1
|
Matskova L, Zheng S, Kashuba E, Ernberg I, Aspenström P. MTSS1: beyond the integration of actin and membrane dynamics. Cell Mol Life Sci 2024; 81:472. [PMID: 39625546 PMCID: PMC11615175 DOI: 10.1007/s00018-024-05511-w] [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: 08/24/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 12/06/2024]
Abstract
MTSS1 is a ubiquitously expressed intracellular protein known mainly for its involvement in basic cellular processes, such as the regulation of actin organization and membrane architecture. MTSS1 has attracted much attention for its role as a tumor suppressor, being absent or expressed at reduced levels in advanced and metastasizing cancers. Occasionally, MTSS1 is, instead, upregulated in metastasis and, in some cases, even in primary tumors. In addition to these well-established functions of MTSS1 linked to its I-BAR- and WH2-domains, the protein is involved in modulating cell-cell contacts, cell differentiation, lipid metabolism, and vesicle formation and acts as a scaffolding protein for several E3 ubiquitin ligases. MTSS1 is classified as a housekeeping protein and is never mutated despite the several pathologic phenotypes linked to its dysregulation. Despite MTSS1's involvement in fundamental signaling pathways, MTSS1 gene ablation is not ubiquitously lethal, although it affects embryonic development. Due to MTSS1´s involvement in many seemingly disparate processes, with many cases lacking mechanistic explanations, we found it timely to review the recent data on MTSS1's role at the cellular level, as well as in health and disease, to direct further studies on this interesting multifunctional protein.
Collapse
Affiliation(s)
- Liudmila Matskova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, FE 280, 17177, Sweden
| | - Shixing Zheng
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Elena Kashuba
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, FE 280, 17177, Sweden
- RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, FE 280, 17177, Sweden.
| | - Pontus Aspenström
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 75185, Sweden.
| |
Collapse
|
2
|
Podieh F, Overboom MC, Knol JC, Piersma SR, Goeij-de Haas R, Pham TV, Jimenez CR, Hordijk PL. AAMP and MTSS1 Are Novel Negative Regulators of Endothelial Barrier Function Identified in a Proteomics Screen. Cells 2024; 13:1609. [PMID: 39404373 PMCID: PMC11476176 DOI: 10.3390/cells13191609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Cell-cell adhesion in endothelial monolayers is tightly controlled and crucial for vascular integrity. Recently, we reported on the importance of fast protein turnover for maintenance of endothelial barrier function. Specifically, continuous ubiquitination and degradation of the Rho GTPase RhoB is crucial to preserve quiescent endothelial integrity. Here, we sought to identify other barrier regulators, which are characterized by a short half-life, using a proteomics approach. Following short-term inhibition of ubiquitination with E1 ligase inhibitor MLN7243 or Cullin E3 ligase inhibitor MLN4924 in primary human endothelial cells, we identified sixty significantly differentially expressed proteins. Intriguingly, our data showed that AAMP and MTSS1 are novel negative regulators of endothelial barrier function and that their turnover is tightly controlled by ubiquitination. Mechanistically, AAMP regulates the stability and activity of RhoA and RhoB, and colocalizes with F-actin and cortactin at membrane ruffles, possibly regulating F-actin dynamics. Taken together, these findings demonstrate the critical role of protein turnover of specific proteins in the regulation of endothelial barrier function, contributing to our options to target dysregulation of vascular permeability.
Collapse
Affiliation(s)
- Fabienne Podieh
- Department of Physiology, Microcirculation, Amsterdam Cardiovascular Science, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (F.P.)
| | - Max C. Overboom
- Department of Physiology, Microcirculation, Amsterdam Cardiovascular Science, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (F.P.)
| | - Jaco C. Knol
- Department of Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands (C.R.J.)
| | - Sander R. Piersma
- Department of Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands (C.R.J.)
| | - Richard Goeij-de Haas
- Department of Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands (C.R.J.)
| | - Thang V. Pham
- Department of Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands (C.R.J.)
| | - Connie R. Jimenez
- Department of Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands (C.R.J.)
| | - Peter L. Hordijk
- Department of Physiology, Microcirculation, Amsterdam Cardiovascular Science, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (F.P.)
| |
Collapse
|
3
|
Kim N, Li Y, Yu R, Kwon HS, Song A, Jun MH, Jeong JY, Lee JH, Lim HH, Kim MJ, Kim JW, Oh WJ. Repulsive Sema3E-Plexin-D1 signaling coordinates both axonal extension and steering via activating an autoregulatory factor, Mtss1. eLife 2024; 13:e96891. [PMID: 38526535 PMCID: PMC11001299 DOI: 10.7554/elife.96891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024] Open
Abstract
Axon guidance molecules are critical for neuronal pathfinding because they regulate directionality and growth pace during nervous system development. However, the molecular mechanisms coordinating proper axonal extension and turning are poorly understood. Here, metastasis suppressor 1 (Mtss1), a membrane protrusion protein, ensured axonal extension while sensitizing axons to the Semaphorin 3E (Sema3E)-Plexin-D1 repulsive cue. Sema3E-Plexin-D1 signaling enhanced Mtss1 expression in projecting striatonigral neurons. Mtss1 localized to the neurite axonal side and regulated neurite outgrowth in cultured neurons. Mtss1 also aided Plexin-D1 trafficking to the growth cone, where it signaled a repulsive cue to Sema3E. Mtss1 ablation reduced neurite extension and growth cone collapse in cultured neurons. Mtss1-knockout mice exhibited fewer striatonigral projections and irregular axonal routes, and these defects were recapitulated in Plxnd1- or Sema3e-knockout mice. These findings demonstrate that repulsive axon guidance activates an exquisite autoregulatory program coordinating both axonal extension and steering during neuronal pathfinding.
Collapse
Affiliation(s)
- Namsuk Kim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Yan Li
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Ri Yu
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyo-Shin Kwon
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Anji Song
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Hee Jun
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Jin-Young Jeong
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and TechnologyDaeguRepublic of Korea
| | - Ji Hyun Lee
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| | - Mi-Jin Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Jung-Woong Kim
- Department of Life Sciences, Chung-Ang UniversitySeoulRepublic of Korea
| | - Won-Jong Oh
- Neurovascular Unit Research Group, Korea Brain Research InstituteDaeguRepublic of Korea
| |
Collapse
|
4
|
Biton T, Scher N, Carmon S, Elbaz-Alon Y, Schejter ED, Shilo BZ, Avinoam O. Fusion pore dynamics of large secretory vesicles define a distinct mechanism of exocytosis. J Cell Biol 2023; 222:e202302112. [PMID: 37707500 PMCID: PMC10501449 DOI: 10.1083/jcb.202302112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/06/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
Exocrine cells utilize large secretory vesicles (LSVs) up to 10 μm in diameter. LSVs fuse with the apical surface, often recruiting actomyosin to extrude their content through dynamic fusion pores. The molecular mechanism regulating pore dynamics remains largely uncharacterized. We observe that the fusion pores of LSVs in the Drosophila larval salivary glands expand, stabilize, and constrict. Arp2/3 is essential for pore expansion and stabilization, while myosin II is essential for pore constriction. We identify several Bin-Amphiphysin-Rvs (BAR) homology domain proteins that regulate fusion pore expansion and stabilization. We show that the I-BAR protein Missing-in-Metastasis (MIM) localizes to the fusion site and is essential for pore expansion and stabilization. The MIM I-BAR domain is essential but not sufficient for localization and function. We conclude that MIM acts in concert with actin, myosin II, and additional BAR-domain proteins to control fusion pore dynamics, mediating a distinct mode of exocytosis, which facilitates actomyosin-dependent content release that maintains apical membrane homeostasis during secretion.
Collapse
Affiliation(s)
- Tom Biton
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Scher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shari Carmon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Elbaz-Alon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D. Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
5
|
Yang C, Chen F, Li S, Zeng X, Wang S, Lan J. Association of rs35006907 Polymorphism with Risk of Dilated Cardiomyopathy in Han Chinese Population. Balkan J Med Genet 2023; 26:27-34. [PMID: 38711908 PMCID: PMC11071056 DOI: 10.2478/bjmg-2023-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024] Open
Abstract
Background Several investigations have demonstrated the association of MTSS1 with left ventricular (LV) structure and function. A recently published study has even revealed that rs35006907 was associated with both MTSS1 expression and the risk of dilated cardiomyopathy (DCM). Objective Our study intended to investigate the relationship between rs35006907 and the risk of DCM in the Han Chinese population. Methods A total of 529 DCM and 600 healthy controls were recruited. We conducted genotyping for rs35006907 in all participants. Gene association studies were performed to assess the association between rs35006907 and the risk of DCM. A series of functional assays including western blot, realtime PCR and firefly luciferase reporter gene assays were conducted to illuminate the underlying mechanism. Results We found that rs35006907-A allele was significantly associated with reduced risk of DCM in additive (p= 0.004; OR=0.78; 95% CI=0.66-0.93) and recessive models (p= 0.0005; OR=0.56; 95%CI=0.41-0.78) when compared with the rs35006907-C allele. There were significant differences in the left ventricular end-diastolic diameter (LVEDD) and left ventricular ejection fraction (LVEF) between rs35006907-CC/AC and AA genotypes. Furthermore, the variant rs35006907-A allele presented lower reporter gene activity, reduced mRNA and protein expression levels when compared with the C allele. Conclusions Our findings demonstrated that rs35006907-C allele increased the risk of DCM in Han Chinese population. Besides, rs35006907-C displayed higher reporter gene activity and increased MTSS1 expression in human samples.
Collapse
Affiliation(s)
- C Yang
- Division of Cardiology, Panzhihua Central Hospital, Panzhihua, China
| | - F Chen
- Department of Hematology, Panzhihua Central Hospital, Panzhihua, China
| | - Sh Li
- Division of Cardiology, Panzhihua Central Hospital, Panzhihua, China
| | - X Zeng
- Division of Cardiology, Panzhihua Central Hospital, Panzhihua, China
| | - Sh Wang
- Division of Cardiology, Panzhihua Central Hospital, Panzhihua, China
| | - J Lan
- Division of Cardiology, Panzhihua Central Hospital, Panzhihua, China
| |
Collapse
|
6
|
Parker SS, Ly KT, Grant AD, Sweetland J, Wang AM, Parker JD, Roman MR, Saboda K, Roe DJ, Padi M, Wolgemuth CW, Langlais P, Mouneimne G. EVL and MIM/MTSS1 regulate actin cytoskeletal remodeling to promote dendritic filopodia in neurons. J Cell Biol 2023; 222:e202106081. [PMID: 36828364 PMCID: PMC9998662 DOI: 10.1083/jcb.202106081] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/22/2022] [Accepted: 01/20/2023] [Indexed: 02/25/2023] Open
Abstract
Dendritic spines are the postsynaptic compartment of a neuronal synapse and are critical for synaptic connectivity and plasticity. A developmental precursor to dendritic spines, dendritic filopodia (DF), facilitate synapse formation by sampling the environment for suitable axon partners during neurodevelopment and learning. Despite the significance of the actin cytoskeleton in driving these dynamic protrusions, the actin elongation factors involved are not well characterized. We identified the Ena/VASP protein EVL as uniquely required for the morphogenesis and dynamics of DF. Using a combination of genetic and optogenetic manipulations, we demonstrated that EVL promotes protrusive motility through membrane-direct actin polymerization at DF tips. EVL forms a complex at nascent protrusions and DF tips with MIM/MTSS1, an I-BAR protein important for the initiation of DF. We proposed a model in which EVL cooperates with MIM to coalesce and elongate branched actin filaments, establishing the dynamic lamellipodia-like architecture of DF.
