1
|
Spagnol G, Trease A, Zheng L, Sobota S, Schmidt M, Cheku S, Sorgen PL. Cx45 regulation by kinases and impact of expression in heart failure. J Mol Cell Cardiol 2025; 203:91-105. [PMID: 40280467 DOI: 10.1016/j.yjmcc.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Phosphorylation plays a crucial role in connexin regulation by modulating gap junction intercellular communication (GJIC), localization, stability, and interactions with signaling proteins. Few kinases are known to phosphorylate Cx45, and their target residues remain unknown. A phosphorylation screen identified several Cx45-targeting kinases activated in heart disease, among which c-Src was found by mass spectroscopy to phosphorylate residues Y324 and Y356. Unlike Cx43, c-Src phosphorylation of Cx45 did not impair GJIC, alter junctional localization, or affect interactions with cytoskeletal proteins β-tubulin, Drebrin, and ZO-1. In LA-25 cells where Cx43 is internalized after temperature sensitive activation of v-Src, expression of Cx45 unexpectedly maintained Cx43 at the plasma membrane. Phospho-specific antibodies helped identify that while Cx43 had a tyrosine phosphorylation pattern favoring turnover, the serine phosphorylation pattern was conducive for GJIC. Furthermore, in a rat model of heart failure, Cx45 was expressed in the ventricle and co-localized with Cx43, leading to altered dye coupling indicative of a shift toward Cx45-like channel permeability. Altogether, our data suggests that in heart failure, c-Src activation on its own would not have an adverse effect on Cx45 function and that aberrant Cx45 expression helps Cx43 transport to and maintain at the intercalated disc. Yet the dominant effect of Cx45 in heteromeric channels could ultimately make Cx45 a key driver of cardiac dysfunction. Finally, the observation that Cx45-mediated coupling remains functional even in the same pathological environment where Cx43-mediated communication is inhibited suggests that kinase regulation of connexins is isoform-specific and not universally predictable.
Collapse
Affiliation(s)
- Gaelle Spagnol
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Andrew Trease
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Li Zheng
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Stephen Sobota
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Marissa Schmidt
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sunayn Cheku
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Paul L Sorgen
- From the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
2
|
Gusev E, Sarapultsev A. Interplay of G-proteins and Serotonin in the Neuroimmunoinflammatory Model of Chronic Stress and Depression: A Narrative Review. Curr Pharm Des 2024; 30:180-214. [PMID: 38151838 DOI: 10.2174/0113816128285578231218102020] [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: 10/04/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023]
Abstract
INTRODUCTION This narrative review addresses the clinical challenges in stress-related disorders such as depression, focusing on the interplay between neuron-specific and pro-inflammatory mechanisms at the cellular, cerebral, and systemic levels. OBJECTIVE We aim to elucidate the molecular mechanisms linking chronic psychological stress with low-grade neuroinflammation in key brain regions, particularly focusing on the roles of G proteins and serotonin (5-HT) receptors. METHODS This comprehensive review of the literature employs systematic, narrative, and scoping review methodologies, combined with systemic approaches to general pathology. It synthesizes current research on shared signaling pathways involved in stress responses and neuroinflammation, including calcium-dependent mechanisms, mitogen-activated protein kinases, and key transcription factors like NF-κB and p53. The review also focuses on the role of G protein-coupled neurotransmitter receptors (GPCRs) in immune and pro-inflammatory responses, with a detailed analysis of how 13 of 14 types of human 5-HT receptors contribute to depression and neuroinflammation. RESULTS The review reveals a complex interaction between neurotransmitter signals and immunoinflammatory responses in stress-related pathologies. It highlights the role of GPCRs and canonical inflammatory mediators in influencing both pathological and physiological processes in nervous tissue. CONCLUSION The proposed Neuroimmunoinflammatory Stress Model (NIIS Model) suggests that proinflammatory signaling pathways, mediated by metabotropic and ionotropic neurotransmitter receptors, are crucial for maintaining neuronal homeostasis. Chronic mental stress can disrupt this balance, leading to increased pro-inflammatory states in the brain and contributing to neuropsychiatric and psychosomatic disorders, including depression. This model integrates traditional theories on depression pathogenesis, offering a comprehensive understanding of the multifaceted nature of the condition.
Collapse
Affiliation(s)
- Evgenii Gusev
- Laboratory of Inflammation Immunology, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, Ekaterinburg 620049, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, Chelyabinsk 454080, Russia
| | - Alexey Sarapultsev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, Chelyabinsk 454080, Russia
- Laboratory of Immunopathophysiology, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, Ekaterinburg 620049, Russia
| |
Collapse
|
3
|
Matsumoto M, Modliszewski JL, Shinozaki K, Maezawa R, Perez VM, Ishikawa Y, Suzuki R, McKnight KL, Masaki T, Hirai-Yuki A, Kohara M, Lemon SM, Selitsky SR, Yamane D. CSNK2B modulates IRF1 binding to functional DNA elements and promotes basal and agonist-induced antiviral signaling. Nucleic Acids Res 2023; 51:4451-4466. [PMID: 37094077 DOI: 10.1093/nar/gkad298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/21/2023] [Accepted: 04/14/2023] [Indexed: 04/26/2023] Open
Abstract
Interferon regulatory factor 1 (IRF1) is a critical component of cell-intrinsic innate immunity that regulates both constitutive and induced antiviral defenses. Due to its short half-life, IRF1 function is generally considered to be regulated by its synthesis. However, how IRF1 activity is controlled post-translationally has remained poorly characterized. Here, we employed a proteomics approach to identify proteins interacting with IRF1, and found that CSNK2B, a regulatory subunit of casein kinase 2, interacts directly with IRF1 and constitutively modulates its transcriptional activity. Genome-wide CUT&RUN analysis of IRF1 binding loci revealed that CSNK2B acts generally to enhance the binding of IRF1 to chromatin, thereby enhancing transcription of key antiviral genes, such as PLAAT4 (also known as RARRES3/RIG1/TIG3). On the other hand, depleting CSNK2B triggered abnormal accumulation of IRF1 at AFAP1 loci, thereby down-regulating transcription of AFAP1, revealing contrary effects of CSNK2B on IRF1 binding at different loci. AFAP1 encodes an actin crosslinking factor that mediates Src activation. Importantly, CSNK2B was also found to mediate phosphorylation-dependent activation of AFAP1-Src signaling and exert suppressive effects against flaviviruses, including dengue virus. These findings reveal a previously unappreciated mode of IRF1 regulation and identify important effector genes mediating multiple cellular functions governed by CSNK2B and IRF1.
Collapse
Affiliation(s)
- Moe Matsumoto
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | | | - Kotomi Shinozaki
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Reona Maezawa
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | | | - Yuki Ishikawa
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Kevin L McKnight
- Lineberger Comprehensive Cancer Center, and Departments of Medicine and Microbiology & Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7292, USA
| | - Takahiro Masaki
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Asuka Hirai-Yuki
- Management Department of Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Michinori Kohara
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Stanley M Lemon
- Lineberger Comprehensive Cancer Center, and Departments of Medicine and Microbiology & Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7292, USA
| | | | - Daisuke Yamane
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
4
|
Peláez R, Pariente A, Pérez-Sala Á, Larrayoz IM. Integrins: Moonlighting Proteins in Invadosome Formation. Cancers (Basel) 2019; 11:cancers11050615. [PMID: 31052560 PMCID: PMC6562994 DOI: 10.3390/cancers11050615] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/24/2022] Open
Abstract
Invadopodia are actin-rich protrusions developed by transformed cells in 2D/3D environments that are implicated in extracellular matrix (ECM) remodeling and degradation. These structures have an undoubted association with cancer invasion and metastasis because invadopodium formation in vivo is a key step for intra/extravasation of tumor cells. Invadopodia are closely related to other actin-rich structures known as podosomes, which are typical structures of normal cells necessary for different physiological processes during development and organogenesis. Invadopodia and podosomes are included in the general term 'invadosomes,' as they both appear as actin puncta on plasma membranes next to extracellular matrix metalloproteinases, although organization, regulation, and function are slightly different. Integrins are transmembrane proteins implicated in cell-cell and cell-matrix interactions and other important processes such as molecular signaling, mechano-transduction, and cell functions, e.g., adhesion, migration, or invasion. It is noteworthy that integrin expression is altered in many tumors, and other pathologies such as cardiovascular or immune dysfunctions. Over the last few years, growing evidence has suggested a role of integrins in the formation of invadopodia. However, their implication in invadopodia formation and adhesion to the ECM is still not well known. This review focuses on the role of integrins in invadopodium formation and provides a general overview of the involvement of these proteins in the mechanisms of metastasis, taking into account classic research through to the latest and most advanced work in the field.
Collapse
Affiliation(s)
- Rafael Peláez
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Ana Pariente
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Álvaro Pérez-Sala
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| |
Collapse
|
5
|
Thatcher SE, Black JE, Tanaka H, Kohama K, Fultz ME, Cassis LA, Wright GL. Matrix Metalloproteinases -14, -9 and -2 are Localized to the Podosome and Involved in Podosome Development in the A7r5 Smooth Muscle Cell. ACTA ACUST UNITED AC 2017; 5. [PMID: 30931350 PMCID: PMC6436839 DOI: 10.13188/2332-3671.1000020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Aim The purpose of the study was to localize matrix metalloproteinase (MMP)-14, -9, and -2 in the A7r5 smooth muscle cell and to understand the interaction between these MMPs and the cytoskeleton. This interaction was observed under non-stimulating and phorbol 12, 13-dibutyrate (PDBu)-stimulating conditions. Methods Confocal microscopy was utilized to define the localizations of MMPs and tissue inhibitor of matrix metalloproteinases (TIMPs) in the A7r5 cell and to determine interaction between MMPs and the cytoskeleton. Under PDBu-stimulating conditions, the presence of MMP active forms and activity by gel zymography was evaluated in the A7r5 cell. Actin and microtubule-polymerization inhibitors were used to evaluate MMP interaction with the cytoskeleton and the cytoskeleton was observed on matrix and within a Type I collagen gel. Results MMP-14, -9, and -2 were localized to the podosome in the A7r5 smooth muscle cell and interactions were seen with these MMPs and the actin cytoskeleton. PDBu-stimulation induced increases in the protein abundance of the active forms of the MMPs and MMP-2 activity was increased. MMPs also interact with a-actin and not β-tubulin in the A7r5 cell. Galardin, also known as GM-6001, was shown to inhibit podosome formation and prevented MMP localization to the podosome. This broad spectrum MMP inhibitor also prevented collagen gel contraction and prevented cell adhesion and spreading of A7r5 cells within this collagen matrix. Conclusion MMPs are important in the formation and function of podosomes in the A7r5 smooth muscle cell. MMPs interact with a-actin and not β-tubulin in the A7r5 cell. Podosomes play an important role in cell migration and understanding the function of podosomes can lead to insights into cancer metastasis and cardiovascular disease.
