1
|
Spagnuolo FD, Kronemberger GS, Storey KJ, Kelly DJ. The maturation state and density of human cartilage microtissues influence their fusion and development into scaled-up grafts. Acta Biomater 2025; 194:109-121. [PMID: 39818242 DOI: 10.1016/j.actbio.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/09/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Functional cartilaginous tissues can potentially be engineered by bringing together numerous microtissues (µTs) and allowing them to fuse and re-organize into larger, structurally organized grafts. The maturation level of individual microtissues is known to influence their capacity to fuse, however its impact on the long-term development of the resulting tissue remains unclear. The first objective of this study was to investigate the influence of the maturation state of human bone-marrow mesenchymal stem/stromal cells (hBM-MSCSs) derived microtissues on their fusion capacity and the phenotype of the final engineered tissue. Less mature (day 2) cartilage microtissues were found to fuse faster, supporting the development of a matrix that was richer in sulphated glycosaminoglycans (sGAG) and collagen, while low in calcium deposits. This enhanced fusion in less mature microtissues correlated with enhanced expression of N-cadherin, followed by a progressive increase in markers associated with cell-extracellular matrix (ECM) interactions. We then engineered larger constructs with varying initial numbers (50, 150 or 300 µTs per well) of less mature microtissues, observing enhanced sGAG synthesis with increased microtissue density. We finally sought to engineer a scaled-up cartilage graft by fusing 4,000 microtissues and maintaining the resulting constructs under either dynamic or static culture conditions. Robust and reliable fusion was observed between microtissues at this scale, with no clear benefit of dynamic culture on the levels of matrix accumulation or the tensile modulus of the resulting construct. These results support the use of BM-MSCs derived microtissues for the development of large-scale, engineered functional cartilaginous grafts. STATEMENT OF SIGNIFICANCE: Microtissues are gaining attention for their use as biological building blocks in the field of tissue engineering. The fusion of multiple microtissues is crucial for achieving a cohesive engineered tissue of scale, however the impact of their maturation level on the long-term properties of the engineered graft is poorly understood. This paper emphasizes the importance of using less mature cartilage microtissues for supporting appropriate cell-cell interactions and robust chondrogenesis in vitro. We demonstrate that tissue development is not negatively impacted by increasing the initial numbers of microtissues within the graft. This biofabrication strategy has significant translation potential, as it enables the engineering of scaled-up cartilage grafts of clinically relevant sizes using bone marrow derived MSCs.
Collapse
Affiliation(s)
- Francesca D Spagnuolo
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Gabriela S Kronemberger
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Kyle J Storey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
2
|
Zhao L, Chen Y, Li H, Ding X, Li J. Deciphering the neuroprotective mechanisms of RACK1 in cerebral ischemia-reperfusion injury: Pioneering insights into mitochondrial autophagy and the PINK1/Parkin axis. CNS Neurosci Ther 2024; 30:e14836. [PMID: 39097918 PMCID: PMC11298203 DOI: 10.1111/cns.14836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 05/23/2024] [Accepted: 06/16/2024] [Indexed: 08/06/2024] Open
Abstract
INTRODUCTION Cerebral ischemia-reperfusion injury (CIRI) is a common and debilitating complication of cerebrovascular diseases such as stroke, characterized by mitochondrial dysfunction and cell apoptosis. Unraveling the molecular mechanisms behind these processes is essential for developing effective CIRI treatments. This study investigates the role of RACK1 (receptor for activated C kinase 1) in CIRI and its impact on mitochondrial autophagy. METHODS We utilized high-throughput transcriptome sequencing and weighted gene co-expression network analysis (WGCNA) to identify core genes associated with CIRI. In vitro experiments used human neuroblastoma SK-N-SH cells subjected to oxygen and glucose deprivation (OGD) to simulate ischemia, followed by reperfusion (OGD/R). RACK1 knockout cells were created using CRISPR/Cas9 technology, and cell viability, apoptosis, and mitochondrial function were assessed. In vivo experiments involved middle cerebral artery occlusion/reperfusion (MCAO/R) surgery in rats, evaluating neurological function and cell apoptosis. RESULTS Our findings revealed that RACK1 expression increases during CIRI and is protective by regulating mitochondrial autophagy through the PINK1/Parkin pathway. In vitro, RACK1 knockout exacerbated cell apoptosis, while overexpression of RACK1 reversed this process, enhancing mitochondrial function. In vivo, RACK1 overexpression reduced cerebral infarct volume and improved neurological deficits. The regulatory role of RACK1 depended on the PINK1/Parkin pathway, with RACK1 knockout inhibiting PINK1 and Parkin expression, while RACK1 overexpression restored them. CONCLUSION This study demonstrates that RACK1 safeguards against neural damage in CIRI by promoting mitochondrial autophagy through the PINK1/Parkin pathway. These findings offer crucial insights into the regulation of mitochondrial autophagy and cell apoptosis by RACK1, providing a promising foundation for future CIRI treatments.