Collapse
Affiliation(s)
- Sara S. Parker
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Kenneth Tran Ly
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Adam D. Grant
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Jillian Sweetland
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Ashley M. Wang
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - James D. Parker
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Mackenzie R. Roman
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Kathylynn Saboda
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Denise J. Roe
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Megha Padi
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
- Department of Molecular and Cellular Biology, College of Science, University of Arizona, Tucson, AZ, USA
| | - Charles W. Wolgemuth
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
- Department of Molecular and Cellular Biology, College of Science, University of Arizona, Tucson, AZ, USA
- Department of Physics, College of Science, University of Arizona, Tucson, AZ, USA
- Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, USA
| | - Paul Langlais
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Ghassan Mouneimne
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
- Cancer Biology Program, University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
7
|
Wang Y, Jia Z, Liang C, He Y, Cong M, Wu Q, Tian P, He D, Miao X, Sun B, Yin Y, Peng C, Yao F, Fu D, Liang Y, Zhang P, Xiong H, Hu G. MTSS1 curtails lung adenocarcinoma immune evasion by promoting AIP4-mediated PD-L1 monoubiquitination and lysosomal degradation. Cell Discov 2023; 9:20. [PMID: 36810288 PMCID: PMC9944270 DOI: 10.1038/s41421-022-00507-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/07/2022] [Indexed: 02/23/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy targeting PD-1/PD-L1 has shown durable clinical benefits in lung cancer. However, many patients respond poorly to ICB treatment, underscoring an incomplete understanding of PD-L1 regulation and therapy resistance. Here, we find that MTSS1 is downregulated in lung adenocarcinoma, leading to PD-L1 upregulation, impairment of CD8+ lymphocyte function, and enhanced tumor progression. MTSS1 downregulation correlates with improved ICB efficacy in patients. Mechanistically, MTSS1 interacts with the E3 ligase AIP4 for PD-L1 monoubiquitination at Lysine 263, leading to PD-L1 endocytic sorting and lysosomal degradation. In addition, EGFR-KRAS signaling in lung adenocarcinoma suppresses MTSS1 and upregulates PD-L1. More importantly, combining AIP4-targeting via the clinical antidepressant drug clomipramine and ICB treatment improves therapy response and effectively suppresses the growth of ICB-resistant tumors in immunocompetent mice and humanized mice. Overall, our study discovers an MTSS1-AIP4 axis for PD-L1 monoubiquitination and reveals a potential combinatory therapy with antidepressants and ICB.
Collapse
Affiliation(s)
- Yuan Wang
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenchang Jia
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chenxi Liang
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yunfei He
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Min Cong
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiuyao Wu
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pu Tian
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dasa He
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiang Miao
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Beibei Sun
- grid.16821.3c0000 0004 0368 8293Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yue Yin
- grid.9227.e0000000119573309National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Chao Peng
- grid.9227.e0000000119573309National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Feng Yao
- grid.16821.3c0000 0004 0368 8293Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Da Fu
- grid.412538.90000 0004 0527 0050Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293General Surgery, Ruijin Hospital & Institute of Pancreatic Diseases, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yajun Liang
- grid.410726.60000 0004 1797 8419Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peiyuan Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Hua Xiong
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
8
|
Pokrant T, Hein JI, Körber S, Disanza A, Pich A, Scita G, Rottner K, Faix J. Ena/VASP clustering at microspike tips involves lamellipodin but not I-BAR proteins, and absolutely requires unconventional myosin-X. Proc Natl Acad Sci U S A 2023. [PMID: 36598940 DOI: 10.1101/2022.05.12.491613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Sheet-like membrane protrusions at the leading edge, termed lamellipodia, drive 2D-cell migration using active actin polymerization. Microspikes comprise actin-filament bundles embedded within lamellipodia, but the molecular mechanisms driving their formation and their potential functional relevance have remained elusive. Microspike formation requires the specific activity of clustered Ena/VASP proteins at their tips to enable processive actin assembly in the presence of capping protein, but the factors and mechanisms mediating Ena/VASP clustering are poorly understood. Systematic analyses of B16-F1 melanoma mutants lacking potential candidate proteins revealed that neither inverse BAR-domain proteins, nor lamellipodin or Abi is essential for clustering, although they differentially contribute to lamellipodial VASP accumulation. In contrast, unconventional myosin-X (MyoX) identified here as proximal to VASP was obligatory for Ena/VASP clustering and microspike formation. Interestingly, and despite the invariable distribution of other relevant marker proteins, the width of lamellipodia in MyoX-KO mutants was significantly reduced as compared with B16-F1 control, suggesting that microspikes contribute to lamellipodium stability. Consistently, MyoX removal caused marked defects in protrusion and random 2D-cell migration. Strikingly, Ena/VASP-deficiency also uncoupled MyoX cluster dynamics from actin assembly in lamellipodia, establishing their tight functional association in microspike formation.
Collapse
Affiliation(s)
- Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Ingo Hein
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Sarah Körber
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Andrea Disanza
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
| | - Andreas Pich
- Research Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Giorgio Scita
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, 20139 Milan, Italy
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
9
|
Moritz MNDO, Casali BC, Stotzer US, Karina dos Santos P, Selistre-de-Araujo HS. Alternagin-C, an alpha2beta1 integrin ligand, attenuates collagen-based adhesion, stimulating the metastasis suppressor 1 expression in triple-negative breast tumor cells. Toxicon 2022; 210:1-10. [DOI: 10.1016/j.toxicon.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/17/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022]
|
10
|
Liang L, Liang X, Jiang P, Zhou L, Zhong L, Wang M, Lin S, Guo Z, Yu J, Yang C, Chen Y, Zhuo C, Chen P, Wang Y. Metastasis suppressor 1 interacts with α-actinin 4 to affect its localization and regulate formation of membrane ruffling. Cytoskeleton (Hoboken) 2021; 78:337-348. [PMID: 34435464 DOI: 10.1002/cm.21686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 01/21/2023]
Abstract
Membrane ruffling plays an important role in the directed cell migration and escape of tumor cells from the monolayer. Metastasis suppressor 1 (MTSS1), also known as missing in metastasis, has been implicated in cell morphology, motility, metastasis, and development. Here, the dynamic interaction proteins associated with MTSS1 and involved in membrane ruffling were determined by cross-linking and mass spectrometry analysis. We identified α-actinin 4 (ACTN4) as an interacting protein and confirmed a direct interaction between MTSS1 and ACTN4. Moreover, co-expression of MTSS1 in fibroblasts recruited cytoplasmic ACTN4 to the cell periphery, at which point ruffling became thick and rigid. In MCF-7 cells, MTSS1 knockdown did not show an obvious effect on the cell shape or the distribution of endogenous ACTN4; however, ACTN4 overexpression transformed cell morphology from an epidermal- to a fibroblast-like shape, and further MTSS1 depletion significantly increased the ratio of fibroblast cells exhibiting prominent ruffling. Furthermore, biochemical data suggested that MTSS1 cross-linking with ACTN4 induced the formation of actin fiber bundles into more organized structures in vitro. These data indicated that MTSS1 might recruit cytoplasmic ACTN4 to the cell periphery and regulate cytoskeleton dynamics to restrict its performance in membrane ruffling.
Collapse
Affiliation(s)
- Lijun Liang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaoping Liang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Peng Jiang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Lu Zhou
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Luanluan Zhong
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Mei Wang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Shuyun Lin
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zhen Guo
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Juan Yu
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Changcheng Yang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yu Chen
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Chengjie Zhuo
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ping Chen
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ying Wang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
11
|
Brücker L, Kretschmer V, May-Simera HL. The entangled relationship between cilia and actin. Int J Biochem Cell Biol 2020; 129:105877. [PMID: 33166678 DOI: 10.1016/j.biocel.2020.105877] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Primary cilia are microtubule-based sensory cell organelles that are vital for tissue and organ development. They act as an antenna, receiving and transducing signals, enabling communication between cells. Defects in ciliogenesis result in severe genetic disorders collectively termed ciliopathies. In recent years, the importance of the direct and indirect involvement of actin regulators in ciliogenesis came into focus as it was shown that F-actin polymerisation impacts ciliation. The ciliary basal body was further identified as both a microtubule and actin organising centre. In the current review, we summarize recent studies on F-actin in and around primary cilia, focusing on different actin regulators and their effect on ciliogenesis, from the initial steps of basal body positioning and regulation of ciliary assembly and disassembly. Since primary cilia are also involved in several intracellular signalling pathways such as planar cell polarity (PCP), subsequently affecting actin rearrangements, the multiple effectors of this pathway are highlighted in more detail with a focus on the feedback loops connecting actin networks and cilia proteins. Finally, we elucidate the role of actin regulators in the development of ciliopathy symptoms and cancer.
Collapse
Affiliation(s)
- Lena Brücker
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany
| | - Viola Kretschmer
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany
| | - Helen Louise May-Simera
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany.
| |
Collapse
|
12
|
Lian Y, Wen D, Meng X, Wang X, Li H, Hao L, Xue H, Zhao J. Inhibition of invadopodia formation by diosgenin in tumor cells. Oncol Lett 2020; 20:283. [PMID: 33014161 PMCID: PMC7520800 DOI: 10.3892/ol.2020.12148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Diosgenin is a type of steroid extracted from the rhizome of Dioscorea plants. In traditional Chinese medicine, Dioscorea has the effect of ‘eliminating phlegm, promoting digestion, relaxing tendons, promoting blood circulation and inhibiting malaria’. Recent studies have confirmed that diosgenin exhibits a number of pharmacological effects, including antitumor activities. Through its antitumor effect, diosgenin is able to block tumor progression and increase the survival rate of patients with cancer; ultimately improving their quality of life. However, the mechanism underlying its pharmacological action remains unclear. Once tumor cells reach a metastatic phase, it can be fatal. Increased migration and invasiveness are the hallmarks of metastatic tumor cells. Invadopodia formation is key to maintaining the high migration and invasive ability of tumor cells. Invadopodia are a type of membrane structure process rich in filamentous-actin and are common in highly invasive tumor cells. In addition to actin, numerous actin regulators, including cortical actin-binding protein (Cortactin), accumulate in invadopodia. Cortactin is a microfilament actin-binding protein with special repetitive domains that are directly involved in the formation of the cortical microfilament actin cell skeleton. Cortactin is also one of the main substrates of intracellular Src-type tyrosine protein kinases and represents a highly conserved family of intracellular cortical signaling proteins. In recent years, great progress has been made in understanding the role of Cortactin and its molecular mechanism in cell motility. However, the diosgenin-Cortactin-invadopodia mechanism is still under investigation. Therefore, the present review focused on the current research on the regulation of invadopodia by diosgenin via Cortactin.