Collapse
Affiliation(s)
- S E Thatcher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, USA
| | - J E Black
- Department of Physiology, Pharmacology and Toxicology, Marshall University, USA
| | - H Tanaka
- Department of Health Sciences, Gunma University, Japan
| | - K Kohama
- Research Institute of Pharmaceutical Sciences, Musashino University, Japan
| | - M E Fultz
- Department of Biology and Chemistry, Morehead State University, USA
| | - L A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, USA
| | - G L Wright
- Department of Physiology, Pharmacology and Toxicology, Marshall University, USA
| |
Collapse
|
6
|
Chi Q, Shan J, Ding X, Yin T, Wang Y, Jia D, Wang G. Smart mechanosensing machineries enable migration of vascular smooth muscle cells in atherosclerosis-relevant 3D matrices. Cell Biol Int 2017; 41:586-598. [PMID: 28328100 DOI: 10.1002/cbin.10764] [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: 01/18/2017] [Accepted: 03/19/2017] [Indexed: 11/05/2022]
Abstract
At the early stage of atherosclerosis, neointima is formed due to the migration of vascular smooth muscle cells (VSMCs) from the media to the intima. VSMCs are surrounded by highly adhesive 3D matrices. They take specific strategies to cross various 3D matrices in the media, including heterogeneous collagen and mechanically strong basement membrane. Migration of VSMCs is potentially caused by biomechanical mechanism. Most in vitro studies focus on cell migration on 2D substrates in response to biochemical factors. How the cells move through 3D matrices under the action of mechanosensing machineries remains unexplored. In this review, we propose that several interesting tension-dependent machineries act as "tractor"-posterior myosin II accumulation, and "wrecker"-anterior podosome maintaining, to power VSMCs ahead. VSMCs embedded in 3D matrices may accumulate a minor myosin II isoform, myosin IIB, at the cell rear. Anisotropic myosin IIB distribution creates cell rear, polarizes cell body, pushes the nucleus and reshapes the cell body, and cooperates with a uniformly distributed myosin IIA to propel the cell forward. On the other hand, matrix digestion by podosome further promote the migration when the matrix becomes denser. Actomyosin tension activates Src to induce podosome in soft 3D matrices and retain the podosome integrity to steadily digest the matrix.
Collapse
Affiliation(s)
- Qingjia Chi
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jieling Shan
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xiaorong Ding
- Department of Electronic Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Yazhou Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Dongyu Jia
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
7
|
Goicoechea SM, Zinn A, Awadia SS, Snyder K, Garcia-Mata R. A RhoG-mediated signaling pathway that modulates invadopodia dynamics in breast cancer cells. J Cell Sci 2017; 130:1064-1077. [PMID: 28202690 DOI: 10.1242/jcs.195552] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/14/2017] [Indexed: 01/11/2023] Open
Abstract
One of the hallmarks of cancer is the ability of tumor cells to invade surrounding tissues and metastasize. During metastasis, cancer cells degrade the extracellular matrix, which acts as a physical barrier, by developing specialized actin-rich membrane protrusion structures called invadopodia. The formation of invadopodia is regulated by Rho GTPases, a family of proteins that regulates the actin cytoskeleton. Here, we describe a novel role for RhoG in the regulation of invadopodia disassembly in human breast cancer cells. Our results show that RhoG and Rac1 have independent and opposite roles in the regulation of invadopodia dynamics. We also show that SGEF (also known as ARHGEF26) is the exchange factor responsible for the activation of RhoG during invadopodia disassembly. When the expression of either RhoG or SGEF is silenced, invadopodia are more stable and have a longer lifetime than in control cells. Our findings also demonstrate that RhoG and SGEF modulate the phosphorylation of paxillin, which plays a key role during invadopodia disassembly. In summary, we have identified a novel signaling pathway involving SGEF, RhoG and paxillin phosphorylation, which functions in the regulation of invadopodia disassembly in breast cancer cells.
Collapse
Affiliation(s)
- Silvia M Goicoechea
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Ashtyn Zinn
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Sahezeel S Awadia
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Kyle Snyder
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|
8
|
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
|
9
|
Liu FT, Xue QZ, Zhu PQ, Luo HL, Zhang Y, Hao T. Long noncoding RNA AFAP1-AS1, a potential novel biomarker to predict the clinical outcome of cancer patients: a meta-analysis. Onco Targets Ther 2016; 9:4247-54. [PMID: 27471399 PMCID: PMC4948684 DOI: 10.2147/ott.s107188] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
A number of studies have demonstrated that the expression level of actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) was upregulated in various cancers. High expression of AFAP1-AS1 is associated with an increased risk of metastasis and a poor prognosis in cancer patients. The electronic search was conducted in PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, and Wanfang database. We collected relevant articles to explore the association between the expression levels of AFAP1-AS1 and lymph node metastasis, distant metastasis, overall survival, relapse-free survival, and progression-free survival. A total of 1,017 patients from eight studies were finally included. The results showed that cancer patients with high AFAP1-AS1 expression suffered an increased risk of developing lymph node metastasis (odds ratio =3.19, 95% confidence interval [CI]: 2.11-4.83, P<0.00001) and distant metastasis (odds ratio =3.05, 95% CI: 1.84-5.04, P<0.0001). Moreover, we found that patients with high AFAP1-AS1 expression also had a poorer overall survival (hazard ratio [HR]: 1.98, 95% CI: 1.57-2.38, P=0.000), a worse progression-free survival (HR: 1.73, 95% CI: 1.11-2.35, P=0.000), and a shorter recurrence-free survival (HR: 1.96, 95% CI: 1.02-2.90, P=0.000) than those with low AFAP1-AS1 expression. High expression of AFAP1-AS1 was associated with poor clinical outcome. AFAP1-AS1 might serve as a potential novel biomarker for indicating the clinical outcomes in human cancers.
Collapse
Affiliation(s)
- Fang-Teng Liu
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province
| | - Qi-Zhen Xue
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, People's Republic of China
| | - Pei-Qian Zhu
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province
| | - Hong-Liang Luo
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province
| | - Yi Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province
| | - Tengfei Hao
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province
| |
Collapse
|
10
|
Chen Y, Liu Y, Guo J, Tang T, Gao J, Huang T, Wang B, Liu S. Preparation and Characterization of a Polyclonal Antibody against Human Actin Filament-Associated Protein-120 kD. Int J Mol Sci 2016; 17:ijms17060942. [PMID: 27322249 PMCID: PMC4926475 DOI: 10.3390/ijms17060942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/02/2016] [Accepted: 06/08/2016] [Indexed: 12/17/2022] Open
Abstract
Actin filament-associated protein-120kD (AFAP-120) is an alternatively spliced isoform of actin filament-associated protein-110kD (AFAP-110) and contains an additional neuronal insert (NINS) fragment in addition to identical domains to the AFAP-110. Unlike AFAP-110 widely expressed in tissues, AFAP-120 is specifically expressed in the nervous system and plays a role in organizing dynamic actin structures during neuronal differentiation. However, anti-AFAP-120 antibody is still commercially unavailable, and this may hinder the function research for AFAP-120. In this study, we simultaneously used the ABCpred online server and the BepiPred 1.0 server to predict B-cell epitopes in the exclusive NINS sequence of human AFAP-120 protein, and found that a 16aa-peptide sequence was the consensus epitope predicted by both tools. This peptide was chemically synthesized and used as an immunogen to develop polyclonal antibody against AFAP-120 (anti-AFAP-120). The sensitivity and specificity of anti-AFAP-120 were analyzed with immunoblotting, immunoprecipitation, and immunofluorescence assays. Our results indicated that anti-AFAP-120 could react with over-expressed and endogenous human AFAP-120 protein under denatured condition, but not with human AFAP-110 protein. Moreover, native human AFAP-120 protein could also be recognized by the anti-AFAP-120 antibody. These results suggested that the prepared anit-AFAP-120 antibody would be a useful tool for studying the biochemical and biological functions of AFAP-120.
Collapse
Affiliation(s)
- Yujian Chen
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Yong Liu
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Jiayu Guo
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Tao Tang
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Jian Gao
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Tao Huang
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Bin Wang
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Shaojun Liu
- Department of Neurobiology, Institute of Basic Medical Sciences, Beijing 100850, China.
| |
Collapse
|
11
|
Watson LJ, Alexander KM, Mohan ML, Bowman AL, Mangmool S, Xiao K, Naga Prasad SV, Rockman HA. Phosphorylation of Src by phosphoinositide 3-kinase regulates beta-adrenergic receptor-mediated EGFR transactivation. Cell Signal 2016; 28:1580-92. [PMID: 27169346 DOI: 10.1016/j.cellsig.2016.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/03/2016] [Accepted: 05/05/2016] [Indexed: 01/08/2023]
Abstract
β2-Adrenergic receptors (β2AR) transactivate epidermal growth factor receptors (EGFR) through formation of a β2AR-EGFR complex that requires activation of Src to mediate signaling. Here, we show that both lipid and protein kinase activities of the bifunctional phosphoinositide 3-kinase (PI3K) enzyme are required for β2AR-stimulated EGFR transactivation. Mechanistically, the generation of phosphatidylinositol (3,4,5)-tris-phosphate (PIP3) by the lipid kinase function stabilizes β2AR-EGFR complexes while the protein kinase activity of PI3K regulates Src activation by direct phosphorylation. The protein kinase activity of PI3K phosphorylates serine residue 70 on Src to enhance its activity and induce EGFR transactivation following βAR stimulation. This newly identified function for PI3K, whereby Src is a substrate for the protein kinase activity of PI3K, is of importance since Src plays a key role in pathological and physiological signaling.
