Affiliation(s)
- Michael Howell
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Howard Brickner
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | | | - Justin Choi
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Jean-Michel Saffin
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037
| | - Daniel Miller
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | | | | | - Arun Fotedar
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | | | - Rati Fotedar
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| |
Collapse
|
41
|
Markwell SM, Weed SA. Tumor and stromal-based contributions to head and neck squamous cell carcinoma invasion. Cancers (Basel) 2015; 7:382-406. [PMID: 25734659 PMCID: PMC4381264 DOI: 10.3390/cancers7010382] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/10/2015] [Accepted: 02/15/2015] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is typically diagnosed at advanced stages with evident loco-regional and/or distal metastases. The prevalence of metastatic lesions directly correlates with poor patient outcome, resulting in high patient mortality rates following metastatic development. The progression to metastatic disease requires changes not only in the carcinoma cells, but also in the surrounding stromal cells and tumor microenvironment. Within the microenvironment, acellular contributions from the surrounding extracellular matrix, along with contributions from various infiltrating immune cells, tumor associated fibroblasts, and endothelial cells facilitate the spread of tumor cells from the primary site to the rest of the body. Thus far, most attempts to limit metastatic spread through therapeutic intervention have failed to show patient benefit in clinic trails. The goal of this review is highlight the complexity of invasion-promoting interactions in the HNSCC tumor microenvironment, focusing on contributions from tumor and stromal cells in order to assist future therapeutic development and patient treatment.
Collapse
Affiliation(s)
- Steven M Markwell
- Department of Neurobiology and Anatomy, Program in Cancer Cell Biology, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA.
| | - Scott A Weed
- Department of Neurobiology and Anatomy, Program in Cancer Cell Biology, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
42
|
Hammer A, Diakonova M. Tyrosyl phosphorylated serine-threonine kinase PAK1 is a novel regulator of prolactin-dependent breast cancer cell motility and invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 846:97-137. [PMID: 25472536 DOI: 10.1007/978-3-319-12114-7_5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite efforts to discover the cellular pathways regulating breast cancer metastasis, little is known as to how prolactin (PRL) cooperates with extracellular environment and cytoskeletal proteins to regulate breast cancer cell motility and invasion. We implicated serine-threonine kinase p21-activated kinase 1 (PAK1) as a novel target for PRL-activated Janus-kinase 2 (JAK2). JAK2-dependent PAK1 tyrosyl phosphorylation plays a critical role in regulation of both PAK1 kinase activity and scaffolding properties of PAK1. Tyrosyl phosphorylated PAK1 facilitates PRL-dependent motility via at least two mechanisms: formation of paxillin/GIT1/βPIX/pTyr-PAK1 complexes resulting in increased adhesion turnover and phosphorylation of actin-binding protein filamin A. Increased adhesion turnover is the basis for cell migration and phosphorylated filamin A stimulates the kinase activity of PAK1 and increases actin-regulating activity to facilitate cell motility. Tyrosyl phosphorylated PAK1 also stimulates invasion of breast cancer cells in response to PRL and three-dimensional (3D) collagen IV via transcription and secretion of MMP-1 and MMP-3 in a MAPK-dependent manner. These data illustrate the complex interaction between PRL and the cell microenvironment in breast cancer cells and suggest a pivotal role for PRL/PAK1 signaling in breast cancer metastasis.