Collapse
Affiliation(s)
- Yaxin Lian
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dezhong Wen
- Department of Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaoting Meng
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaozhen Wang
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hongcheng Li
- GeneScience Pharmaceuticals Co., Ltd., Changchun, Jilin 130021, P.R. China
| | - Liming Hao
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hui Xue
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia Zhao
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
13
|
Zhang L, Wang X, Liu X, Lv M, Shen E, Zhu G, Sun Z. miR-28-5p targets MTSS1 to regulate cell proliferation and apoptosis in esophageal cancer. Acta Biochim Biophys Sin (Shanghai) 2020; 52:842-852. [PMID: 32645138 DOI: 10.1093/abbs/gmaa059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/10/2020] [Accepted: 03/31/2020] [Indexed: 01/10/2023] Open
Abstract
Esophageal cancer (EC) is one of the most common aggressive malignant diseases worldwide. miR-28-5p plays important regulatory roles in many cancers including human EC. However, the molecular mechanism and potential role of miR-28-5p in EC remain uncertain. In this study, qRT-PCR and western blot analysis revealed that miR-28-5p expression was up-regulated and metastasis suppressor-1 (MTSS1) was down-regulated in EC tissues relative to matched para-cancer tissues. Cell counting kit-8 (CCK-8) assay demonstrated that miR-28-5p mimics increased cell viability, and miR-28-5p inhibitor decreased it. Flow cytometry (FCM) assay indicated that miR-28-5p mimics promoted cell cycle entry, while miR-28-5p inhibitor reduced it and induced cell apoptosis. Moreover, miR-28-5p mimics up-regulated the expressions of cyclin A, cyclin dependent kinase 2 (CDK2), cyclin D1, and cyclin E but down-regulated the expressions of cleaved caspase-3 and cleaved caspase-9, which was abolished by miR-28-5p inhibitor. Furthermore, luciferase reporter assay verified that miR-28-5p directly targeted MTSS1 3'UTR and down-regulated its expression. MTSS1 overexpression in TE-1 cells inhibited cell proliferation and promoted apoptosis induced by miR-28-5p mimics, whereas silencing of MTSS1 reversed cell progression induced by miR-28-5p inhibitor. We also demonstrated that miR-28-5p could promote esophageal tumor formation in vivo. Hematoxylin-eosin staining, immunohistochemistry, and TUNEL assays confirmed that miR-28-5p antagomir inhibited cell growth and accelerated apoptosis. Our results suggest that miR-28-5p may induce cell proliferation and suppress apoptosis to promote EC tumor formation via decreasing MTSS1 expression. Thus, miR-28-5p may be a potential target for human EC therapy.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Xin Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Xin Liu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Mingyue Lv
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Erdong Shen
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Guolian Zhu
- Department of Oncology, Shenyang Fifth People’s Hospital, Shenyang 110001, China
| | - Zhe Sun
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
14
|
Sarapulov AV, Petrov P, Hernández-Pérez S, Šuštar V, Kuokkanen E, Cords L, Samuel RVM, Vainio M, Fritzsche M, Carrasco YR, Mattila PK. Missing-in-Metastasis/Metastasis Suppressor 1 Regulates B Cell Receptor Signaling, B Cell Metabolic Potential, and T Cell-Independent Immune Responses. Front Immunol 2020; 11:599. [PMID: 32373113 PMCID: PMC7176992 DOI: 10.3389/fimmu.2020.00599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/16/2020] [Indexed: 11/13/2022] Open
Abstract
Efficient generation of antibodies by B cells is one of the prerequisites of protective immunity. B cell activation by cognate antigens via B cell receptors (BCRs), or pathogen-associated molecules through pattern-recognition receptors, such as Toll-like receptors (TLRs), leads to transcriptional and metabolic changes that ultimately transform B cells into antibody-producing plasma cells or memory cells. BCR signaling and a number of steps downstream of it rely on coordinated action of cellular membranes and the actin cytoskeleton, tightly controlled by concerted action of multiple regulatory proteins, some of them exclusive to B cells. Here, we dissect the role of Missing-In-Metastasis (MIM), or Metastasis suppressor 1 (MTSS1), a cancer-associated membrane and actin cytoskeleton regulating protein, in B cell-mediated immunity by taking advantage of MIM knockout mouse strain. We show undisturbed B cell development and largely normal composition of B cell compartments in the periphery. Interestingly, we found that MIM-/- B cells are defected in BCR signaling in response to surface-bound antigens but, on the other hand, show increased metabolic activity after stimulation with LPS or CpG. In vivo, MIM knockout animals exhibit impaired IgM antibody responses to immunization with T cell-independent antigen. This study provides the first comprehensive characterization of MIM in B cells, demonstrates its regulatory role for B cell-mediated immunity, as well as proposes new functions for MIM in tuning receptor signaling and cellular metabolism, processes, which may also contribute to the poorly understood functions of MIM in cancer.
Collapse
Affiliation(s)
- Alexey V. Sarapulov
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Petar Petrov
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Sara Hernández-Pérez
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Vid Šuštar
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
| | - Elina Kuokkanen
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
| | - Lena Cords
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Rufus V. M. Samuel
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
| | - Marika Vainio
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Marco Fritzsche
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, United Kingdom
- Rosalind Franklin Institute, Didcot, United Kingdom
| | - Yolanda R. Carrasco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Pieta K. Mattila
- Institute of Biomedicine and MediCity Research Laboratories, University of Turku, Turku, Finland
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
15
|
Cong M, Wang Y, Yang Y, Lian C, Zhuang X, Li X, Zhang P, Liu Y, Tang J, Yang Q, Zhang X, Xiong H, Hu R, Hu G. MTSS1 suppresses mammary tumor-initiating cells by enhancing RBCK1-mediated p65 ubiquitination. NATURE CANCER 2020; 1:222-234. [PMID: 35122005 DOI: 10.1038/s43018-019-0021-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022]
Abstract
Tumor-initiating cells (TICs) are considered the culprits of cancer development and progression. Dysregulation of metastasis suppressor protein 1 (MTSS1) has been widely observed in tumor metastasis, but its functional contribution and mechanism in cancer is poorly understood. Here we report a role of MTSS1 in suppressing TICs in breast cancer. Mtss1 knockout (KO) enhances the mammary epithelial TIC subpopulation in both luminal and basal-like breast cancer mouse models. MTSS1 also suppresses tumorsphere formation in breast cancer cells. Mechanistically, MTSS1 interacts with the E3 ligase RanBP2-type and C3HC4-type zinc finger containing 1 (RBCK1) to facilitate RBCK1-mediated p65 ubiquitination and degradation, thus suppressing the NF-κB signaling pathway and tumorigenesis. In addition, actin beta-like 2 (ACTBL2) competes with RBCK1 for MTSS1 binding, leading to p65 stabilization. Importantly, MTSS1 silencing promotes patient-derived organoid formation and xenograft growth. MTSS1 downregulation in clinical tumors is also linked to worse prognosis. Overall our data reveal a new paradigm of NF-κB regulation and may have important implications in therapeutics targeting TICs.
Collapse
Affiliation(s)
- Min Cong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yang Yang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Shanghai, China
| | - Cheng Lian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xueqian Zhuang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoxun Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peiyuan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingjie Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Tang
- State Key Laboratory of Oncology in South China; Department of Breast Oncology, Sun Yat-Sen University, Guangzhou, China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Ji'nan, China
| | - Xue Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hua Xiong
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | - Ronggui Hu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Shanghai, China.
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
16
|
Petrov P, Sarapulov AV, Eöry L, Scielzo C, Scarfò L, Smith J, Burt DW, Mattila PK. Computational analysis of the evolutionarily conserved Missing In Metastasis/Metastasis Suppressor 1 gene predicts novel interactions, regulatory regions and transcriptional control. Sci Rep 2019; 9:4155. [PMID: 30858428 PMCID: PMC6411742 DOI: 10.1038/s41598-019-40697-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 02/21/2019] [Indexed: 12/25/2022] Open
Abstract
Missing in Metastasis (MIM), or Metastasis Suppressor 1 (MTSS1), is a highly conserved protein, which links the plasma membrane to the actin cytoskeleton. MIM has been implicated in various cancers, however, its modes of action remain largely enigmatic. Here, we performed an extensive in silico characterisation of MIM to gain better understanding of its function. We detected previously unappreciated functional motifs including adaptor protein (AP) complex interaction site and a C-helix, pointing to a role in endocytosis and regulation of actin dynamics, respectively. We also identified new functional regions, characterised with phosphorylation sites or distinct hydrophilic properties. Strong negative selection during evolution, yielding high conservation of MIM, has been combined with positive selection at key sites. Interestingly, our analysis of intra-molecular co-evolution revealed potential regulatory hotspots that coincided with reduced potentially pathogenic polymorphisms. We explored databases for the mutations and expression levels of MIM in cancer. Experimentally, we focused on chronic lymphocytic leukaemia (CLL), where MIM showed high overall expression, however, downregulation on poor prognosis samples. Finally, we propose strong conservation of MTSS1 also on the transcriptional level and predict novel transcriptional regulators. Our data highlight important targets for future studies on the role of MIM in different tissues and cancers.