Collapse
Affiliation(s)
- Lewis J Watson
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Kevin M Alexander
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Maradumane L Mohan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, United States
| | - Amber L Bowman
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand
| | - Kunhong Xiao
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States; Department of Pharmacology and Chemical Biology, University of Pittsburg School of Medicine, Pittsburgh, PA 15261, United States
| | - Sathyamangla V Naga Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, United States.
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
12
|
Revach OY, Winograd-Katz SE, Samuels Y, Geiger B. The involvement of mutant Rac1 in the formation of invadopodia in cultured melanoma cells. Exp Cell Res 2016; 343:82-88. [PMID: 26873115 DOI: 10.1016/j.yexcr.2016.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 02/04/2016] [Indexed: 01/07/2023]
Abstract
In this article, we discuss the complex involvement of a Rho-family GTPase, Rac1, in cell migration and in invadopodia-mediated matrix degradation. We discuss the involvement of invadopodia in invasive cell migration, and their capacity to promote cancer metastasis. Considering the regulation of invadopodia formation, we describe studies that demonstrate the role of Rac1 in the metastatic process, and the suggestion that this effect is attributable to the capacity of Rac1 to promote invadopodia formation. This notion is demonstrated here by showing that knockdown of Rac1 in melanoma cells expressing a wild-type form of this GTPase, reduces invadopodia-dependent matrix degradation. Interestingly, we also show that excessive activity of Rac1, displayed by the P29S, hyperactive, "fast cycling" mutant of Rac1, which is present in 5-10% of melanoma tumors, inhibits invadopodia function. Moreover, knockdown of this hyperactive mutant enhanced matrix degradation, indicating that excessive Rac1 activity by this mutant can negatively regulate invadopodia formation and function.
Collapse
Affiliation(s)
- Or-Yam Revach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sabina E Winograd-Katz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
13
|
Zhang R, Zhang J, Wu Q, Meng F, Liu C. XB130: A novel adaptor protein in cancer signal transduction. Biomed Rep 2016; 4:300-306. [PMID: 26998266 DOI: 10.3892/br.2016.588] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022] Open
Abstract
Adaptor proteins are functional proteins that contain two or more protein-binding modules to link signaling proteins together, which affect cell growth and shape and have no enzymatic activity. The actin filament-associated protein (AFAP) family is an important member of the adaptor proteins, including AFAP1, AFAP1L1 and AFAP1L2/XB130. AFAP1 and AFAP1L1 share certain common characteristics and function as an actin-binding protein and a cSrc-activating protein. XB130 exhibits certain unique features in structure and function. The mRNA of XB130 is expressed in human spleen, thyroid, kidney, brain, lung, pancreas, liver, colon and stomach, and the most prominent disease associated with XB130 is cancer. XB130 has a controversial effect on cancer. Studies have shown that XB130 can promote cancer progression and downregulation of XB130-reduced growth of tumors derived from certain cell lines. A higher mRNA level of XB130 was shown to be associated with a better survival in non-small cell lung cancer. Previous studies have shown that XB130 can regulate cell growth, migration and invasion and possibly has the effect through the cAMP-cSrc-phosphoinositide 3-kinase/Akt pathway. Except for cancer, XB130 is also associated with other pathological or physiological procedures, such as airway repair and regeneration.
Collapse
Affiliation(s)
- Ruiyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Qifei Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Fandi Meng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| |
Collapse
|
14
|
AFAP1 Is a Novel Downstream Mediator of TGF-β1 for CCN2 Induction in Osteoblasts. PLoS One 2015; 10:e0136712. [PMID: 26340021 PMCID: PMC4560384 DOI: 10.1371/journal.pone.0136712] [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] [Received: 09/10/2014] [Accepted: 08/07/2015] [Indexed: 02/06/2023] Open
Abstract
Background CCN2 acts as an anabolic growth factor to regulate osteoblast differentiation and function. CCN2 is induced by TGF-β1 and acts as a mediator of TGF-β1 induced matrix production in osteoblasts and Src is required for CCN2 induction by TGF-β1; however, the molecular mechanisms that control CCN2 induction in osteoblasts are poorly understood. AFAP1 binds activated forms of Src and can direct the activation of Src in certain cell types, however a role for AFAP1 downstream of TGF-β1 or in osteoblats is undefined. In this study, we investigated the role of AFAP1 for CCN2 induction by TGF-β1 in primary osteoblasts. Results We demonstrated that AFAP1 expression in osteoblasts occurs in a biphasic pattern with maximal expression levels occurring during osteoblast proliferation (~day 3), reduced expression during matrix production/maturation (~day 14–21), an a further increase in expression during mineralization (~day 21). AFAP1 expression is induced by TGF-β1 treatment in osteoblasts during days 7, 14 and 21. In osteoblasts, AFAP1 binds to Src and is required for Src activation by TGF-β1 and CCN2 promoter activity and protein induction by TGF-β1 treatment was impaired using AFAP1 siRNA, indicating the requirement of AFAP1 for CCN2 induction by TGF-β1. We also demonstrated that TGF-β1 induction of extracellular matrix protein collagen XIIa occurs in an AFAP1 dependent fashion. Conclusions This study demonstrates that AFAP1 is an essential downstream signaling component of TGF-β1 for Src activation, CCN2 induction and collagen XIIa in osteoblasts.
Collapse
|
15
|
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]
|
16
|
Revach OY, Geiger B. The interplay between the proteolytic, invasive, and adhesive domains of invadopodia and their roles in cancer invasion. Cell Adh Migr 2015; 8:215-25. [PMID: 24714132 DOI: 10.4161/cam.27842] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Invadopodia are actin-based protrusions of the plasma membrane that penetrate into the extracellular matrix (ECM), and enzymatically degrade it. Invadopodia and podosomes, often referred to, collectively, as "invadosomes," are actin-based membrane protrusions that facilitate matrix remodeling and cell invasion across tissues, processes that occur under specific physiological conditions such as bone remodeling, as well as under pathological states such as bone, immune disorders, and cancer metastasis. In this review, we specifically focus on the functional architecture of invadopodia in cancer cells; we discuss here three functional domains of invadopodia responsible for the metalloproteinase-based degradation of the ECM, the cytoskeleton-based mechanical penetration into the matrix, and the integrin adhesome-based adhesion to the ECM. We will describe the structural and molecular organization of each domain and the cross-talk between them during the invasion process.
Collapse
Affiliation(s)
- Or-Yam Revach
- Department of Molecular Cell Biology; Weizmann Institute of Science; Rehovot, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology; Weizmann Institute of Science; Rehovot, Israel
| |
Collapse
|
17
|
Genetic disruption of the sh3pxd2a gene reveals an essential role in mouse development and the existence of a novel isoform of tks5. PLoS One 2014; 9:e107674. [PMID: 25259869 PMCID: PMC4178035 DOI: 10.1371/journal.pone.0107674] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/12/2014] [Indexed: 01/07/2023] Open
Abstract
Tks5 is a scaffold protein and Src substrate involved in cell migration and matrix degradation through its essential role in invadosome formation and function. We have previously described that Tks5 is fundamental for zebrafish neural crest cell migration in vivo. In the present study, we sought to investigate the function of Tks5 in mammalian development by analyzing mice mutant for sh3pxd2a, the gene encoding Tks5. Homozygous disruption of the sh3pxd2a gene by gene-trapping in mouse resulted in neonatal death and the presence of a complete cleft of the secondary palate. Interestingly, embryonic fibroblasts from homozygous gene-trap sh3pxd2a mice lacked only the highest molecular weight band of the characteristic Tks5 triplet observed in protein extracts, leaving the lower molecular weight bands unaffected. This finding, together with the existence of two human Expressed Sequence Tags lacking the first 5 exons of SH3PXD2A, made us hypothesize about the presence of a second alternative transcription start site located in intron V. We performed 5′RACE on mouse fibroblasts and isolated a new transcript of the sh3pxd2a gene encoding a novel Tks5 isoform, that we named Tks5β. This novel isoform diverges from the long form of Tks5 in that it lacks the PX-domain, which confers affinity for phosphatidylinositol-3,4-bisphosphate. Instead, Tks5β has a short unique amino terminal sequence encoded by the newly discovered exon 6β; this exon includes a start codon located 29 bp from the 5'-end of exon 6. Tks5β mRNA is expressed in MEFs and all mouse adult tissues analyzed. Tks5β is a substrate for the Src tyrosine kinase and its expression is regulated through the proteasome degradation pathway. Together, these findings indicate the essentiality of the larger Tks5 isoform for correct mammalian development and the transcriptional complexity of the sh3pxd2a gene.