Collapse
Affiliation(s)
- Lanqing Zhao
- Department of Sleep Medicine CenterThe Shengjing Affiliated Hospital, China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Yu Chen
- Department of Otorhinolaryngology Head and NeckShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Hongxi Li
- Department of Pain ManagementShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Xiaoxu Ding
- Department of Otorhinolaryngology Head and NeckShengjing Hospital of China Medical UniversityShenyangLiaoningPeople's Republic of China
| | - Jinwei Li
- Department of Neurology/Stroke CenterThe First Affiliated Hospital of China Medical University, China Medical UniversityShenyangLiaoningPeople's Republic of China
| |
Collapse
|
3
|
Abbey CA, Duran CL, Chen Z, Chen Y, Roy S, Coffell A, Sveeggen TM, Chakraborty S, Wells GB, Chang J, Bayless KJ. Identification of New Markers of Angiogenic Sprouting Using Transcriptomics: New Role for RND3. Arterioscler Thromb Vasc Biol 2024; 44:e145-e167. [PMID: 38482696 PMCID: PMC11043006 DOI: 10.1161/atvbaha.123.320599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/28/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND New blood vessel formation requires endothelial cells to transition from a quiescent to an invasive phenotype. Transcriptional changes are vital for this switch, but a comprehensive genome-wide approach focused exclusively on endothelial cell sprout initiation has not been reported. METHODS Using a model of human endothelial cell sprout initiation, we developed a protocol to physically separate cells that initiate the process of new blood vessel formation (invading cells) from noninvading cells. We used this model to perform multiple transcriptomics analyses from independent donors to monitor endothelial gene expression changes. RESULTS Single-cell population analyses, single-cell cluster analyses, and bulk RNA sequencing revealed common transcriptomic changes associated with invading cells. We also found that collagenase digestion used to isolate single cells upregulated the Fos proto-oncogene transcription factor. Exclusion of Fos proto-oncogene expressing cells revealed a gene signature consistent with activation of signal transduction, morphogenesis, and immune responses. Many of the genes were previously shown to regulate angiogenesis and included multiple tip cell markers. Upregulation of SNAI1 (snail family transcriptional repressor 1), PTGS2 (prostaglandin synthase 2), and JUNB (JunB proto-oncogene) protein expression was confirmed in invading cells, and silencing JunB and SNAI1 significantly reduced invasion responses. Separate studies investigated rounding 3, also known as RhoE, which has not yet been implicated in angiogenesis. Silencing rounding 3 reduced endothelial invasion distance as well as filopodia length, fitting with a pathfinding role for rounding 3 via regulation of filopodial extensions. Analysis of in vivo retinal angiogenesis in Rnd3 heterozygous mice confirmed a decrease in filopodial length compared with wild-type littermates. CONCLUSIONS Validation of multiple genes, including rounding 3, revealed a functional role for this gene signature early in the angiogenic process. This study expands the list of genes associated with the acquisition of a tip cell phenotype during endothelial cell sprout initiation.
Collapse
Affiliation(s)
- Colette A. Abbey
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Camille L. Duran
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Zhishi Chen
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Yanping Chen
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Sukanya Roy
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
| | - Ashley Coffell
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Timothy M. Sveeggen
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Sanjukta Chakraborty
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
| | - Gregg B. Wells
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, TX
| | - Jiang Chang
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Kayla J. Bayless
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| |
Collapse
|
4
|
Gao Z, Sun W, Ni X, Wan W, Suo T, Ni X, Liu H, Li N, Sheng Shen, Liu H. Low expression of RACK1 is associated with metastasis and worse prognosis in cholangiocarcinoma. Heliyon 2024; 10:e27366. [PMID: 38509930 PMCID: PMC10950496 DOI: 10.1016/j.heliyon.2024.e27366] [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] [Received: 06/29/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Cholangiocarcinoma is a poorly prognostic malignant tumor, and the metastatic stage of cancer is not an early stage when diagnosed. Lymph node metastasis is common in the early stage. Ribosomal receptor for activated C-kinase 1 (RACK1) has found involved in the oncogenesis of various tumors and in the epithelial-mesenchymal transition (EMT). Nevertheless, its role in cholangiocarcinoma remains unknown. MATERIAL AND METHODS The possible correlation between RACK1 and tumor prognosis was analyzed in cholangiocarcinoma patients. The GEO and TCGA databases were used to evaluate the level of RACK1 in cholangiocarcinoma. The RBE and HCCC-9810 cell lines were used to examine the effects of RACK1 in the behavior of tumor cells in vitro. RESULTS The Kaplan-Meier analysis indicated that low expression of RACK1 was associated with poor prognosis and RACK1 was negatively related to lymph node metastasis, which were verified in databases TCGA and GEO; downregulation of RACK1 via RNA interference correlated with changes in the expression of EMT biomarkers and promoted the migration of cholangiocarcinoma cell lines. CONCLUSION The protein expression of RACK1 is significantly higher in cholangiocarcinoma tissues than in peritumoral tissues, however, the high RACK1 expression indicates better overall survival and less risk for lymph node metastasis. In vitro, RACK1 may suppress the migratory ability of cholangiocarcinoma cells by inhibiting EMT.