Collapse
Affiliation(s)
- Petar Petrov
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.
| | - Alexey V Sarapulov
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Lel Eöry
- Division of Genetics and Genomics, The Roslin Institute and R(D)SVS, University of Edinburgh, Roslin, Easter Bush campus, Midlothian, EH25 9RG, United Kingdom
| | - Cristina Scielzo
- Unit of B Cell Neoplasia, Division of Molecular Oncology, IRCCS, San Raffaele Scientific Institute, Milano, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Lydia Scarfò
- Unit of B Cell Neoplasia, Division of Molecular Oncology, IRCCS, San Raffaele Scientific Institute, Milano, Italy.,Università Vita-Salute San Raffaele, Milan, Italy.,Strategic Research Program on CLL, Division of Experimental Oncology, IRCCS, San Raffaele Scientific Institute, Milano, Italy
| | - Jacqueline Smith
- Division of Genetics and Genomics, The Roslin Institute and R(D)SVS, University of Edinburgh, Roslin, Easter Bush campus, Midlothian, EH25 9RG, United Kingdom
| | - David W Burt
- University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Pieta K Mattila
- Institute of Biomedicine, and MediCity Research Laboratories, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.
| |
Collapse
|
17
|
MiR-423-5p in brain metastasis: potential role in diagnostics and molecular biology. Cell Death Dis 2018; 9:936. [PMID: 30224667 PMCID: PMC6141540 DOI: 10.1038/s41419-018-0955-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 01/05/2023]
Abstract
During the last several years, a growing number of studies have shown that microRNAs (miRNAs) participate in cancer metastasis. Brain metastasis (BM) is a frequent complication of lung adenocarcinoma (LAD), and the incidence of locally advanced LAD with BM can be as high as 30-50%. This study was performed to identify the miRNA expression patterns of LAD with BM and to determine the biological role that miRNAs play in tumorigenesis. To this end, we conducted microarray and quantitative PCR analyses to evaluate BM-related miRNAs independently validated from a total of 155 patients with LAD. A series of in vivo and in vitro assays were also conducted to verify the impact of miRNAs on BM. We found significantly increased expression of miR-423-5p, and BM was predicted in non-small cell lung cancer when compared to LAD without BM. We next examined the function of miR-423-5p and discovered that it significantly promoted colony formation, cell motility, migration, and invasion in vitro. We computationally and experimentally confirmed that metastasis suppressor 1 (MTSS1) was a direct miR-423-5p target. Through a combination of image, histological, and molecular analyses, we found that miR-423-5p overexpression significantly increased tumor burden, local invasion, and distant BM. The level of MTSS1 expression was inversely correlated with miR-423-5p upregulation in the LAD specimens and was associated with survival of patients with BM. MiR-423-5p promoted BM in LAD and inhibited MTSS1 expression. Together, these results show that MiR-423-5p has the potential to be a marker of BM and/or a therapeutic target in LAD.
Collapse
|
18
|
Ramos-García P, González-Moles MÁ, González-Ruiz L, Ayén Á, Ruiz-Ávila I, Navarro-Triviño FJ, Gil-Montoya JA. An update of knowledge on cortactin as a metastatic driver and potential therapeutic target in oral squamous cell carcinoma. Oral Dis 2018; 25:949-971. [PMID: 29878474 DOI: 10.1111/odi.12913] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/15/2018] [Accepted: 06/05/2018] [Indexed: 12/12/2022]
Abstract
Cortactin is a protein encoded by the CTTN gene, localized on chromosome band 11q13. As a result of the amplification of this band, an important event in oral carcinogenesis, CTTN is also usually amplified, promoting the frequent overexpression of cortactin. Cortactin enhances cell migration in oral cancer, playing a key role in the regulation of filamentous actin and of protrusive structures (invadopodia and lamellipodia) on the cell membrane that are necessary for the acquisition of a migratory phenotype. We also analyze a series of emerging functions that cortactin may exert in oral cancer (cell proliferation, angiogenesis, regulation of exosomes, and interactions with the tumor microenvironment). We review its molecular structure, its most important interactions (with Src, Arp2/3 complex, and SH3-binding partners), the regulation of its functions, and its specific oncogenic role in oral cancer. We explore the mechanisms of its overexpression in cancer, mainly related to genetic amplification. We analyze the prognostic implications of the oncogenic activation of cortactin in potentially malignant disorders and in head and neck cancer, where it appears to be relevant in the development of lymph node metastasis. Finally, we discuss its usefulness as a therapeutic target and suggest future research lines.
Collapse
Affiliation(s)
| | - Miguel Ángel González-Moles
- School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, Granada, Spain
| | - Lucía González-Ruiz
- Servicio de Dermatología, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Ángela Ayén
- School of Medicine, University of Granada, Granada, Spain
| | - Isabel Ruiz-Ávila
- Instituto de Investigación Biosanitaria, Granada, Spain.,Servicio de Anatomía Patológica, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | | | - José Antonio Gil-Montoya
- School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, Granada, Spain
| |
Collapse
|
19
|
Kawabata Galbraith K, Fujishima K, Mizuno H, Lee SJ, Uemura T, Sakimura K, Mishina M, Watanabe N, Kengaku M. MTSS1 Regulation of Actin-Nucleating Formin DAAM1 in Dendritic Filopodia Determines Final Dendritic Configuration of Purkinje Cells. Cell Rep 2018; 24:95-106.e9. [DOI: 10.1016/j.celrep.2018.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/01/2018] [Accepted: 06/01/2018] [Indexed: 10/28/2022] Open
|
20
|
Hong SW, Hur W, Choi JE, Kim JH, Hwang D, Yoon SK. Role of ADAM17 in invasion and migration of CD133-expressing liver cancer stem cells after irradiation. Oncotarget 2018; 7:23482-97. [PMID: 26993601 PMCID: PMC5029641 DOI: 10.18632/oncotarget.8112] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 02/28/2016] [Indexed: 12/29/2022] Open
Abstract
We investigated the biological role of CD133-expressing liver cancer stem cells (CSCs) enriched after irradiation of Huh7 cells in cell invasion and migration. We also explored whether a disintegrin and metalloproteinase-17 (ADAM17) influences the metastatic potential of CSC-enriched hepatocellular carcinoma (HCC) cells after irradiation. A CD133-expressing Huh7 cell subpopulation showed greater resistance to sublethal irradiation and specifically enhanced cell invasion and migration capabilities. We also demonstrated that the radiation-induced MMP-2 and MMP-9 enzyme activities as well as the secretion of vascular endothelial growth factor were increased more predominantly in Huh7CD133+ cell subpopulations than Huh7CD133− cell subpopulations. Furthermore, we showed that silencing ADAM17 significantly inhibited the migration and invasiveness of enriched Huh7CD133+ cells after irradiation; moreover, Notch signaling was significantly reduced in irradiated CD133-expressing liver CSCs following stable knockdown of the ADAM17 gene. In conclusion, our findings indicate that CD133-expressing liver CSCs have considerable metastatic capabilities after irradiation of HCC cells, and their metastatic capabilities might be maintained by ADAM17. Therefore, suppression of ADAM17 shows promise for improving the efficiency of current radiotherapies and reducing the metastatic potential of liver CSCs during HCC treatment.
Collapse
Affiliation(s)
- Sung Woo Hong
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Wonhee Hur
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Eun Choi
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung-Hee Kim
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Daehee Hwang
- Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, DGIST, Daegu, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
21
|
Hong SW, Hur W, Choi JE, Kim JH, Hwang D, Yoon SK. Role of ADAM17 in invasion and migration of CD133-expressing liver cancer stem cells after irradiation. Oncotarget 2018. [PMID: 26993601 DOI: 0.18632/oncotarget.8112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
We investigated the biological role of CD133-expressing liver cancer stem cells (CSCs) enriched after irradiation of Huh7 cells in cell invasion and migration. We also explored whether a disintegrin and metalloproteinase-17 (ADAM17) influences the metastatic potential of CSC-enriched hepatocellular carcinoma (HCC) cells after irradiation. A CD133-expressing Huh7 cell subpopulation showed greater resistance to sublethal irradiation and specifically enhanced cell invasion and migration capabilities. We also demonstrated that the radiation-induced MMP-2 and MMP-9 enzyme activities as well as the secretion of vascular endothelial growth factor were increased more predominantly in Huh7CD133+ cell subpopulations than Huh7CD133- cell subpopulations. Furthermore, we showed that silencing ADAM17 significantly inhibited the migration and invasiveness of enriched Huh7CD133+ cells after irradiation; moreover, Notch signaling was significantly reduced in irradiated CD133-expressing liver CSCs following stable knockdown of the ADAM17 gene. In conclusion, our findings indicate that CD133-expressing liver CSCs have considerable metastatic capabilities after irradiation of HCC cells, and their metastatic capabilities might be maintained by ADAM17. Therefore, suppression of ADAM17 shows promise for improving the efficiency of current radiotherapies and reducing the metastatic potential of liver CSCs during HCC treatment.
Collapse
Affiliation(s)
- Sung Woo Hong
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Wonhee Hur
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Eun Choi
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung-Hee Kim
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Daehee Hwang
- Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, DGIST, Daegu, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
22
|
Bracalente C, Ibañez IL, Berenstein A, Notcovich C, Cerda MB, Klamt F, Chernomoretz A, Durán H. Reprogramming human A375 amelanotic melanoma cells by catalase overexpression: Upregulation of antioxidant genes correlates with regression of melanoma malignancy and with malignant progression when downregulated. Oncotarget 2018; 7:41154-41171. [PMID: 27206673 PMCID: PMC5173049 DOI: 10.18632/oncotarget.9273] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/02/2016] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are implicated in tumor transformation. The antioxidant system (AOS) protects cells from ROS damage. However, it is also hijacked by cancers cells to proliferate within the tumor. Thus, identifying proteins altered by redox imbalance in cancer cells is an attractive prognostic and therapeutic tool. Gene expression microarrays in A375 melanoma cells with different ROS levels after overexpressing catalase were performed. Dissimilar phenotypes by differential compensation to hydrogen peroxide scavenging were generated. The melanotic A375-A7 (A7) upregulated TYRP1, CNTN1 and UCHL1 promoting melanogenesis. The metastatic A375-G10 (G10) downregulated MTSS1 and TIAM1, proteins absent in metastasis. Moreover, differential coexpression of AOS genes (EPHX2, GSTM3, MGST1, MSRA, TXNRD3, MGST3 and GSR) was found in A7 and G10. Their increase in A7 improved its AOS ability and therefore, oxidative stress response, resembling less aggressive tumor cells. Meanwhile, their decrease in G10 revealed a disruption in the AOS and therefore, enhanced its metastatic capacity. These gene signatures, not only bring new insights into the physiopathology of melanoma, but also could be relevant in clinical prognostic to classify between non aggressive and metastatic melanomas.
Collapse
Affiliation(s)
- Candelaria Bracalente
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina
| | - Irene L Ibañez
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina
| | - Ariel Berenstein
- Fundación Instituto Leloir and Departamento de Física, Facultad Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cintia Notcovich
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina
| | - María B Cerda
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina
| | - Fabio Klamt
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brasil
| | - Ariel Chernomoretz
- Fundación Instituto Leloir and Departamento de Física, Facultad Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Hebe Durán
- Departamento de Micro y Nanotecnología, Comisión Nacional de Energía Atómica, San Martín, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina.,Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, San Martín, Buenos Aires, Argentina
| |
Collapse
|
23
|
Abstract
Actin remodeling plays an essential role in diverse cellular processes such as cell motility, vesicle trafficking or cytokinesis. The scaffold protein and actin nucleation promoting factor Cortactin is present in virtually all actin-based structures, participating in the formation of branched actin networks. It has been involved in the control of endocytosis, and vesicle trafficking, axon guidance and organization, as well as adhesion, migration and invasion. To migrate and invade through three-dimensional environments, cells have developed specialized actin-based structures called invadosomes, a generic term to designate invadopodia and podosomes. Cortactin has emerged as a critical regulator of invadosome formation, function and disassembly. Underscoring this role, Cortactin is frequently overexpressed in several types of invasive cancers. Herein we will review the roles played by Cortactin in these specific invasive structures.