Collapse
|
18
|
The Role of Src Kinase in the Caspase-1 Pathway After Hypoxia in the Brain of Newborn Piglets. Neurochem Res 2014; 39:2118-26. [DOI: 10.1007/s11064-014-1404-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/28/2014] [Accepted: 07/28/2014] [Indexed: 01/06/2023]
|
19
|
Cunnick JM, Kim S, Hadsell J, Collins S, Cerra C, Reiser P, Flynn DC, Cho Y. Actin filament-associated protein 1 is required for cSrc activity and secretory activation in the lactating mammary gland. Oncogene 2014; 34:2640-9. [PMID: 25043309 PMCID: PMC4302073 DOI: 10.1038/onc.2014.205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/25/2014] [Accepted: 06/09/2014] [Indexed: 12/12/2022]
Abstract
Actin filament-associated protein 1 (AFAP1) is an adaptor protein of cSrc that binds to filamentous actin and regulates the activity of this tyrosine kinase to affect changes to the organization of the actin cytoskeleton. In breast and prostate cancer cells, AFAP1 has been shown to regulate cellular responses requiring actin cytoskeletal changes such as adhesion, invadopodia formation and invasion. However, a normal physiologic role for AFAP1 has remained elusive. In this study, we generated an AFAP1 knockout mouse model that establishes a novel physiologic role for AFAP1 in lactation. Specifically, these animals displayed a defect in lactation that resulted in an inability to nurse efficiently. Histologically, the mammary glands of the lactating knockout mice were distinguished by the accumulation of large cytoplasmic lipid droplets in the alveolar epithelial cells. There was a reduction in lipid synthesis and the expression of lipogenic genes without a corresponding reduction in the production of β-casein, a milk protein. Furthermore, these defects were associated with histologic and biochemical signs of precocious involution. This study also demonstrated that AFAP1 responds to prolactin, a lactogenic hormone, by forming a complex with cSrc and becoming tyrosine phosphorylated. Taken together, these observations pointed to a defect in secretory activation. Certain characteristics of this phenotype mirrored the defect in secretory activation in the cSrc knockout mouse, but most importantly, the activity of cSrc in the mammary gland was reduced during early lactation in the AFAP1-null mouse and the localization of active cSrc at the apical surface of luminal epithelial cells during lactation was selectively lost in the absence of AFAP1. These data define, for the first time, the requirement of AFAP1 for the spatial and temporal regulation of cSrc activity in the normal breast, specifically for milk production.
Collapse
Affiliation(s)
- J M Cunnick
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA, USA
| | - S Kim
- Graduate School of Medicine, The Commonwealth Medical College, Scranton, PA, USA
| | - J Hadsell
- Fortis Institute Scranton, Scranton, PA, USA
| | - S Collins
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA, USA
| | - C Cerra
- Department of Pathology, Pocono Health System, East Stroudsburg, PA, USA
| | - P Reiser
- Department of Pathology, Pocono Health System, East Stroudsburg, PA, USA
| | - D C Flynn
- College of Health Science, University of Delaware, Newark, DE, USA
| | - Y Cho
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA, USA
| |
Collapse
|
20
|
Abstract
The occurrence of invadopodia has been, since its characterization, a hallmark of cancerous cell invasion and metastasis. These structures are now the subject of a controversy concerning their cellular function, molecular regulation, and assembly. The terms invadopodia and podosomes have been used interchangeably since their discovery back in 1980. Since then, these phenotypes are now more established and accepted by the scientific community as vital structures for 3D cancer cell motility. Many characteristics relating to invadopodia and podosomes have been elucidated, which might prove these structures as good targets for metastasis treatment. In this review, we briefly review the actin reorganization process needed in most types of cancer cell motility. We also review the important characteristics of invadopodia, including molecular components, assembly, markers, and the signaling pathways, providing a comprehensive model for invadopodia regulation.
Collapse
Affiliation(s)
- Bechara A Saykali
- Department of Natural Sciences, The Lebanese American University , Beirut , Lebanon
| | | |
Collapse
|
21
|
Takahashi R, Nagayama S, Furu M, Kajita Y, Jin Y, Kato T, Imoto S, Sakai Y, Toguchida J. AFAP1L1, a novel associating partner with vinculin, modulates cellular morphology and motility, and promotes the progression of colorectal cancers. Cancer Med 2014; 3:759-74. [PMID: 24723436 PMCID: PMC4303145 DOI: 10.1002/cam4.237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/17/2014] [Accepted: 03/03/2014] [Indexed: 12/18/2022] Open
Abstract
We have previously identified actin filament-associated protein 1-like 1 (AFAP1L1) as a metastasis-predicting marker for spindle cell sarcomas by gene expression profiling, and demonstrated that AFAP1L1 is involved in the cell invasion process by in vitro analyses. However, its precise molecular function has not been fully elucidated, and it remains unknown whether AFAP1L1 could be a prognostic marker and/or therapeutic target of other malignancies. In this study, we found a marked elevation of AFAP1L1 gene expression in colorectal cancer (CRC) tissues as compared to the adjacent normal mucosa. Multivariate analysis revealed that AFAP1L1 was an independent and significant factor for the recurrence of rectal cancers. Moreover, the addition of the AFAP1L1 expression level to the lymph node metastasis status provided more predictive information regarding postoperative recurrence in rectal cancers. AFAP1L1-transduced CRC cells exhibited a rounded shape, increased cell motility on planar substrates, and resistance to anoikis in vitro. AFAP1L1 localized to the ringed structure of the invadopodia, together with vinculin, and AFAP1L1 was identified as a novel associating partner of vinculin by immunoprecipitation assay. AFAP1L1-transduced cells showed accelerated tumor growth in vivo, presumably reflecting the anoikis resistance of these AFAP1L1-expressing cells. Furthermore, the local administration of a siRNA against AFAP1L1 significantly suppressed the in vivo tumor growth of xenografts, suggesting that AFAP1L1 might be a candidate therapeutic target for CRCs. These results suggest that AFAP1L1 plays a role in the progression of CRCs by modulating cell shape and motility and by inhibiting anoikis, presumably through interactions with vinculin-including protein complexes.
Collapse
Affiliation(s)
- Ryo Takahashi
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shi M, Zheng D, Sun L, Wang L, Lin L, Wu Y, Zhou M, Liao W, Liao Y, Zuo Q, Liao W. XB130 promotes proliferation and invasion of gastric cancer cells. J Transl Med 2014; 12:1. [PMID: 24387290 PMCID: PMC3882781 DOI: 10.1186/1479-5876-12-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 12/27/2013] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND XB130 has been reported to be expressed by various types of cells such as thyroid cancer and esophageal cancer cells, and it promotes the proliferation and invasion of thyroid cancer cells. Our previous study demonstrated that XB130 is also expressed in gastric cancer (GC), and that its expression is associated with the prognosis, but the role of XB130 in GC has not been well characterized. METHODS In this study, we investigated the influence of XB130 on gastric tumorigenesis and metastasis in vivo and in vitro using the MTT assay, clonogenic assay, BrdU incorporation assay, 3D culture, immunohistochemistry and immunofluorescence. Western blot analysis was also performed to identify the potential mechanisms involved. RESULTS The proliferation, migration, and invasion of SGC7901 and MNK45 gastric adenocarcinoma cell lines were all significantly inhibited by knockdown of XB130 using small hairpin RNA. In a xenograft model, tumor growth was markedly inhibited after shXB130-transfected GC cells were implanted into nude mice. After XB130 knockdown, GC cells showed a more epithelial-like phenotype, suggesting an inhibition of the epithelial-mesenchymal transition (EMT) process. In addition, silencing of XB130 reduced the expression of p-Akt/Akt, upregulated expression of epithelial markers including E-cadherin, α-catenin and β-catenin, and downregulated mesenchymal markers including fibronectin and vimentin. Expression of oncoproteins related to tumor metastasis, such as MMP2, MMP9, and CD44, was also significantly reduced. CONCLUSIONS These findings indicate that XB130 enhances cell motility and invasiveness by modulating the EMT-like process, while silencing XB130 in GC suppresses tumorigenesis and metastasis, suggesting that it may be a potential therapeutic target.
Collapse
Affiliation(s)
- Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Dayong Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Lin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Li Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Yajun Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Minyu Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Wenjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Qiang Zuo
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| |
Collapse
|
23
|
Reynolds AB, Kanner SB, Bouton AH, Schaller MD, Weed SA, Flynn DC, Parsons JT. SRChing for the substrates of Src. Oncogene 2013; 33:4537-47. [PMID: 24121272 DOI: 10.1038/onc.2013.416] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/16/2013] [Accepted: 08/17/2013] [Indexed: 12/12/2022]
Abstract
By the mid 1980's, it was clear that the transforming activity of oncogenic Src was linked to the activity of its tyrosine kinase domain and attention turned to identifying substrates, the putative next level of control in the pathway to transformation. Among the first to recognize the potential of phosphotyrosine-specific antibodies, Parsons and colleagues launched a risky shotgun-based approach that led ultimately to the cDNA cloning and functional characterization of many of today's best-known Src substrates (for example, p85-Cortactin, p110-AFAP1, p130Cas, p125FAK and p120-catenin). Two decades and over 6000 citations later, the original goals of the project may be seen as secondary to the enormous impact of these protein substrates in many areas of biology. At the request of the editors, this review is not restricted to the current status of the substrates, but reflects also on the anatomy of the project itself and some of the challenges and decisions encountered along the way.
Collapse
Affiliation(s)
- A B Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - S B Kanner
- Arrowhead Research Corporation, Madison, WI, USA
| | - A H Bouton
- Departments of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - M D Schaller
- Department of Biochemistry, 3124 HSN, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - S A Weed
- Department of Neurobiology and Anatomy, 1833 Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - D C Flynn
- Department of Medical Lab Sciences, College of Health Sciences, University of Delaware, Newark, DE, USA
| | - J T Parsons
- Departments of Microbiology, Immunology and Cancer Biology, University of Virginia Cancer Center, Charlottesville, VA, USA
| |
Collapse
|
24
|
Xiao H, Liu M. Atypical protein kinase C in cell motility. Cell Mol Life Sci 2013; 70:3057-66. [PMID: 23096778 PMCID: PMC11113714 DOI: 10.1007/s00018-012-1192-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 09/03/2012] [Accepted: 10/08/2012] [Indexed: 01/01/2023]
Abstract
Cell motility is defined as cell movement in the three-dimensional space leading to repositioning of the cell. Atypical protein kinase C (aPKC, including ζ and λ/ι) are a subfamily of PKC. Different from classic PKC and novel PKC, the activation of atypical PKC is not dependent on diacylglycerol or calcium. PKCζ can be activated by lipid components, such as phosphatidylinositols, phosphatidic acid, arachidonic acid, and ceramide. Both phosphatidylinositol (3,4,5)-trisphosphate and PDK1 are necessary for the complete and stable activation of PKCζ. Atypical PKC is involved in the regulation of cell polarization, directional sensing, formation of filopodia, and cell motility. It is essential for migration and invasion of multiple cancer cell types. Particularly, atypical PKC has been found in the regulation of the motility of hematopoietic cells. It also participates in the regulation of proteolytic activity of podosomes and invadopodia. It has been found that atypical PKC can work coordinately with other PKC subfamily members and other signaling pathways. Research on the roles of atypical PKC in cell motility may lead to new therapeutic strategies for cancer and other diseases.