Collapse
Affiliation(s)
- Alan Hammer
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | | |
Collapse
|
43
|
Hammer A, Oladimeji P, De Las Casas LE, Diakonova M. Phosphorylation of tyrosine 285 of PAK1 facilitates βPIX/GIT1 binding and adhesion turnover. FASEB J 2014; 29:943-59. [PMID: 25466889 DOI: 10.1096/fj.14-259366] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The p21-activated serine-threonine kinase (PAK1) regulates cell motility and adhesion. We have previously shown that the prolactin (PRL)-activated tyrosine kinase JAK2 phosphorylates PAK1 in vivo and in vitro and identified tyrosines 153, 201, and 285 in PAK1 as sites of JAK2 tyrosyl phosphorylation. Here, we further investigate the role of the tyrosyl phosphorylated PAK1 (pTyr-PAK1) in regulation of cell adhesion. We use human breast cancer T47D cell lines that stably overexpress PAK1 wild type or PAK1 Y3F mutant in which these 3 JAK2 phosphorylation sites were mutated to phenylalanine. We demonstrate that PRL/JAK2-dependent phosphorylation of these tyrosines promotes a motile phenotype in the cells upon adhesion, participates in regulation of cell adhesion on collagen IV, and is required for maximal PAK1 kinase activity. Down-regulation of PAK1 abolishes the effect of PAK1 on cell adhesion. We show that the tyrosyl phosphorylation of PAK1 promotes PAK1 binding to β-PAK1-interacting guanine-nucleotide exchange factor (βPIX) and G protein-coupled receptor kinase-interacting target 1 (GIT1), phosphorylation of paxillin on Ser273, and formation and distribution of adhesion complexes. Using phosphospecific antibodies (Abs) directed to single phosphorylated tyrosines on PAK1, we identified Tyr285 as a site of PRL-dependent phosphorylation of PAK1 by JAK2. Furthermore, using PAK1 Y285F mutant, we provide evidence for a role of pTyr285 in cell adhesion, enhanced βPIX/GIT1 binding, and adhesion turnover. Our immunohistochemistry analysis demonstrates that pTyr285- PAK1 may modulate PAK1 signaling during tumor progression.
Collapse
Affiliation(s)
- Alan Hammer
- Departments of *Biological Sciences and Pathology, University of Toledo, Toledo, Ohio, USA
| | - Peter Oladimeji
- Departments of *Biological Sciences and Pathology, University of Toledo, Toledo, Ohio, USA
| | - Luis E De Las Casas
- Departments of *Biological Sciences and Pathology, University of Toledo, Toledo, Ohio, USA
| | - Maria Diakonova
- Departments of *Biological Sciences and Pathology, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
44
|
Xu X, Wang W, Kratz K, Fang L, Li Z, Kurtz A, Ma N, Lendlein A. Controlling major cellular processes of human mesenchymal stem cells using microwell structures. Adv Healthc Mater 2014; 3:1991-2003. [PMID: 25313500 DOI: 10.1002/adhm.201400415] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/02/2014] [Indexed: 01/17/2023]
Abstract
Directing stem cells towards a desired location and function by utilizing the structural cues of biomaterials is a promising approach for inducing effective tissue regeneration. Here, the cellular response of human adipose-derived mesenchymal stem cells (hADSCs) to structural signals from microstructured substrates comprising arrays of square-shaped or round-shaped microwells is explored as a transitional model between 2D and 3D systems. Microwells with a side length/diameter of 50 μm show advantages over 10 μm and 25 μm microwells for accommodating hADSCs within single microwells rather than in the inter-microwell area. The cell morphologies are three-dimensionally modulated by the microwell structure due to differences in focal adhesion and consequent alterations of the cytoskeleton. In contrast to the substrate with 50 μm round-shaped microwells, the substrate with 50 μm square-shaped microwells promotes the proliferation and osteogenic differentiation potential of hADSCs but reduces the cell migration velocity and distance. Such microwell shape-dependent modulatory effects are highly associated with Rho/ROCK signaling. Following ROCK inhibition, the differences in migration, proliferation, and osteogenesis between cells on different substrates are diminished. These results highlight the possibility to control stem cell functions through the use of structured microwells combined with the manipulation of Rho/ROCK signaling.