Collapse
Affiliation(s)
- Pauline Jeannot
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester , Manchester M20 4BX, UK
| | - Arnaud Besson
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,LBCMCP , Centre de Biologie Intégrative, Université de Toulouse , CNRS, UPS, Toulouse Cedex, France
| |
Collapse
|
24
|
Samsonraj RM, Dudakovic A, Manzar B, Sen B, Dietz AB, Cool SM, Rubin J, van Wijnen AJ. Osteogenic Stimulation of Human Adipose-Derived Mesenchymal Stem Cells Using a Fungal Metabolite That Suppresses the Polycomb Group Protein EZH2. Stem Cells Transl Med 2017; 7:197-209. [PMID: 29280310 PMCID: PMC5788881 DOI: 10.1002/sctm.17-0086] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/25/2017] [Indexed: 12/19/2022] Open
Abstract
Strategies for musculoskeletal tissue regeneration apply adult mesenchymal stem/stromal cells (MSCs) that can be sourced from bone marrow- and lipo-aspirates. Adipose tissue-derived MSCs are more easily harvested in the large quantities required for skeletal tissue-engineering approaches, but are generally considered to be less osteogenic than bone marrow MSCs. Therefore, we tested a new molecular strategy to improve their osteogenic lineage-differentiation potential using the fungal metabolite cytochalasin D (CytoD). We show that CytoD, which may function by redistributing the intracellular location of β-actin (ACTB), is a potent osteogenic stimulant as reflected by significant increases in alkaline phosphatase activity, extracellular matrix mineralization, and osteoblast-related gene expression (e.g., RUNX2, ALPL, SPARC, and TGFB3). RNA sequencing analyses of MSCs revealed that acute CytoD treatment (24 hours) stimulates a broad program of osteogenic biomarkers and epigenetic regulators. CytoD decreases mRNA and protein levels of the Polycomb chromatin regulator Enhancer of Zeste Homolog 2 (EZH2), which controls heterochromatin formation by mediating trimethylation of histone 3 lysine 27 (H3K27me3). Reduced EZH2 expression decreases cellular H3K27me3 marks indicating a global reduction in heterochromatin. We conclude that CytoD is an effective osteogenic stimulant that mechanistically functions by blocking both cytoplasmic actin polymerization and gene-suppressive epigenetic mechanisms required for the acquisition of the osteogenic phenotype in adipose tissue-derived MSCs. This finding supports the use of CytoD in advancing the osteogenic potential of MSCs in skeletal regenerative strategies. Stem Cells Translational Medicine 2018;7:197-209.
Collapse
Affiliation(s)
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Bushra Manzar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Buer Sen
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
25
|
Li L, Baxter SS, Gu N, Ji M, Zhan X. Missing-in-metastasis protein downregulates CXCR4 by promoting ubiquitylation and interaction with small Rab GTPases. J Cell Sci 2017; 130:1475-1485. [PMID: 28264927 DOI: 10.1242/jcs.198937] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/27/2017] [Indexed: 01/09/2023] Open
Abstract
Surface expression of chemokine receptor CXCR4 is downregulated by missing-in-metastasis protein (MIM; also known as MTSS1), a member of the inverse BAR (I-BAR)-domain protein family that recognizes and generates membranes with negative curvature. Yet, the mechanism for the regulation is unknown. Here, we show that MIM forms a complex with CXCR4 by binding to E3 ubiquitin ligase AIP4 (also known as ITCH) in response to stromal cell-derived factor 1 (SDF-1; also known as CXCL12). Overexpression of MIM promoted CXCR4 ubiquitylation, inhibited cellular response to SDF-1, caused accumulation and aggregation of multivesicular bodies (MVBs) in the cytoplasm, and promoted CXCR4 sorting into MVBs in a manner depending on binding to AIP4. In response to SDF-1, MIM also bound transiently to the small GTPase Rab5 at 5 min and to Rab7 at 30 min. Binding to Rab7 requires an N-terminal coiled-coil motif, deletion of which abolished MIM-mediated MVB formation and CXCR4 internalization. Our results unveil a previously unknown property of MIM that establishes the linkage of protein ubiquitylation with Rab-guided trafficking of CXCR4 in endocytic vesicles.
Collapse
Affiliation(s)
- Lushen Li
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shaneen S Baxter
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ning Gu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xi Zhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA .,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
26
|
Role of Akt2 in regulation of metastasis suppressor 1 expression and colorectal cancer metastasis. Oncogene 2017; 36:3104-3118. [PMID: 28068324 DOI: 10.1038/onc.2016.460] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/27/2016] [Accepted: 10/31/2016] [Indexed: 12/15/2022]
Abstract
Survival signaling is critical for the metastatic program of cancer cells. The current study investigated the role of Akt survival proteins in colorectal cancer (CRC) metastasis and explored potential mechanisms of Akt-mediated metastasis regulation. Using an orthotopic implantation model in mice, which uniquely recapitulates the entire multistep process of CRC metastasis, combined with an inducible system of short hairpin RNA-mediated Akt isoform knockdown in human CRC cells, our studies confirm a role of Akt2 in CRC cell dissemination to distant organs in vivo. Akt2 deficiency profoundly inhibited the development of liver lesions in mice, whereas Akt1 had no effect under the experimental conditions used in the study. Array analysis of human metastatic genes identified the scaffolding protein metastasis suppressor 1 (MTSS1) as a novel Akt2-regulated gene. Inducible loss of Akt2 in CRC cells robustly upregulated MTSS1 at the messenger RNA and protein level, and the accumulated protein was functionally active as shown by its ability to engage an MTSS1-Src-cortactin inhibitory axis. MTSS1 expression led to a marked reduction in levels of functional cortacin (pcortactin Y421), an actin nucleation-promoting factor that has a crucial role in cancer cell invasion and metastasis. MTSS1 was also shown to mediate suppressive effects of Akt2 deficiency on CRC cell viability, survival, migration and actin polymerization in vitro. The relevance of these findings to human CRC is supported by analysis of The Cancer Genome Atlas (TCGA) and NCBI GEO data sets, which demonstrated inverse changes in expression of Akt2 and MTSS1 during CRC progression. Taken together, the data identify MTSS1 as a new Akt2-regulated gene, and point to suppression of MTSS1 as a key step in the metastasis-promoting effects of Akt2 in CRC cells.
Collapse
|
27
|
Li L, Liu H, Baxter SS, Gu N, Ji M, Zhan X. The SH3 domain distinguishes the role of I-BAR proteins IRTKS and MIM in chemotactic response to serum. Biochem Biophys Res Commun 2016; 479:787-792. [PMID: 27693783 DOI: 10.1016/j.bbrc.2016.09.131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/26/2016] [Indexed: 02/03/2023]
Abstract
The family of inverse BAR (I-BAR) domain proteins participates in a range of cellular processes associated with membrane dynamics and consists of five distinct members. Three of the I-BAR proteins, including insulin receptor tyrosine kinase substrate (IRTKS), contain an SH3 domain near their C-termini. Yet, the function of the SH3 domain of IRTKS remains uncharacterized. Here we report that in contrast to MIM, which is a prototype of I-BAR proteins and does not contain an SH3 domain, IRTKS promoted serum-induced cell migration along with enhanced phosphorylation of mitogen activated kinases Erk1/2 and p38, and activation of small GTPases Rac1 and Cdc42. In addition, cells overexpressing IRTKS exhibited an increased polarity characterized by elongated cytoplasm and extensive lamellipodia at leading edges. However, a mutant with deletion of the SH3 domain attenuated both cellular motility and p38 phosphorylation but had little effect on Erk1/2 phosphorylation. Also, a chimeric mutant in which the N-terminal portion of MIM is fused with the C-terminal IRTKS, including the SH3 domain, was able to promote chemotactic response to serum and cellular polarity. In contrast, a chimeric mutant in which the N-terminal IRTKS is fused with the C-terminal MIM failed to do so. Furthermore, treatment of cells with SB203580, a selective inhibitor of p38, also neutralized the effect of IRTKS on cell migration. These data indicate that the SH3 domain distinguishes the function of IRTKS in promoting cell migration and inducing signal transduction from those of SH3-less I-BAR proteins.
Collapse
Affiliation(s)
- Lushen Li
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; The Center for Vascular and Inflammatory Diseases and The Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hongyu Liu
- China-Japan Union Hospital of Jilin University, Changchun, 130031, China
| | - Shaneen S Baxter
- The Center for Vascular and Inflammatory Diseases and The Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ning Gu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Xi Zhan
- The Center for Vascular and Inflammatory Diseases and The Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
28
|
Li T, Xue Y, Wang G, Gu T, Li Y, Zhu YY, Chen L. Multi-target siRNA: Therapeutic Strategy for Hepatocellular Carcinoma. J Cancer 2016; 7:1317-27. [PMID: 27390607 PMCID: PMC4934040 DOI: 10.7150/jca.15157] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/18/2016] [Indexed: 12/23/2022] Open
Abstract
Multiple targets RNAi strategy is a preferred way to treat multigenic diseases, especially cancers. In the study, multi-target siRNAs were designed to inhibit NET-1, EMS1 and VEGF genes in hepatocellular carcinoma (HCC) cells. And multi-target siRNAs showed better silencing effects on NET-1, EMS1 and VEGF, compared with single target siRNA. Moreover, multi-target siRNA showed greater suppression effects on proliferation, migration, invasion, angiogenesis and induced apoptosis in HCC cells. The results suggested that multi-target siRNA might be a preferred strategy for cancer therapy and NET-1, EMS1 and VEGF could be effective targets for HCC treatments.