Collapse
Affiliation(s)
- Helan Xiao
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| | | |
Collapse
|
25
|
Abstract
Remodeling of extracellular matrix (ECM) is a fundamental cell property that allows cells to alter their microenvironment and move through tissues. Invadopodia and podosomes are subcellular actin-rich structures that are specialized for matrix degradation and are formed by cancer and normal cells, respectively. Although initial studies focused on defining the core machinery of these two structures, recent studies have identified inputs from both growth factor and adhesion signaling as crucial for invasive activity. This Commentary will outline the current knowledge on the upstream signaling inputs to invadopodia and podosomes and their role in governing distinct stages of these invasive structures. We discuss invadopodia and podosomes as adhesion structures and highlight new data showing that invadopodia-associated adhesion rings promote the maturation of already-formed invadopodia. We present a model in which growth factor stimulation leads to phosphoinositide 3-kinase (PI3K) activity and formation of invadopodia, whereas adhesion signaling promotes exocytosis of proteinases at invadopodia.
Collapse
Affiliation(s)
- Daisuke Hoshino
- Department of Cancer Biology, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN 37232-6840, USA
| | | | | |
Collapse
|
26
|
Gulia J, Navedo MF, Gui P, Chao JT, Mercado JL, Santana LF, Davis MJ. Regulation of L-type calcium channel sparklet activity by c-Src and PKC-α. Am J Physiol Cell Physiol 2013; 305:C568-77. [PMID: 23804206 DOI: 10.1152/ajpcell.00381.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The activity of persistent Ca²⁺ sparklets, which are characterized by longer and more frequent channel open events than low-activity sparklets, contributes substantially to steady-state Ca²⁺ entry under physiological conditions. Here, we addressed two questions related to the regulation of Ca²⁺ sparklets by PKC-α and c-Src, both of which increase whole cell Cav1.2 current: 1) Does c-Src activation enhance persistent Ca²⁺ sparklet activity? 2) Does PKC-α activate c-Src to produce persistent Ca²⁺ sparklets? With the use of total internal reflection fluorescence microscopy, Ca²⁺ sparklets were recorded from voltage-clamped tsA-201 cells coexpressing wild-type (WT) or mutant Cav1.2c (the neuronal isoform of Cav1.2) constructs ± active or inactive PKC-α/c-Src. Cells expressing Cav1.2c exhibited both low-activity and persistent Ca²⁺ sparklets. Persistent Ca²⁺ sparklet activity was significantly reduced by acute application of the c-Src inhibitor PP2 or coexpression of kinase-dead c-Src. Cav1.2c constructs mutated at one of two COOH-terminal residues (Y²¹²²F and Y²¹³⁹F) were used to test the effect of blocking putative phosphorylation sites for c-Src. Expression of Y²¹²²F but not Y²¹³⁹F Cav1.2c abrogated the potentiating effect of c-Src on Ca²⁺ sparklet activity. We could not detect a significant change in persistent Ca²⁺ sparklet activity or density in cells coexpressing Cav1.2c + PKC-α, regardless of whether WT or Y²¹²²F Cav1.2c was used, or after PP2 application, suggesting that PKC-α does not act upstream of c-Src to produce persistent Ca²⁺ sparklets. However, our results indicate that persistent Ca²⁺ sparklet activity is promoted by the action of c-Src on residue Y²¹²² of the Cav1.2c COOH terminus.
Collapse
Affiliation(s)
- Jyoti Gulia
- Department of Biological Engineering University of Missouri, Columbia, Missouri, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Xiao H, Bai XH, Wang Y, Kim H, Mak AS, Liu M. MEK/ERK pathway mediates PKC activation-induced recruitment of PKCζ and MMP-9 to podosomes. J Cell Physiol 2013; 228:416-27. [PMID: 22740332 DOI: 10.1002/jcp.24146] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Podosomes are adhesive structures on the ventral surface of cells that invade and degrade the extracellular matrix. Recently, we reported that phorbol 12,13-dibutyrate (PDBu), a protein kinase C (PKC) activator, induced podosome formation in normal human bronchial epithelial (NHBE) cells, and atypical PKCζ regulated MMP-9 recruitment to podosomes for its release and activation. The objective of this study was to explore signaling pathways that are involved in PKC activation-induced podosome formation and matrix degradation. Herein, we found that PDBu increased phosphorylation of PI3K p85, Akt, Src, ERK1/2, and JNK. Inhibitors for PI3K, Akt, and Src suppressed PDBu-induced podosome formation and matrix degradation. In contrast, blockers for MEK/ERK or JNK did not inhibit podosome formation but reduced proteolytic activity of podosomes. Inhibition of PKCζ activity with its pseudosubstrate peptide (PS)-inhibited PDBu-induced phosphorylation of MEK/ERK and JNK. On the other hand, inhibition of MEK/ERK or JNK pathway did not affect PKCζ phosphorylation, but reduced the recruitment of PKCζ and MMP-9 to podosomes. We conclude that PKCζ may regulate MEK/ERK and JNK phosphorylation and in turn activated MEK/ERK and JNK may regulate the proteolytic activity of PDBu-induced podosomes by influencing the recruitment of PKCζ and MMP-9 to podosomes.
Collapse
Affiliation(s)
- Helan Xiao
- Division of Cell and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
28
|
Junctional adhesion molecules 2 and 3 may potentially be involved in progression of gastric adenocarcinoma tumors. Med Oncol 2013; 30:380. [PMID: 23277282 DOI: 10.1007/s12032-012-0380-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 12/03/2012] [Indexed: 12/13/2022]
Abstract
Tight junctions (TJs) of epithelia are responsible for integrity of polarized epithelial cells. It is now well established that the deregulation of their functions and expressions contribute to initiation and progression of cancer through activation of cytoskeleton machinery. The aim of this study was to examine the expression level of two genes encoding tight junction-associated proteins of Jam2 and Jam3 in gastric adenocarcinoma and compare with normal gastric tissues dissected from same patients. Significant difference of expression level for these genes was observed between tumor and adjacent normal tissues. Also, we analyzed the expression level of actin filament-associated protein gene that appears to be a downstream factor of JAM2 and JAM3. The expression level of this gene was significantly higher in tumor tissues. Some correlations between the expression level of these genes with each other and with pathological features were observed. These data brought new evidences for the role of these three genes in progression of gastric adenocarcinoma.
Collapse
|
29
|
Hoshino D, Jourquin J, Emmons SW, Miller T, Goldgof M, Costello K, Tyson DR, Brown B, Lu Y, Prasad NK, Zhang B, Mills GB, Yarbrough WG, Quaranta V, Seiki M, Weaver AM. Network analysis of the focal adhesion to invadopodia transition identifies a PI3K-PKCα invasive signaling axis. Sci Signal 2012; 5:ra66. [PMID: 22969158 DOI: 10.1126/scisignal.2002964] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In cancer, deregulated signaling can produce an invasive cellular phenotype. We modeled the invasive transition as a theoretical switch between two cytoskeletal structures: focal adhesions and extracellular matrix-degrading invadopodia. We constructed molecular interaction networks of each structure and identified upstream regulatory hubs through computational analyses. We compared these regulatory hubs to the status of signaling components from head and neck carcinomas, which led us to analyze phosphatidylinositol 3-kinase (PI3K) and protein kinase C α (PKCα). Consistent with previous studies, PI3K activity promoted both the formation and the activity of invadopodia. We found that PI3K induction of invadopodia was increased by overexpression of SH2 (Src homology 2) domain-containing inositol 5'-phosphatase 2 (SHIP2), which converts the phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P(3)] that is produced by PI3K activity to phosphatidylinositol 3,4-bisphosphate [PI(3,4)P(2)], which is believed to promote invadopodia formation. Knockdown of PKCα had divergent effects on invadopodia formation, depending on the status of PI3K. Loss of PKCα inhibited invadopodia formation in cells with wild-type PI3K pathway status. Conversely, in cells with constitutively active PI3K (through activating PI3K mutants or lacking the endogenous opposing enzyme PTEN), PKCα knockdown increased invadopodia formation. Mechanistic studies revealed a negative feedback loop from PKCα that dampened PI3K activity and invasive behavior in cells with genetic hyperactivation of the PI3K pathway. These studies demonstrated the potential of network modeling as a discovery tool and identified PI3K and PKCα as interacting regulators of invasive behavior.
Collapse
Affiliation(s)
- Daisuke Hoshino
- Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Spatiotemporal regulation of Src and its substrates at invadosomes. Eur J Cell Biol 2012; 91:878-88. [PMID: 22823952 DOI: 10.1016/j.ejcb.2012.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 06/19/2012] [Accepted: 06/19/2012] [Indexed: 01/07/2023] Open
Abstract
In the past decade, substantial progress has been made in understanding how Src family kinases regulate the formation and function of invadosomes. Invadosomes are organized actin-rich structures that contain an F-actin core surrounded by an adhesive ring and mediate invasive migration. Src kinases orchestrate, either directly or indirectly, each phase of the invadosome life cycle including invadosome assembly, maturation and matrix degradation and disassembly. Complex arrays of Src effector proteins are involved at different stages of invadosome maturation and their spatiotemporal activity must be tightly regulated to achieve effective invasive migration. In this review, we highlight some recent progress and the challenges of understanding how Src is regulated temporally and spatially to orchestrate the dynamics of invadosomes and mediate cell invasion.