Collapse
Affiliation(s)
- Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| | - Liang Fang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
| | - Zhengdong Li
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
| | - Andreas Kurtz
- Berlin-Brandenburg Center for Regenerative Therapies; Charité - University Medicine Berlin; Augustenburger Platz 1 13353 Berlin Germany
- College of Veterinary Medicine and Research Institute for Veterinary Science; Seoul National University; Gwangk-ro 1 Gwanak-gu Seoul 151-747 Republic of Korea
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| |
Collapse
|
45
|
Komaravolu RK, Adam C, Moonen JRA, Harmsen MC, Goebeler M, Schmidt M. Erk5 inhibits endothelial migration via KLF2-dependent down-regulation of PAK1. Cardiovasc Res 2014; 105:86-95. [DOI: 10.1093/cvr/cvu236] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
46
|
Dammann K, Khare V, Gasche C. Republished: tracing PAKs from GI inflammation to cancer. Postgrad Med J 2014; 90:657-68. [PMID: 25335797 PMCID: PMC4222351 DOI: 10.1136/postgradmedj-2014-306768rep] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 04/07/2014] [Accepted: 04/10/2014] [Indexed: 12/20/2022]
Abstract
P-21 activated kinases (PAKs) are effectors of Rac1/Cdc42 which coordinate signals from the cell membrane to the nucleus. Activation of PAKs drive important signalling pathways including mitogen activated protein kinase, phospoinositide 3-kinase (PI3K/AKT), NF-κB and Wnt/β-catenin. Intestinal PAK1 expression increases with inflammation and malignant transformation, although the biological relevance of PAKs in the development and progression of GI disease is only incompletely understood. This review highlights the importance of altered PAK activation within GI inflammation, emphasises its effect on oncogenic signalling and discusses PAKs as therapeutic targets of chemoprevention.
Collapse
Affiliation(s)
- Kyle Dammann
- Department of Medicine III, Division of Gastroenterology and Hepatology and Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Medical University of Vienna, Vienna, Austria
| | - Vineeta Khare
- Department of Medicine III, Division of Gastroenterology and Hepatology and Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Medical University of Vienna, Vienna, Austria
| | - Christoph Gasche
- Department of Medicine III, Division of Gastroenterology and Hepatology and Christian Doppler Laboratory for Molecular Cancer Chemoprevention, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Field J, Manser E. The PAKs come of age: Celebrating 18 years of discovery. CELLULAR LOGISTICS 2014; 2:54-58. [PMID: 23125949 PMCID: PMC3485743 DOI: 10.4161/cl.22084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Protein kinases are versatile signaling molecules that are involved in the regulation most physiological responses. The p21-activated kinases (PAKs) can be activated directly by the small GTPases Rac and Cdc42 and are among the best characterized downstream effectors of these Rho proteins. The structure, substrate specificity and functional role of PAKS are evolutionarily conserved from protozoa to mammals. Vertebrate PAKs are particularly important for cytoskeletal remodeling and focal adhesion assembly, thereby contributing to dynamic processes such as cell migration and synaptic plasticity. This issue of Cellular Logistics focuses on the PAK family of kinases, with ten reviews written by researchers currently working in the field. Here in this introductory overview we highlight some of the most interesting recent discoveries regarding PAK biochemistry and biology. The reviews in this issue cover a range of topics including the atomic structures of PAK1 and PAK4, their role in animals as assessed by knockout studies, and how PAKs are likely to contribute to cancer and neurodegenerative diseases. The promise remains that PAK inhibitors will emerge that validate current pre-clinical studies suggesting that blocking PAK activity will positively contribute to human health.
Collapse
Affiliation(s)
- Jeffrey Field
- Department of Pharmacology; Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | | |
Collapse
|
48
|
Parrini MC. Untangling the complexity of PAK1 dynamics: The future challenge. CELLULAR LOGISTICS 2014; 2:78-83. [PMID: 23125950 PMCID: PMC3485744 DOI: 10.4161/cl.19817] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PAK1 kinase is a crucial regulator of a variety of cellular processes, such as motility, cell division, gene transcription and apoptosis. Its deregulation is involved in several pathologies, including cancer, viral infection and neurodegenerative diseases. Due to this strong implication in human health, the complex network of signaling pathways centered on PAK1 is a subject of intensive investigations. This review summarizes the present knowledge on the multiple PAK1 intracellular localizations and on its shuttling between different compartments. The dynamics of PAK1 localization and activation are finely tuned by the cell and it is this tight control that underlies the capacity of PAK1 to participate in the regulation of many fundamental cell functions. Recently, PAK1 biosensors have been developed to visualize PAK1 activation in live cells. These new imaging tools should be of great help to better understand PAK1 biology and to conceive strategies for efficient and specific PAK1 inhibitors.