Collapse
Affiliation(s)
- Tiejun Li
- 1. Department of Pathological Anatomy, Nantong University, Nantong, China;; 2. Small RNA Technology and Application Institute, Nantong University, Nantong, China;; 3. Biomics Biotechnologies Co., Ltd., Nantong, China
| | - Yuwen Xue
- 1. Department of Pathological Anatomy, Nantong University, Nantong, China
| | - Guilan Wang
- 1. Department of Pathological Anatomy, Nantong University, Nantong, China
| | - Tingting Gu
- 1. Department of Pathological Anatomy, Nantong University, Nantong, China
| | - Yunlong Li
- 1. Department of Pathological Anatomy, Nantong University, Nantong, China
| | - York Yuanyuan Zhu
- 2. Small RNA Technology and Application Institute, Nantong University, Nantong, China;; 3. Biomics Biotechnologies Co., Ltd., Nantong, China
| | - Li Chen
- 1. Department of Pathological Anatomy, Nantong University, Nantong, China
| |
Collapse
|
29
|
Zhan T, Cao C, Li L, Gu N, Civin CI, Zhan X. MIM regulates the trafficking of bone marrow cells via modulating surface expression of CXCR4. Leukemia 2016; 30:1327-34. [PMID: 26965284 PMCID: PMC4889520 DOI: 10.1038/leu.2016.39] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/15/2015] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Missing in metastasis (MIM) is abundantly expressed in hematopoietic cells. Here we characterized the impact of MIM deficiency on murine bone marrow (BM) cells. Although MIM-/- cells proliferated similarly to wild type (WT), they exhibited stronger response to chemokine SDF-1, increase in surface expression of CXCR4, impaired CXCR4 internalization and constitutive activation of Rac, Cdc42 and p38. Transplantation of MIM-/- BM cells into lethally irradiated mice showed enhanced homing to BM, which was abolished when mice were pretreated with a p38 antagonist. Interestingly, MIM-/- BM cells, including hematopoietic stem and progenitor cells (HSPCs), showed 2 to 5-fold increase in mobilization into the peripheral blood upon treatment with AMD3100. In vitro, MIM-/- leukocytes were susceptible to AMD3100 and maintained increased response to AMD3100 for mobilization even after transfer into wild type mice. MIM-/- mice had also a higher level of SDF-1 in the circulation. Our data highlighted an unprecedented role of MIM in the homoeostasis of BM cells, including HSPCs, through modulation of the CXCR4/SDF-1 axis and interactions of BM leukocytes with their microenvironments.
Collapse
Affiliation(s)
- T Zhan
- Department of Pathology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - C Cao
- Department of Pathology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - L Li
- Department of Pathology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA.,China Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - N Gu
- China Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - C I Civin
- Department of Pediatrics, Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Departments of Pediatrics and Physiology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - X Zhan
- Department of Pathology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA.,Departments of Pediatrics and Physiology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
Zhao P, Cao M, Song L, Wu H, Hu K, Chen B, Wang Q, Gu N. Downregulation of MIM protein inhibits the cellular endocytosis process of magnetic nanoparticles in macrophages. RSC Adv 2016. [DOI: 10.1039/c6ra21530k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
MIM plays a positive role in the RAW 264.7 cellular endocytosis process of iron oxide nanoparticles mainly in clathrin-mediated pathway, which is a meaningful molecular basis for biomedical applications of nanomaterials.
Collapse
Affiliation(s)
- Peng Zhao
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Meng Cao
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Lina Song
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Hao Wu
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Ke Hu
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Bo Chen
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Qiwei Wang
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| | - Ning Gu
- State Key Laboratory of Bioelectronics
- Jiangsu Key Laboratory for Biomaterials and Devices
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
| |
Collapse
|
31
|
Yan MD, Yao CJ, Chow JM, Chang CL, Hwang PA, Chuang SE, Whang-Peng J, Lai GM. Fucoidan Elevates MicroRNA-29b to Regulate DNMT3B-MTSS1 Axis and Inhibit EMT in Human Hepatocellular Carcinoma Cells. Mar Drugs 2015; 13:6099-116. [PMID: 26404322 PMCID: PMC4626681 DOI: 10.3390/md13106099] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 09/08/2015] [Accepted: 09/14/2015] [Indexed: 12/16/2022] Open
Abstract
Accumulating evidence has revealed that fucoidan exhibits anti-tumor activities by arresting cell cycle and inducing apoptosis in many types of cancer cells including hepatocellular carcinoma (HCC). Exploring its effect on microRNA expression, we found that fucoidan markedly upregulated miR-29b of human HCC cells. The induction of miR-29b was accompanied with suppression of its downstream target DNMT3B in a dose-dependent manner. The reduction of luciferase activity of DNMT3B 3'-UTR reporter by fucoidan was as markedly as that by miR-29b mimic, indicating that fucoidan induced miR-29b to suppress DNMT3B. Accordingly, the mRNA and protein levels of MTSS1 (metastasis suppressor 1), a target silenced by DNMT3B, were increased after fucoidan treatment. Furthermore, fucoidan also down-regulated TGF-β receptor and Smad signaling of HCC cells. All these effects leaded to the inhibition of EMT (increased E-cadherin and decreased N-cadherin) and prevention of extracellular matrix degradation (increased TIMP-1 and decreased MMP2, 9), by which the invasion activity of HCC cells was diminished. Our results demonstrate the profound effect of fucoidan not only on the regulation of miR-29b-DNMT3B-MTSS1 axis but also on the inhibition of TGF-β signaling in HCC cells, suggesting the potential of using fucoidan as integrative therapeutics against invasion and metastasis of HCC.
Collapse
Affiliation(s)
- Ming-De Yan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; E-Mails: (M.-D.Y.); (C.-J.Y.); (J.W.-P.)
| | - Chih-Jung Yao
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; E-Mails: (M.-D.Y.); (C.-J.Y.); (J.W.-P.)
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; E-Mail:
- Comprehensive Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Jyh-Ming Chow
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; E-Mail:
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; E-Mail:
| | - Chia-Lun Chang
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; E-Mail:
| | - Pai-An Hwang
- Seafood Technology Division, Fisheries Research Institute, Council of Agriculture, Keelung 20246, Taiwan; E-Mail:
| | - Shuang-En Chuang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County 35053, Taiwan; E-Mail:
| | - Jacqueline Whang-Peng
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; E-Mails: (M.-D.Y.); (C.-J.Y.); (J.W.-P.)
- Comprehensive Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Gi-Ming Lai
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; E-Mails: (M.-D.Y.); (C.-J.Y.); (J.W.-P.)
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; E-Mail:
- Comprehensive Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; E-Mail:
- National Institute of Cancer Research, National Health Research Institutes, Miaoli County 35053, Taiwan; E-Mail:
| |
Collapse
|
32
|
Kedmi M, Ben-Chetrit N, Körner C, Mancini M, Ben-Moshe NB, Lauriola M, Lavi S, Biagioni F, Carvalho S, Cohen-Dvashi H, Schmitt F, Wiemann S, Blandino G, Yarden Y. EGF induces microRNAs that target suppressors of cell migration: miR-15b targets MTSS1 in breast cancer. Sci Signal 2015; 8:ra29. [PMID: 25783158 DOI: 10.1126/scisignal.2005866] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Growth factors promote tumor growth and metastasis. We found that epidermal growth factor (EGF) induced a set of 22 microRNAs (miRNAs) before promoting the migration of mammary cells. These miRNAs were more abundant in human breast tumors relative to the surrounding tissue, and their abundance varied among breast cancer subtypes. One of these miRNAs, miR-15b, targeted the 3' untranslated region of MTSS1 (metastasis suppressor protein 1). Although xenografts in which MTSS1 was knocked down grew more slowly in mice initially, longer-term growth was unaffected. Knocking down MTSS1 increased migration and Matrigel invasion of nontransformed mammary epithelial cells. Overexpressing MTSS1 in an invasive cell line decreased cell migration and invasiveness, decreased the formation of invadopodia and actin stress fibers, and increased the formation of cellular junctions. In tissues from breast cancer patients with the aggressive basal subtype, an inverse correlation occurred with the high expression of miRNA-15b and the low expression of MTSS1. Furthermore, low abundance of MTSS1 correlated with poor patient prognosis. Thus, growth factor-inducible miRNAs mediate mechanisms underlying the progression of cancer.
Collapse
Affiliation(s)
- Merav Kedmi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nir Ben-Chetrit
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Cindy Körner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Maicol Mancini
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Noa Bossel Ben-Moshe
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mattia Lauriola
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sara Lavi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Francesca Biagioni
- Translational Oncogenomics Unit, Italian National Cancer Institute "Regina Elena," Rome 00144, Italy
| | - Silvia Carvalho
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hadas Cohen-Dvashi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Fernando Schmitt
- Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto and Department of Pathology, University Health Network, Toronto, Ontario M5C 2C4, Canada. IPATIMUP, University of Porto, Porto 4200-465, Portugal
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Giovanni Blandino
- Translational Oncogenomics Unit, Italian National Cancer Institute "Regina Elena," Rome 00144, Italy
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
33
|
Downregulation of MTSS1 expression is an independent prognosticator in squamous cell carcinoma of the lung. Br J Cancer 2015; 112:866-73. [PMID: 25625275 PMCID: PMC4453956 DOI: 10.1038/bjc.2015.2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/22/2014] [Indexed: 12/29/2022] Open
Abstract
Background: The metastasis suppressor 1 (MTSS1) is a newly discovered protein putatively involved in tumour progression and metastasis. Material and Methods: Immunohistochemical expression of MTSS1 was analysed in 264 non-small-cell lung carcinomas (NSCLCs). Results: The metastasis suppressor 1 was significantly overexpressed in NSCLC compared with normal lung (P=0.01). Within NSCLC, MTSS1 expression was inversely correlated with pT-stage (P=0.019) and histological grading (P<0.001). NSCLC with MTSS1 downregulation (<20%) showed a significantly worse outcome (P=0.007). This proved to be an independent prognostic factor in squamous cell carcinomas (SCCs; P=0.041), especially in early cancer stages (P=0.006). Conclusion: The metastasis suppressor 1 downregulation could thus serve as a stratifying marker for adjuvant therapy in early-stage SCC of the lung.
Collapse
|
34
|
Zhong J, Shaik S, Wan L, Tron AE, Wang Z, Sun L, Inuzuka H, Wei W. SCF β-TRCP targets MTSS1 for ubiquitination-mediated destruction to regulate cancer cell proliferation and migration. Oncotarget 2014; 4:2339-53. [PMID: 24318128 PMCID: PMC3926831 DOI: 10.18632/oncotarget.1446] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Metastasis suppressor 1 (MTSS1) is an important tumor suppressor protein, and loss of MTSS1 expression has been observed in several types of human cancers. Importantly, decreased MTSS1 expression is associated with more aggressive forms of breast and prostate cancers, and with poor survival rate. Currently, it remains unclear how MTSS1 is regulated in cancer cells, and whether reduced MTSS1 expression contributes to elevated cancer cell proliferation and migration. Here we report that the SCFβ-TRCP regulates MTSS1 protein stability by targeting it for ubiquitination and subsequent destruction via the 26S proteasome. Notably, depletion of either Cullin 1 or β-TRCP1 led to increased levels of MTSS1. We further demonstrated a crucial role for Ser322 in the DSGXXS degron of MTSS1 in governing SCFβ-TRCP-mediated MTSS1 degradation. Mechanistically, we defined that Casein Kinase Iδ (CKIδ) phosphorylates Ser322 to trigger MTSS1's interaction with β-TRCP for subsequent ubiquitination and degradation. Importantly, introducing wild-type MTSS1 or a non-degradable MTSS1 (S322A) into breast or prostate cancer cells with low MTSS1 expression significantly inhibited cellular proliferation and migration. Moreover, S322A-MTSS1 exhibited stronger effects in inhibiting cell proliferation and migration when compared to ectopic expression of wild-type MTSS1. Therefore, our study provides a novel molecular mechanism for the negative regulation of MTSS1 by β-TRCP in cancer cells. It further suggests that preventing MTSS1 degradation could be a possible novel strategy for clinical treatment of more aggressive breast and prostate cancers.