Collapse
|
31
|
Yamanaka D, Akama T, Fukushima T, Nedachi T, Kawasaki C, Chida K, Minami S, Suzuki K, Hakuno F, Takahashi SI. Phosphatidylinositol 3-kinase-binding protein, PI3KAP/XB130, is required for cAMP-induced amplification of IGF mitogenic activity in FRTL-5 thyroid cells. Mol Endocrinol 2012; 26:1043-55. [PMID: 22496359 DOI: 10.1210/me.2011-1349] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
We previously demonstrated that long-term pretreatment of rat FRTL-5 thyroid cells with TSH or cAMP-generating reagents potentiated IGF-I-dependent DNA synthesis. Under these conditions, cAMP treatment increased tyrosine phosphorylation of a 125-kDa protein (p125) and its association with a p85 regulatory subunit of phosphatidylinositol 3-kinase (p85 PI3K), which were suggested to mediate potentiation of DNA synthesis. This study was undertaken to identify p125 and to elucidate its roles in potentiation of DNA synthesis induced by IGF-I. Matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) analysis revealed p125 to be a rat ortholog of human XB130, which we named PI3K-associated protein (PI3KAP). cAMP treatment elevated PI3KAP/XB130 mRNA and protein levels as well as tyrosine phosphorylation and interaction with p85 PI3K leading to increased PI3K activities associated with PI3KAP/XB130, supporting the role of PI3KAP/XB130 in DNA synthesis potentiation. Importantly, PI3KAP/XB130 knockdown attenuated cAMP-dependent potentiation of IGF-I-induced DNA synthesis. Furthermore, c-Src was associated with PI3KAP/XB130 and was activated in response to cAMP. Addition of Src family kinase inhibitors, PP1 or PP2, during cAMP treatment abolished tyrosine phosphorylation of PI3KAP/XB130 and its interaction with p85 PI3K. Finally, introduction of PI3KAP/XB130 into NIH3T3 fibroblasts lacking endogenous PI3KAP/XB130 enhanced IGF-I-induced DNA synthesis; however, a mutant Y72F incapable of binding to p85 PI3K did not show this response. Together, these data indicate that cAMP-dependent induction of PI3KAP/XB130, which is associated with PI3K, is required for enhancement of IGF mitogenic activities.
Collapse
Affiliation(s)
- Daisuke Yamanaka
- Department of Animal Sciences, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Xiao H, Han B, Lodyga M, Bai XH, Wang Y, Liu M. The actin-binding domain of actin filament-associated protein (AFAP) is involved in the regulation of cytoskeletal structure. Cell Mol Life Sci 2012; 69:1137-51. [PMID: 21984596 PMCID: PMC11114525 DOI: 10.1007/s00018-011-0812-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 08/15/2011] [Accepted: 09/01/2011] [Indexed: 11/26/2022]
Abstract
Actin filament-associated protein (AFAP) plays a critical role in the regulation of actin filament integrity, formation and maintenance of the actin network, function of focal contacts, and cell migration. Here, we show that endogenous AFAP was present not only in the cytoskeletal but also in the cytosolic fraction. Depolymerization of actin filaments with cytochalasin D or latrunculin A increased AFAP in the cytosolic fraction. AFAP harbors an actin-binding domain (ABD) in its C-terminus. AFAPΔABD, an AFAP mutant with selective ABD deletion, was mainly in the cytosolic fraction when overexpressed in the cells, which was associated with a disorganized cytoskeleton with reduced stress fibers, accumulation of F-actin on cellular membrane, and formation of actin-rich small dots. Cortactin, a well-known podosome marker, was colocalized with AFAPΔABD in these small dots at the ventral surface of the cell, indicating that these small dots fulfill certain criteria of podosomes. However, these podosome-like small dots did not digest gelatin matrix. This may be due to the reduced interaction between AFAPΔABD and c-Src. When AFAPΔABD-transfected cells were stimulated with phorbol ester, they formed podosome-like structures with larger sizes, less numerous and longer life span, in comparison with wild-type AFAP-transfected cells. These results indicate that the association of AFAP with F-actin through ABD is crucial for AFAP to regulate cytoskeletal structures. The AFAPΔABD, as cytosolic proteins, may be more accessible to the cellular membrane, podosome-like structures, and thus be more interactive for the regulation of cellular functions.
Collapse
Affiliation(s)
- Helan Xiao
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, ON Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Bing Han
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, ON Canada
| | - Monika Lodyga
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, ON Canada
| | - Xiao-Hui Bai
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, ON Canada
| | - Yingchun Wang
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, ON Canada
| | - Mingyao Liu
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, ON Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Room TMDT 2-814, 101 College Street, Toronto, ON M5G 1L7 Canada
| |
Collapse
|
33
|
Invadosome regulation by adhesion signaling. Curr Opin Cell Biol 2011; 23:597-606. [DOI: 10.1016/j.ceb.2011.04.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 04/11/2011] [Accepted: 04/11/2011] [Indexed: 12/16/2022]
|
34
|
Linder S, Wiesner C, Himmel M. Degrading devices: invadosomes in proteolytic cell invasion. Annu Rev Cell Dev Biol 2011; 27:185-211. [PMID: 21801014 DOI: 10.1146/annurev-cellbio-092910-154216] [Citation(s) in RCA: 294] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Podosomes and invadopodia, collectively known as invadosomes, are cell-matrix contacts in a variety of cell types, such as monocytic cells or cancer cells, that have to cross tissue barriers. Both structures share an actin-rich core, which distinguishes them from other matrix contacts, and are regulated by a multitude of signaling pathways including RhoGTPases, kinases, actin-associated proteins, and microtubule-dependent transport. Invadosomes recruit and secrete proteinases and are thus able to lyse extracellular matrix components. They are therefore considered to be potential key structures in proteolytic cell invasion in both physiological and pathological settings. This review provides an overview of the field, with special focus on current developments such as intracellular transport processes, ultrastructural analysis, the possible involvement of invadosomes in disease, and the tentative identification of invadosomes in 3D environments and in vivo.
Collapse
Affiliation(s)
- Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany.
| | | | | |
Collapse
|
35
|
The 'ins' and 'outs' of podosomes and invadopodia: characteristics, formation and function. Nat Rev Mol Cell Biol 2011; 12:413-26. [PMID: 21697900 DOI: 10.1038/nrm3141] [Citation(s) in RCA: 815] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Podosomes and invadopodia are actin-based dynamic protrusions of the plasma membrane of metazoan cells that represent sites of attachment to - and degradation of - the extracellular matrix. The key proteins in these structures include the actin regulators cortactin and neural Wiskott-Aldrich syndrome protein (N-WASP), the adaptor proteins Tyr kinase substrate with four SH3 domains (TKS4) and Tyr kinase substrate with five SH3 domains (TKS5), and the metalloprotease membrane type 1 matrix metalloprotease (MT1MMP; also known as MMP14). Many cell types can produce these structures, including invasive cancer cells, vascular smooth muscle and endothelial cells, and immune cells such as macrophages and dendritic cells. Recently, progress has been made in our understanding of the regulatory and functional aspects of podosome and invadopodium biology and their role in human disease.
Collapse
|
36
|
Hu J, Mukhopadhyay A, Truesdell P, Chander H, Mukhopadhyay UK, Mak AS, Craig AWB. Cdc42-interacting protein 4 is a Src substrate that regulates invadopodia and invasiveness of breast tumors by promoting MT1-MMP endocytosis. J Cell Sci 2011; 124:1739-51. [PMID: 21525036 DOI: 10.1242/jcs.078014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Invadopodia are actin-rich membrane protrusions that promote extracellular matrix degradation and invasiveness of tumor cells. Src protein-tyrosine kinase is a potent inducer of invadopodia and tumor metastases. Cdc42-interacting protein 4 (CIP4) adaptor protein interacts with actin regulatory proteins and regulates endocytosis. Here, we show that CIP4 is a Src substrate that localizes to invadopodia in MDA-MB-231 breast tumor cells expressing activated Src (MDA-SrcYF). To probe the function of CIP4 in invadopodia, we established stable CIP4 knockdown in MDA-SrcYF cell lines by RNA interference. Compared with control cells, CIP4 knockdown cells degrade more extracellular matrix (ECM), have increased numbers of mature invadopodia and are more invasive through matrigel. Similar results are observed with knockdown of CIP4 in EGF-treated MDA-MB-231 cells. This inhibitory role of CIP4 is explained by our finding that CIP4 limits surface expression of transmembrane type I matrix metalloprotease (MT1-MMP), by promoting MT1-MMP internalization. Ectopic expression of CIP4 reduces ECM digestion by MDA-SrcYF cells, and this activity is enhanced by mutation of the major Src phosphorylation site in CIP4 (Y471). Overall, our results identify CIP4 as a suppressor of Src-induced invadopodia and invasion in breast tumor cells by promoting endocytosis of MT1-MMP.