Collapse
Affiliation(s)
- Maria Carla Parrini
- Institut Curie; Centre de Recherche; Paris, France; Inserm U830; Paris, France
| |
Collapse
|
49
|
Ribeiro SA, D'Ambrosio MV, Vale RD. Induction of focal adhesions and motility in Drosophila S2 cells. Mol Biol Cell 2014; 25:3861-9. [PMID: 25273555 PMCID: PMC4244196 DOI: 10.1091/mbc.e14-04-0863] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this study, normally immotile S2 cells are engineered to induce the formation of focal adhesions and cell motility by the transfection of a single gene encoding an integrin subunit. It is demonstrated that the focal adhesions recruit expected components and exhibit mechanosensitive behavior on integrin-ligand substrates of different stiffnesses. Focal adhesions are dynamic structures that interact with the extracellular matrix on the cell exterior and actin filaments on the cell interior, enabling cells to adhere and crawl along surfaces. We describe a system for inducing the formation of focal adhesions in normally non–ECM-adherent, nonmotile Drosophila S2 cells. These focal adhesions contain the expected molecular markers such as talin, vinculin, and p130Cas, and they require talin for their formation. The S2 cells with induced focal adhesions also display a nonpolarized form of motility on vitronectin-coated substrates. Consistent with findings in mammalian cells, the degree of motility can be tuned by changing the stiffness of the substrate and was increased after the depletion of PAK3, a p21-activated kinase. A subset of nonmotile, nonpolarized cells also exhibited focal adhesions that rapidly assembled and disassembled around the cell perimeter. Such cooperative and dynamic fluctuations of focal adhesions were decreased by RNA interference (RNAi) depletion of myosin II and focal adhesion kinase, suggesting that this behavior requires force and focal adhesion maturation. These results demonstrate that S2 cells, a cell line that is well studied for cytoskeletal dynamics and readily amenable to protein manipulation by RNAi, can be used to study the assembly and dynamics of focal adhesions and mechanosensitive cell motility.
Collapse
Affiliation(s)
- Susana A Ribeiro
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Michael V D'Ambrosio
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Ronald D Vale
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
50
|
Signaling of the p21-activated kinase (PAK1) coordinates insulin-stimulated actin remodeling and glucose uptake in skeletal muscle cells. Biochem Pharmacol 2014; 92:380-8. [PMID: 25199455 DOI: 10.1016/j.bcp.2014.08.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 11/22/2022]
Abstract
Skeletal muscle accounts for ∼ 80% of postprandial glucose clearance, and skeletal muscle glucose clearance is crucial for maintaining insulin sensitivity and euglycemia. Insulin-stimulated glucose clearance/uptake entails recruitment of glucose transporter 4 (GLUT4) to the plasma membrane (PM) in a process that requires cortical F-actin remodeling; this process is dysregulated in Type 2 Diabetes. Recent studies have implicated PAK1 as a required element in GLUT4 recruitment in mouse skeletal muscle in vivo, although its underlying mechanism of action and requirement in glucose uptake remains undetermined. Toward this, we have employed the PAK1 inhibitor, IPA3, in studies using L6-GLUT4-myc muscle cells. IPA3 fully ablated insulin-stimulated GLUT4 translocation to the PM, corroborating the observation of ablated insulin-stimulated GLUT4 accumulation in the PM of skeletal muscle from PAK1(-/-) knockout mice. IPA3-treatment also abolished insulin-stimulated glucose uptake into skeletal myotubes. Mechanistically, live-cell imaging of myoblasts expressing the F-actin biosensor LifeAct-GFP treated with IPA3 showed blunting of the normal insulin-induced cortical actin remodeling. This blunting was underpinned by a loss of normal insulin-stimulated cofilin dephosphorylation in IPA3-treated myoblasts. These findings expand upon the existing model of actin remodeling in glucose uptake, by placing insulin-stimulated PAK1 signaling as a required upstream step to facilitate actin remodeling and subsequent cofilin dephosphorylation. Active, dephosphorylated cofilin then provides the G-actin substrate for continued F-actin remodeling to facilitate GLUT4 vesicle translocation for glucose uptake into the skeletal muscle cell.
Collapse
|