Collapse
Affiliation(s)
- Jiateng Zhong
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhang S, Qi Q. MTSS1 suppresses cell migration and invasion by targeting CTTN in glioblastoma. J Neurooncol 2014; 121:425-31. [PMID: 25385572 DOI: 10.1007/s11060-014-1656-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/26/2014] [Indexed: 10/24/2022]
Abstract
Glioblastomas (GBMs) are the highest grade of primary brain tumors with astrocytic similarity and are characterized dispersal of tumor cell. Metastasis suppressor 1 (MTSS1) play an important role in cancer metastasis. Recent studies indicating that MTSS1 as a potential tumor suppressor and its reduced expression associated with poor prognosis in many cancer types. However, the relationship with the prognosis of patients and the molecular mechanism of MTSS1 renders a tumor suppressor effect in GBM is unknown. Here, we showed that low MTSS1 gene expression is associated with poor outcomes in patients with GBM. Overexpression of MTSS1 in U-87 MG cells exhibited inhibited glioma cell growth, colony formation, migration and invasion. Mechanistically, we found that high MTSS1 expression in U-87 MG reduced expression of CTTN. These results implicate that the role of MTSS1 suppresses cell migration and invasion by inhibiting expression of CTTN and as a prognosis biomarker in GBM.
Collapse
Affiliation(s)
- Shoudan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, 121000, China
| | | |
Collapse
|
36
|
Methods of Cell Propulsion through the Local Stroma in Breast Cancer. Int J Breast Cancer 2014; 2014:197480. [PMID: 24808960 PMCID: PMC3997937 DOI: 10.1155/2014/197480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/18/2014] [Indexed: 01/08/2023] Open
Abstract
In the normal breast, cellular structures change cyclically in response to ovarian hormones. Cell proliferation, apoptosis, invasion, and differentiation are integral processes that are precisely regulated. Normal epithelial cells depend on the formation of intercellular adhesion contacts to form a continuous sheet of stratifying cell layers that are attached to one and other horizontally and vertically.
Cells migrate by extending membrane protrusions to explore the extracellular space locating their targets in a chemotactic manner. The formation of cell protrusions is driven by the assembly of actin filaments at the leading edge. Reorganisation is regulated by a highly integrated signalling cascade that transduces extracellular stimuli to the actin filaments. This signalling cascade is governed by GTPases which act as molecular switches leading to actin polymerisation and the formation of filopodia and lamellipodia. This process is linked to downstream molecules known collectively as WASP proteins, which, in the presence of cortactin, form a complex leading to nucleation and formation of branched filaments. In breast cancer, the cortactin is over expressed leading to increased cellular motility and invasiveness. This hugely complex and integrated signalling cascade transduces extracellular stimuli. There are multiple genes related to cell motility which are dysregulated in human breast cancers.
Collapse
|
37
|
Mertz KD, Pathria G, Wagner C, Saarikangas J, Sboner A, Romanov J, Gschaider M, Lenz F, Neumann F, Schreiner W, Nemethova M, Glassmann A, Lappalainen P, Stingl G, Small JV, Fink D, Chin L, Wagner SN. MTSS1 is a metastasis driver in a subset of human melanomas. Nat Commun 2014; 5:3465. [PMID: 24632752 DOI: 10.1038/ncomms4465] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 02/18/2014] [Indexed: 12/21/2022] Open
Abstract
In cancers with a highly altered genome, distinct genetic alterations drive subsets rather than the majority of individual tumours. Here we use a sequential search across human tumour samples for transcript outlier data points with associated gene copy number variations that correlate with patient's survival to identify genes with pro-invasive functionality. Employing loss and gain of function approaches in vitro and in vivo, we show that one such gene, MTSS1, promotes the ability of melanocytic cells to metastasize and engages actin dynamics via Rho-GTPases and cofilin in this process. Indeed, high MTSS1 expression defines a subgroup of primary melanomas with unfavourable prognosis. These data underscore the biological, clinical and potential therapeutic implications of molecular subsets within genetically complex cancers.
Collapse
Affiliation(s)
- Kirsten D Mertz
- 1] Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria [2] [3]
| | - Gaurav Pathria
- 1] Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria [2]
| | - Christine Wagner
- 1] Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria [2]
| | - Juha Saarikangas
- 1] Institute of Biotechnology, University of Helsinki, 00100 Helsinki, Finland [2]
| | - Andrea Sboner
- Department of Pathology and Laboratory Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York 10021, USA
| | - Julia Romanov
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Melanie Gschaider
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Florian Lenz
- Section for Biosimulation and Bioinformatics, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, 1090 Vienna, Austria
| | - Friederike Neumann
- Section for Biosimulation and Bioinformatics, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfgang Schreiner
- Section for Biosimulation and Bioinformatics, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, 1090 Vienna, Austria
| | - Maria Nemethova
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, 1030 Vienna, Austria
| | | | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, 00100 Helsinki, Finland
| | - Georg Stingl
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - J Victor Small
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Dieter Fink
- Institute for Laboratory Animal Sciences, Department of Biomedical Sciences, University for Veterinary Medicine, 1210 Vienna, Austria
| | - Lynda Chin
- Department of Genomic Medicine and Institute for Applied Cancer Science, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Stephan N Wagner
- 1] Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria [2] Center for Molecular Medicine (CeMM), Austrian Academy of Sciences, 1090 Vienna, Austria
| |
Collapse
|
38
|
Wang F, Liu Y, Zhang H. Loss of MTSS1 Expression is an Independent Prognostic Factor for Hilar Cholangiocarcinoma. Pathol Oncol Res 2013; 19:815-20. [DOI: 10.1007/s12253-013-9649-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 05/02/2013] [Indexed: 12/12/2022]
|
39
|
Chen Q, Chen L, Zhao R, Yang XD, Imran K, Xing CG. Microarray analyses reveal liver metastasis-related genes in metastatic colorectal cancer cell model. J Cancer Res Clin Oncol 2013; 139:1169-78. [PMID: 23563852 DOI: 10.1007/s00432-013-1424-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 03/22/2013] [Indexed: 02/04/2023]
Abstract
PURPOSE To study the molecular mechanisms of colorectal cancer liver metastasis. METHODS Cecal wall implantation was performed in nude mice to subclone a highly liver metastatic human colorectal cancer clone (SW1116-M) from SW1116. In vivo and in vitro assays were adopted to confirm the proliferation and metastasis potential. The human tumor metastasis PCR microarrays were used to analyze the differential gene expressions. The results were confirmed further by real-time quantitative PCR. RESULTS SW1116-M and SW1116-S5, two human colon cancer cell clones with different metastatic potential, were subcloned from SW1116. In SW1116-M, in vitro invasion, migration and in vivo metastatic potential were higher, and in vitro proliferation rate was lower than SW1116-S5. In tumor metastasis PCR microarray, 24 genes related to cell invading, adhesion, cellular growth and differentiation were found with a twofold difference between SW1116-S5 and SW1116-M. Sixteen of these, including E-cadherins, MTSS1, TRAIL and TRPM1, were up-regulated; eight genes including cathepsin L, EphB2, HGF, MET, MCAM and RORβ were down-regulated. CONCLUSIONS We have established a highly liver metastatic clone. The subsequent metastasis PCR microarray analysis identified a procedure of cellular differentiation and mesenchymal to epithelial transition (MET) in liver metastasis. The colonization to from macrometastasis is not a switch from cell cycle arrest but a result of cell differentiation and MET.
Collapse
Affiliation(s)
- Qiang Chen
- General Surgery Department, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Rd, Suzhou 215004, China
| | | | | | | | | | | |
Collapse
|
40
|
Suppression of MIM by microRNA-182 activates RhoA and promotes breast cancer metastasis. Oncogene 2013; 33:1287-96. [DOI: 10.1038/onc.2013.65] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/18/2012] [Accepted: 01/05/2013] [Indexed: 02/07/2023]
|
41
|
MacGrath SM, Koleske AJ. Cortactin in cell migration and cancer at a glance. J Cell Sci 2013; 125:1621-6. [PMID: 22566665 DOI: 10.1242/jcs.093781] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Stacey M MacGrath
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA.
| | | |
Collapse
|
42
|
Chen Y, Aardema J, Corey SJ. Biochemical and functional significance of F-BAR domain proteins interaction with WASP/N-WASP. Semin Cell Dev Biol 2013; 24:280-6. [PMID: 23384583 DOI: 10.1016/j.semcdb.2013.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Accepted: 01/16/2013] [Indexed: 01/17/2023]
Abstract
The Bin-Amphiphysin-Rvs (BAR) domain family of proteins includes groups which promote positive (classical BAR, N-BAR, and F-BAR) and negative (I-BAR) membrane deformation. Of these groups, the F-BAR subfamily is the most diverse in its biochemical properties. F-BAR domain proteins dimerize to form a tight scaffold about the membrane. The F-BAR domain provides a banana-shaped, alpha-helical structure that senses membrane curvature. Different types of F-BAR domain proteins contain tyrosine kinase or GTPase activities; some interact with phosphatases and RhoGTPases. Most possess an SH3 domain that facilitates the recruitment and activation of WASP/N-WASP. Thus, F-BAR domain proteins affect remodeling of both membrane and the actin cytoskeleton. The purpose of this review is to highlight the role of F-BAR proteins in coupling WASP/N-WASP to cytoskeletal remodeling. A role for F-BAR/WASP interaction in human diseases affecting nervous, blood, and neoplastic tissues is discussed.
Collapse
Affiliation(s)
- Yolande Chen
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, United States
| | | | | |
Collapse
|
43
|
Abstract
MIM [missing in metastasis; also called MTSS1 (metastasis suppressor 1)] is an intracellular protein that binds to actin and cortactin and has an intrinsic capacity to sense and facilitate the formation of protruded membranous curvatures implicated in cellular polarization, mobilization and endocytosis. The N-terminal 250 amino acids of MIM undergo homodimerization and form a structural module with the characteristic of an I-BAR [inverse BAR (Bin/amphiphysin/Rvs)] domain. To discern the role of the dimeric configuration in the function of MIM, we designed several peptides able to interfere with MIM dimerization in a manner dependent upon their lengths. Overexpression of one of the peptides effectively abolished MIM-mediated membrane protrusions and transferrin uptake. However, a peptide with a high potency inhibiting MIM dimerization failed to affect its binding to actin and cortactin. Thus the results of the present study indicate that the dimeric configuration is essential for MIM-mediated membrane remodelling and serves as a proper target to develop antagonists specifically against an I-BAR-domain-containing protein.