Collapse
Affiliation(s)
- Jinghui Hu
- Department of Biochemistry, Queen's University, Kingston, ON K7L 3N6 Canada
| | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Rangrez AY, Massy ZA, Metzinger-Le Meuth V, Metzinger L. miR-143 and miR-145: molecular keys to switch the phenotype of vascular smooth muscle cells. CIRCULATION. CARDIOVASCULAR GENETICS 2011; 4:197-205. [PMID: 21505201 DOI: 10.1161/circgenetics.110.958702] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Snyder BN, Cho Y, Qian Y, Coad JE, Flynn DC, Cunnick JM. AFAP1L1 is a novel adaptor protein of the AFAP family that interacts with cortactin and localizes to invadosomes. Eur J Cell Biol 2011; 90:376-89. [PMID: 21333378 DOI: 10.1016/j.ejcb.2010.11.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 11/23/2010] [Accepted: 11/24/2010] [Indexed: 11/26/2022] Open
Abstract
The actin-filament associated protein (AFAP) family of adaptor proteins consists of three members: AFAP1, AFAP1L1, and AFAP1L2/XB130 with AFAP1 being the best described as a cSrc binding partner and actin cross-linking protein. A homology search of AFAP1 recently identified AFAP1L1 which has a similar sequence, domain structure and cellular localization; however, based upon sequence variations, AFAP1L1 is hypothesized to have unique functions that are distinct from AFAP1. While AFAP1 has the ability to bind to the SH3 domain of the nonreceptor tyrosine kinase cSrc via an N-terminal SH3 binding motif, it was unable to bind cortactin. However, the SH3 binding motif of AFAP1L1 was more efficient at interacting with the SH3 domain of cortactin and not cSrc. AFAP1L1 was shown by fluorescence microscopy to decorate actin filaments and move to punctate actin structures and colocalize with cortactin, consistent with localization to invadosomes. Upon overexpression in A7r5 cells, AFAP1L1 had the ability to induce podosome formation and move to podosomes without stimulation. Immunohistochemical analysis of AFAP1L1 in human tissues shows differential expression when contrasted with AFAP1 with localization of AFAP1L1 to unique sites in muscle and the dentate nucleus of the brain where AFAP1 was not detectable. We hypothesize AFAP1L1 may play a similar role to AFAP1 in affecting changes in actin filaments and bridging interactions with binding partners, but we hypothesize that AFAP1L1 may forge unique protein interactions in which AFAP1 is less efficient, and these interactions may allow AFAP1L1 to affect invadosome formation.
Collapse
Affiliation(s)
- Brandi N Snyder
- The Mary Babb Randolph Cancer Center and the Department of Cancer Cell Biology, West Virginia University, Morgantown, WV 26505, USA
| | | | | | | | | | | |
Collapse
|
40
|
Han B, Xiao H, Xu J, Lodyga M, Bai XH, Jin T, Liu M. Actin filament associated protein mediates c-Src related SRE/AP-1 transcriptional activation. FEBS Lett 2011; 585:471-7. [PMID: 21236256 DOI: 10.1016/j.febslet.2011.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Revised: 12/26/2010] [Accepted: 01/03/2011] [Indexed: 10/18/2022]
Abstract
AFAP is an adaptor protein involved in cytoskeletal organization and intracellular signaling. AFAP binds and activates c-Src; however, the downstream signals of this interaction remain unknown. Here we show that co-expression of AFAP and c-Src induce transcriptional activation of SRE and AP-1 in a c-Src activity dependent fashion. Structural-functional studies suggest that the proline-rich motif in the N-terminus of AFAP is critical for c-Src activation, and subsequent SRE/AP-1 transactivation and the actin-binding domain in the AFAP C-terminus is negatively involved in the regulation of AFAP/c-Src mediated SRE/AP-1 transactivation. Selective deletion of this domain enhances transactivation of SRE. We conclude that in addition to its role in the regulation of cytoskeletal structures, AFAP may also be involved in the c-Src related transcriptional activities.
Collapse
Affiliation(s)
- Bing Han
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
41
|
Ahluwalia M, de Groot J, Liu W(M, Gladson CL. Targeting SRC in glioblastoma tumors and brain metastases: rationale and preclinical studies. Cancer Lett 2010; 298:139-49. [PMID: 20947248 PMCID: PMC3212431 DOI: 10.1016/j.canlet.2010.08.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 08/22/2010] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is an extremely aggressive, infiltrative tumor with a poor prognosis. The regulatory approval of bevacizumab for recurrent GBM has confirmed that molecularly targeted agents have potential for GBM treatment. Preclinical data showing that SRC and SRC-family kinases (SFKs) mediate intracellular signaling pathways controlling key biologic/oncogenic processes provide a strong rationale for investigating SRC/SFK inhibitors, e.g., dasatinib, in GBM and clinical studies are underway. The activity of these agents against solid tumors suggests that they may also be useful in treating brain metastases. This article reviews the potential for using SRC/SFK inhibitors to treat GBM and brain metastases.
Collapse
Affiliation(s)
- Manmeet Ahluwalia
- Cleveland Clinic Main Campus, Mail Code ND40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-444-6145
| | - John de Groot
- The Brain Tumor Center, The University of Texas, M.D. Anderson Cancer Center, 1515, Holcombe Blvd., Unit 431, Houston, TX 77030, Phone: 713-792-7255
| | - Wei (Michael) Liu
- Lerner Research Institute, Department of Cancer Biology, Cleveland Clinic Mail Code NB40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-636-9494
| | - Candece L Gladson
- Lerner Research Institute, Department of Cancer Biology, Cleveland Clinic Mail Code NB40, 9500 Euclid Avenue, Cleveland, OH 44195, Phone: 216-636-9493, Fax: 216-445-6269
| |
Collapse
|
42
|
Lodyga M, Bai XH, Kapus A, Liu M. Adaptor protein XB130 is a Rac-controlled component of lamellipodia that regulates cell motility and invasion. J Cell Sci 2010; 123:4156-69. [DOI: 10.1242/jcs.071050] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
XB130 is a newly described cytosolic adaptor protein and tyrosine kinase substrate, involved in Src- and RET/PTC-dependent signaling. Although XB130 has been cloned as a homologue of actin-filament-associated protein (AFAP-110), its potential regulation by the actin skeleton and its putative roles in cytoskeleton regulation have not been addressed. Here, we show that XB130 (in contrast to AFAP-110) exhibited robust translocation to the cell periphery in response to various stimuli (including epidermal growth factor, wounding and expression of constitutively active Rac) that elicit lamellipodium formation. In stimulated cells, XB130 localized to the lamellipodial F-actin meshwork. Genetic and pharmacological data suggest that the key trigger for XB130 recruitment is the formation of the branched F-actin itself. Structure-function analysis revealed that both the XB130 N-terminus (167 amino acids) and C-terminus (63 amino acids) harbor crucial regions for its translocation to lamellipodia, whereas the PH domains and Src-targeted tyrosines are dispensable. Importantly, in TPC1 thyroid papillary carcinoma cells, silencing endogenous XB130 decreased the rate of wound closure, inhibited matrigel invasion, reduced lamellipodial persistence and slowed down spreading. Thus, XB130 is a novel Rac- and cytoskeleton-regulated and cytoskeleton-regulating adaptor protein that exhibits high affinity to lamellipodial (branched) F-actin and impacts motility and invasiveness of tumor cells.
Collapse
Affiliation(s)
- Monika Lodyga
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 1L7, Canada
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Ontario M5G 1X8, Canada
| | - Xiao-hui Bai
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 1L7, Canada
| | - András Kapus
- Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Ontario M5G 1X8, Canada
- Department of Surgery, University of Toronto, Ontario M5G 1X8, Canada
| | - Mingyao Liu
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 1L7, Canada
- Institute of Medical Sciences, University of Toronto, Ontario M5G 1X8, Canada
- Department of Surgery, University of Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
43
|
Kelley LC, Ammer AG, Hayes KE, Martin KH, Machida K, Jia L, Mayer BJ, Weed SA. Oncogenic Src requires a wild-type counterpart to regulate invadopodia maturation. J Cell Sci 2010; 123:3923-32. [PMID: 20980387 DOI: 10.1242/jcs.075200] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The proto-oncogene Src tyrosine kinase (Src) is overexpressed in human cancers and is currently a target of anti-invasive therapies. Activation of Src is an essential catalyst of invadopodia production. Invadopodia are cellular structures that mediate extracellular matrix (ECM) proteolysis, allowing invasive cell types to breach confining tissue barriers. Invadopodia assembly and maturation is a multistep process, first requiring the targeting of actin-associated proteins to form pre-invadopodia, which subsequently mature by recruitment and activation of matrix metalloproteases (MMPs) that facilitate ECM degradation. We demonstrate that active, oncogenic Src alleles require the presence of a wild-type counterpart to induce ECM degradation at invadopodia sites. In addition, we identify the phosphorylation of the invadopodia regulatory protein cortactin as an important mediator of invadopodia maturation downstream of wild-type Src. Distinct phosphotyrosine-based protein-binding profiles in cells forming pre-invadopodia and mature invadopodia were identified by SH2-domain array analysis. These results indicate that although elevated Src kinase activity is required to target actin-associated proteins to pre-invadopodia, regulated Src activity is required for invadopodia maturation and matrix degradation activity. Our findings describe a previously unappreciated role for proto-oncogenic Src in enabling the invasive activity of constitutively active Src alleles.
Collapse
Affiliation(s)
- Laura C Kelley
- Department of Neurobiology and Anatomy, Program in Cancer Cell Biology, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia 26506-9300, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
The protein kinase C cascade regulates recruitment of matrix metalloprotease 9 to podosomes and its release and activation. Mol Cell Biol 2010; 30:5545-61. [PMID: 20937775 DOI: 10.1128/mcb.00382-10] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Podosomes are transient cell surface structures essential for degradation of extracellular matrix during cell invasion. Protein kinase C (PKC) is involved in the regulation of podosome formation; however, the roles of individual PKC isoforms in podosome formation and proteolytic function are largely unknown. Recently, we reported that PDBu, a PKC activator, induced podosome formation in normal human bronchial epithelial cells. Here, we demonstrate that phorbol-12,13-dibutyrate (PDBu)-induced podosome formation is mainly mediated through redistribution of conventional PKCs, especially PKCα, from the cytosol to the podosomes. Interestingly, although blocking atypical PKCζ did not affect PDBu-induced podosome formation, it significantly reduced matrix degradation at podosomes. Inhibition of PKCζ reduced recruitment of matrix metalloprotease 9 (MMP-9) to podosomes and its release and activation. Downregulation of MMP-9 by small interfering RNA (siRNA) or neutralization antibody also significantly reduced matrix degradation. The regulatory effects of PKCζ on matrix degradation and recruitment of MMP-9 to podosomes were PKCζ kinase activity dependent. PDBu-induced recruitment of PKCζ and MMP-9 to podosomes was blocked by inhibition of novel PKC with rottlerin or PKCδ siRNA. Our data suggest that multiple PKC isozymes form a signaling cascade that controls podosome formation and dynamics and MMP-9 recruitment, release, and activation in a coordinated fashion.
Collapse
|
45
|
A Polymorphic Variant of AFAP-110 Enhances cSrc Activity. Transl Oncol 2010; 3:276-85. [PMID: 20689769 DOI: 10.1593/tlo.10106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 03/18/2010] [Accepted: 03/31/2010] [Indexed: 12/27/2022] Open
Abstract
Enhanced expression and activity of cSrc are associated with ovarian cancer progression. Generally, cSrc does not contain activating mutations; rather, its activity is increased in response to signals that affect a conformational change that releases its autoinhibition. In this report, we analyzed ovarian cancer tissues for the expression of a cSrc-activating protein, AFAP-110. AFAP-110 activates cSrc through a direct interaction that releases it from its autoinhibited conformation. Immunohistochemical analysis revealed a concomitant increase of AFAP-110 and cSrc in ovarian cancer tissues. An analysis of the AFAP-110 coding sequence revealed the presence of a nonsynonymous, single-nucleotide polymorphism that resulted in a change of Ser403 to Cys403. In cells that express enhanced levels of cSrc, AFAP-110(403C) directed the activation of cSrc and the formation of podosomes independently of input signals, in contrast to wild-type AFAP-110. We therefore propose that, under conditions of cSrc overexpression, the polymorphic variant of AFAP-110 promotes cSrc activation. Further, these data indicate amechanismby which an inherited genetic variation could influence ovarian cancer progression and could be used to predict the response to targeted therapy.
Collapse
|
46
|
Abstract
BACKGROUND INFORMATION Fluoride is a well-known G-protein activator. Exposure of cultured cells to its derivatives results in actin cytoskeleton remodelling. Podosomes are actin-based structures endowed with adhesion and matrix-degradation functions. This study investigates actin cytoskeleton reorganization induced by fluoride in endothelial cells. RESULTS Treatment of cultured endothelial cells with sodium fluoride (NaF) results in a rapid and potent stimulation of podosome formation. Furthermore, we show that Cdc42 (cell-division cycle 42), Rac1 and RhoA activities are stimulated in NaF-treated cells. However, podosome assembly is dependent on Cdc42 and Rac1, but not RhoA. Although the sole activation of Cdc42 is sufficient to induce individual podosomes, a balance between RhoGTPase activities regulates podosome formation in response to NaF, which in this case are often found in groups or rosettes. As in other models, podosome formation in endothelial cells exposed to NaF also involves Src. Finally, we demonstrate that NaF-induced podosomes are fully competent for matrix protein degradation. CONCLUSIONS Taken together, our findings establish NaF as a novel inducer of podosomes in endothelial cells in vitro.
Collapse
|
47
|
Tenan M, Aurrand-Lions M, Widmer V, Alimenti A, Burkhardt K, Lazeyras F, Belkouch MC, Hammel P, Walker PR, Duchosal MA, Imhof BA, Dietrich PY. Cooperative expression of junctional adhesion molecule-C and -B supports growth and invasion of glioma. Glia 2010; 58:524-37. [PMID: 19795504 DOI: 10.1002/glia.20941] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Brain invasion is a biological hallmark of glioma that contributes to its aggressiveness and limits the potential of surgery and irradiation. Deregulated expression of adhesion molecules on glioma cells is thought to contribute to this process. Junctional adhesion molecules (JAMs) include several IgSF members involved in leukocyte trafficking, angiogenesis, and cell polarity. They are expressed mainly by endothelial cells, white blood cells, and platelets. Here, we report JAM-C expression by human gliomas, but not by their normal cellular counterpart. This expression correlates with the expression of genes involved in cytoskeleton remodeling and cell migration. These genes, identified by a transcriptomic approach, include poliovirus receptor and cystein-rich 61, both known to promote glioma invasion, as well as actin filament associated protein, a c-Src binding partner. Gliomas also aberrantly express JAM-B, a high affinity JAM-C ligand. Their interaction activates the c-Src proto-oncogene, a central upstream molecule in the pathways regulating cell migration and invasion. In the tumor microenvironment, this co-expression may thus promote glioma invasion through paracrine stimuli from both tumor cells and endothelial cells. Accordingly, JAM-C/B blocking antibodies impair in vivo glioma growth and invasion, highlighting the potential of JAM-C and JAM-B as new targets for the treatment of human gliomas.
Collapse
Affiliation(s)
- Mirna Tenan
- Service of Oncology, Laboratory of Tumor Immunology, Geneva University Hospitals and University of Geneva, 1211 Geneva 14, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Quintavalle M, Elia L, Condorelli G, Courtneidge SA. MicroRNA control of podosome formation in vascular smooth muscle cells in vivo and in vitro. ACTA ACUST UNITED AC 2010; 189:13-22. [PMID: 20351064 PMCID: PMC2854384 DOI: 10.1083/jcb.200912096] [Citation(s) in RCA: 171] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PDGF enhances podosome formation and cell migration by regulating expression of the microRNAs miR-143 and -145, which target PDGF-R, PKC-ε, and fascin. Smooth muscle cell (SMC) plasticity plays an important role during development and in vascular pathologies such as atherosclerosis and restenosis. It was recently shown that down-regulation of microRNA (miR)-143 and -145, which are coexpressed from a single promoter, regulates the switch from contractile to synthetic phenotype, allowing SMCs to migrate and proliferate. We show in this study that loss of miR-143/145 in vitro and in vivo results in the formation of podosomes, which are actin-rich membrane protrusions involved in the migration of several cell types, including SMCs. We further show that platelet-derived growth factor (PDGF) mediates podosome formation in SMCs through the regulation of miR-143/145 expression via a pathway involving Src and p53. Moreover, we identify key podosome regulators as targets of miR-143 (PDGF receptor α and protein kinase C ε) and miR-145 (fascin). Thus, dysregulation of the miR-143 and -145 genes is causally involved in the aberrant SMC plasticity encountered during vascular disease, in part through the up-regulation of an autoregulatory loop that promotes podosome formation.
Collapse
|
49
|
Albiges-Rizo C, Destaing O, Fourcade B, Planus E, Block MR. Actin machinery and mechanosensitivity in invadopodia, podosomes and focal adhesions. J Cell Sci 2009; 122:3037-49. [PMID: 19692590 DOI: 10.1242/jcs.052704] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The invasiveness of cells is correlated with the presence of dynamic actin-rich membrane structures called invadopodia, which are membrane protrusions that are associated with localized polymerization of sub-membrane actin filaments. Similar to focal adhesions and podosomes, invadopodia are cell-matrix adhesion sites. Indeed, invadopodia share several features with podosomes, but whether they are distinct structures is still a matter of debate. Invadopodia are built upon an N-WASP-dependent branched actin network, and the Rho GTPase Cdc42 is involved in inducing invadopodial-membrane protrusion, which is mediated by actin filaments that are organized in bundles to form an actin core. Actin-core formation is thought to be an early step in invadopodium assembly, and the actin core is perpendicular to the extracellular matrix and the plasma membrane; this contrasts with the tangential orientation of actin stress fibers anchored to focal adhesions. In this Commentary, we attempt to summarize recent insights into the actin dynamics of invadopodia and podosomes, and the forces that are transmitted through these invasive structures. Although the mechanisms underlying force-dependent regulation of invadopodia and podosomes are largely unknown compared with those of focal adhesions, these structures do exhibit mechanosensitivity. Actin dynamics and associated forces might be key elements in discriminating between invadopodia, podosomes and focal adhesions. Targeting actin-regulatory molecules that specifically promote invadopodium formation is an attractive strategy against cancer-cell invasion.
Collapse
Affiliation(s)
- Corinne Albiges-Rizo
- INSERM U823 Institut Albert Bonniot, Université Joseph Fourier, CNRS ERL3148, Equipe DySAD, Site Santé, BP 170, Grenoble, France.
| | | | | | | | | |
Collapse
|
50
|
Crimaldi L, Courtneidge SA, Gimona M. Tks5 recruits AFAP-110, p190RhoGAP, and cortactin for podosome formation. Exp Cell Res 2009; 315:2581-92. [PMID: 19540230 DOI: 10.1016/j.yexcr.2009.06.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 05/29/2009] [Accepted: 06/14/2009] [Indexed: 10/20/2022]
Abstract
Podosome formation in vascular smooth muscle cells is characterized by the recruitment of AFAP-110, p190RhoGAP, and cortactin, which have specific roles in Src activation, local down-regulation of RhoA activity, and actin polymerization, respectively. However, the molecular mechanism that underlies their specific recruitment to podosomes remains unknown. The scaffold protein Tks5 is localized to podosomes in Src-transformed fibroblasts and in smooth muscle cells, and may serve as a specific recruiting adapter for various components during podosome formation. We show here that induced mislocalization of Tks5 to the surface of mitochondria leads to a major subcellular redistribution of AFAP-110, p190RhoGAP, and cortactin, and to inhibition of podosome formation. Analysis of a series of similarly mistargeted deletion mutants of Tks5 indicates that the fifth SH3 domain is essential for this recruitment. A Tks5 mutant lacking the PX domain also inhibits podosome formation and induces the redistribution of AFAP-110, p190RhoGAP, and cortactin to the perinuclear area. By expressing a catalytically inactive point mutant and by siRNA-mediated expression knock-down we also provide evidence that p190RhoGAP is required for podosome formation. Together our findings demonstrate that Tks5 plays a central role in the recruitment of AFAP-110, p190RhoGAP, and cortactin to drive podosome formation.
Collapse
Affiliation(s)
- Luca Crimaldi
- Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Via Nazionale, Santa Maria, Imbaro, Chieti, Italy.
| | | | | |
Collapse
|