Collapse
|
44
|
Branched F-actin as a negative regulator of cilia formation. Exp Cell Res 2012; 319:147-51. [PMID: 22975729 DOI: 10.1016/j.yexcr.2012.08.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 08/30/2012] [Accepted: 08/31/2012] [Indexed: 01/18/2023]
Abstract
Cilia dysfunction leads to developmental defects and also a spectrum of human diseases termed ciliopathies. The actin cytoskeleton is a highly dynamic network and involved in many important biological processes, such as cell migration and membrane trafficking. Recently, actin dynamics has been shown to play a critical role in ciliogenesis. This review summarizes these results and provides insight into possible mechanisms.
Collapse
|
45
|
Abstract
Endocytosis includes a number of processes by which cells internalize segments of their plasma membrane, enclosing a wide variety of material from outside the cell. Endocytosis can contribute to uptake of nutrients, regulation of signaling molecules, control of osmotic pressure, and function of synapses. The actin cytoskeleton plays an essential role in several of these processes. Actin assembly can create protrusions that encompass extracellular materials. Actin can also support the processes of invagination of a membrane segment into the cytoplasm, elongation of the invagination, scission of the new vesicle from the plasma membrane, and movement of the vesicle away from the membrane. We briefly discuss various types of endocytosis, including phagocytosis, macropinocytosis, and clathrin-independent endocytosis. We focus mainly on new findings on the relative importance of actin in clathrin-mediated endocytosis (CME) in yeast versus mammalian cells.
Collapse
Affiliation(s)
- Olivia L Mooren
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA.
| | | | | |
Collapse
|
46
|
Wang J, Li J, Shen J, Wang C, Yang L, Zhang X. MicroRNA-182 downregulates metastasis suppressor 1 and contributes to metastasis of hepatocellular carcinoma. BMC Cancer 2012; 12:227. [PMID: 22681717 PMCID: PMC3492170 DOI: 10.1186/1471-2407-12-227] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 06/01/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND miR-182 is one of the most significantly up-regulated miRNAs in hepatocellular carcinoma (HCC). Metastasis suppressor 1 (MTSS1), one target gene of miR-182, plays an important role in the metastasis of cancers. However, it remains unclear what role does function and mechanism of miR-182 and MTSS1play in HCC. METHODS miR-182 expression was tested in 86 cases of paired HCC and normal tissues by real-time PCR and the relationships between miR-182 expression and clinicopathological parameters were analyzed. The expression of MTSS1 was evaluated by immunohistochemistry and western blot in the above tissues and its correlation with miR-182 expression was analyzed. Moreover, western blot and invasion assays were performed after transfection of pre-miR-182 or anti-miR-182 to HCC cell lines. In addition, luciferase assays was performed to confirm the regulation of miR-182 on MTSS1. RESULTS Compared with normal tissue, miR-182 was up-regulated and MTSS1 was down-regulated in HCC tissues. Moreover, the over-expression of miR-182 was correlated with intrahepatic metastasis (p = 0.034) and poor prognosis (p = 0.039) of HCC patients. There was a negative correlation between miR-182 and MTSS1 expression in both HCC tissues (r = -0.673, p < 0.01) and HCC cell lines (r = -0.931, p = 0.021). Furthermore, the up-regulation of miR-182 resulted in the down-regulation of MTSS1 and increased invasive potential of HUH-1, and reverse results were also confirmed when the expression of miR-182 was inhibited. In addition, the results of the luciferase assay demonstrated the targeted regulation of miR-182 on MTSS1. CONCLUSIONS miR-182 could promote metastasis of HCC and inhibit the expression of MTSS1. miR-182 and MTSS1 are potential prognostic markers and/or therapeutic targets in HCC.
Collapse
Affiliation(s)
- Jian Wang
- Department of 4th Abdominal Oncology, Cancer Hospital and Institute of Tianjin Medical University, Tianjin, 300060, China.
| | | | | | | | | | | |
Collapse
|
47
|
Dawson JC, Timpson P, Kalna G, Machesky LM. Mtss1 regulates epidermal growth factor signaling in head and neck squamous carcinoma cells. Oncogene 2012; 31:1781-93. [PMID: 21927027 PMCID: PMC3245856 DOI: 10.1038/onc.2011.376] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 07/13/2011] [Accepted: 07/22/2011] [Indexed: 12/11/2022]
Abstract
Mtss1 is located within chromosomal region 8q23-24, which is one of the three most commonly amplified regions in head and neck squamous cell carcinoma (HNSCC). Mtss1 is lost in metastatic cells, but confusingly is commonly overexpressed in primary tumors. Here we address possible reasons why Mtss1 is positively selected for in primary tumors. We find that Mtss1 enhances the localization of the epidermal growth factor (EGF) receptor to the plasma membrane, prolonging EGF signaling and resulting in enhanced proliferation in HNSCC. Depletion of Mtss1 results in decreased EGF receptor levels and decreased phosphorylation of Erk1/2 and Akt. However, when cells are at high density and adherent to each other, analogous to conditions in a solid tumor, Mtss1 does not confer any growth advantage, either in basal conditions or following EGF stimulation. This could indicate why Mtss1 might be lost in metastases, but preserved in early primary tumors. This is supported by an organotypic assay showing that Mtss1-expressing cells display a less proliferative more epithelial-like morphology on top of a collagen matrix. Furthermore, xenograft tumors expressing Mtss1 initially grow more rapidly, but later show less proliferation and more differentiation. Mtss1 positively modulates EGF signaling at low cell densities to promote proliferation and, therefore, may be beneficial for the early stages of primary HNSCC tumor growth. However, at high cell densities, Mtss1 impacts negatively on EGF signaling and this suggests why it inhibits metastasis.
Collapse
Affiliation(s)
- John C. Dawson
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, Scotland, UK
| | - Paul Timpson
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, Scotland, UK
| | - Gabriela Kalna
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, Scotland, UK
| | - Laura M. Machesky
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, Scotland, UK
| |
Collapse
|
48
|
Jahid S, Sun J, Edwards RA, Dizon D, Panarelli NC, Milsom JW, Sikandar SS, Gümüs ZH, Lipkin SM. miR-23a promotes the transition from indolent to invasive colorectal cancer. Cancer Discov 2012; 2:540-53. [PMID: 22628407 DOI: 10.1158/2159-8290.cd-11-0267] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Colorectal cancer is a classic example of a tumor that progresses through multiple distinct stages in its evolution. To understand the mechanisms regulating the transition from indolent to invasive disease, we profiled somatic copy number alterations in noninvasive adenomas and invasive adenocarcinomas from Apc and DNA mismatch repair (MMR) mutant mouse models. We identified a recurrent amplicon on mouse chromosome 8 that encodes microRNA (miRNA) 23a and -27a (miR). miR-23a and -27a levels are upregulated in mouse intestinal adenocarcinomas, primary tumors from patients with stage I/II colorectal cancers, as well as in human colorectal cancer cell lines and cancer stem cells. Functionally, miR-23a promotes the migration and invasion of colorectal cancer cells and stem cells, whereas miR-27a primarily promotes proliferation. We computationally and experimentally validated that metastasis suppressor 1 (MTSS1) is a direct miR-23a target and similarly validated that the ubiquitin ligase FBXW7 is a direct miR-27a target. Analyses of computationally predicted target genes in microarray data sets of patients with colorectal cancers are consistent with a role for miR-23a, but not miR-27a, specifically in invasive colorectal cancers.
Collapse
Affiliation(s)
- Sohail Jahid
- Departments of Medicine, Genetic Medicine, Surgery, Pathology and Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Dawson JC, Bruche S, Spence HJ, Braga VMM, Machesky LM. Mtss1 promotes cell-cell junction assembly and stability through the small GTPase Rac1. PLoS One 2012; 7:e31141. [PMID: 22479308 PMCID: PMC3313965 DOI: 10.1371/journal.pone.0031141] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 01/03/2012] [Indexed: 11/19/2022] Open
Abstract
Cell-cell junctions are an integral part of epithelia and are often disrupted in cancer cells during epithelial-to-mesenchymal transition (EMT), which is a main driver of metastatic spread. We show here that Metastasis suppressor-1 (Mtss1; Missing in Metastasis, MIM), a member of the IMD-family of proteins, inhibits cell-cell junction disassembly in wound healing or HGF-induced scatter assays by enhancing cell-cell junction strength. Mtss1 not only makes cells more resistant to cell-cell junction disassembly, but also accelerates the kinetics of adherens junction assembly. Mtss1 drives enhanced junction formation specifically by elevating Rac-GTP. Lastly, we show that Mtss1 depletion reduces recruitment of F-actin at cell-cell junctions. We thus propose that Mtss1 promotes Rac1 activation and actin recruitment driving junction maintenance. We suggest that the observed loss of Mtss1 in cancers may compromise junction stability and thus promote EMT and metastasis.
Collapse
Affiliation(s)
- John C. Dawson
- Beatson Institute for Cancer Research, Glasgow, United Kingdom
| | - Susann Bruche
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Vania M. M. Braga
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
50
|
Grigera PR, Ma L, Borgman CA, Pinto AF, Sherman NE, Parsons JT, Fox JW. Mass spectrometric analysis identifies a cortactin-RCC2/TD60 interaction in mitotic cells. J Proteomics 2012; 75:2153-9. [PMID: 22282019 DOI: 10.1016/j.jprot.2012.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 01/08/2012] [Accepted: 01/09/2012] [Indexed: 10/24/2022]
Abstract
Cortactin is an F-actin binding protein that functions as a scaffold to regulate Arp2/3 mediated actin polymerization in lamellipodia and invadopodia formation as well as functioning in cell migration and endocytosis of many different cell types. In light of the fact that regulated actin polymerization is critical for many cellular processes we launched a search for novel cortactin interactions with cellular proteins that might indicate heretofore undescribed biological activities supported by cortactin. Using a modified stable isotope labeling in cell culture (SILAC) approach in HEK293 cells and Flag-tagged cortactin (F-cortactin) as bait, we identified a limited set of cortactin interactions including several proteins which have not previously been identified as cortactin associated proteins. Among these were serine/threonine-protein phosphatase 2A subunit beta (PP2A-beta) and RCC2/TD60, a Rac guanine nucleotide exchange factor (GEF) required for completion of mitosis and cytokinesis. The interaction between cortactin and RCC2/TD60 was verified in cell lysates immunoprecitated with anti-RCC2/TD60 antibody. Furthermore, cortactin was localized by immunofluorescence in the equatorial plane of dividing HeLa cells in the region where RCC2/TD60 has previously been localized thus providing support for a complex containing cortactin and RCC2/TD60 complex that may play a functional role in cells undergoing mitosis.
Collapse
Affiliation(s)
- Pablo R Grigera
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, United States
| | | | | | | | | | | | | |
Collapse
|