Collapse
Affiliation(s)
- Zhihui Gao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Wentao Sun
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Xiaojian Ni
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Wenze Wan
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Tao Suo
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Xiaoling Ni
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Han Liu
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Na Li
- Basic Medical Institute, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Sheng Shen
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
| | - Houbao Liu
- Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Biliary Tract Disease Institute, Fudan University, Shanghai 200032, China
- Shanghai Biliary Tract Minimal Invasive Surgery and Materials Engineering Research Center, Shanghai 200032, China
- Department of General Surgery, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, China
| |
Collapse
|
5
|
Sveeggen TM, Abbey CA, Smith RL, Salinas ML, Chapkin RS, Bayless KJ. Annexin A2 modulates phospholipid membrane composition upstream of Arp2 to control angiogenic sprout initiation. FASEB J 2023; 37:e22715. [PMID: 36527391 PMCID: PMC10586062 DOI: 10.1096/fj.202201088r] [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: 07/14/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
The intersection of protein and lipid biology is of growing importance for understanding how cells address structural challenges during adhesion and migration. While protein complexes engaged with the cytoskeleton play a vital role, support from the phospholipid membrane is crucial for directing localization and assembly of key protein complexes. During angiogenesis, dramatic cellular remodeling is necessary for endothelial cells to shift from a stable monolayer to invasive structures. However, the molecular dynamics between lipids and proteins during endothelial invasion are not defined. Here, we utilized cell culture, immunofluorescence, and lipidomic analyses to identify a novel role for the membrane binding protein Annexin A2 (ANXA2) in modulating the composition of specific membrane lipids necessary for cortical F-actin organization and adherens junction stabilization. In the absence of ANXA2, there is disorganized cortical F-actin, reduced junctional Arp2, excess sprout initiation, and ultimately failed sprout maturation. Furthermore, we observed reduced filipin III labeling of membrane cholesterol in cells with reduced ANXA2, suggesting there is an alteration in phospholipid membrane dynamics. Lipidomic analyses revealed that 42 lipid species were altered with loss of ANXA2, including an accumulation of phosphatidylcholine (16:0_16:0). We found that supplementation of phosphatidylcholine (16:0_16:0) in wild-type endothelial cells mimicked the ANXA2 knock-down phenotype, indicating that ANXA2 regulated the phospholipid membrane upstream of Arp2 recruitment and organization of cortical F-actin. Altogether, these data indicate a novel role for ANXA2 in coordinating events at endothelial junctions needed to initiate sprouting and show that proper lipid modulation is a critical component of these events.
Collapse
Affiliation(s)
- Timothy M. Sveeggen
- Texas A&M Health Science Center, Texas, Bryan, USA
- Interdisciplinary Graduate Program in Genetics, Texas A&M University, College Station, Texas, USA
| | | | | | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas, USA
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas, USA
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | | |
Collapse
|
6
|
Shakhov AS, Alieva IB. The "Third Violin" in the Cytoskeleton Orchestra-The Role of Intermediate Filaments in the Endothelial Cell's Life. Biomedicines 2022; 10:828. [PMID: 35453578 PMCID: PMC9027429 DOI: 10.3390/biomedicines10040828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/01/2023] Open
Abstract
The endothelium plays an important role in the transcytosis of lipoproteins. According to one of the theories, endothelial injury is a triggering factor for the development of atherosclerosis, and intracellular structures, including components of the endotheliocyte cytoskeleton (microtubules, actin, and intermediate filaments), are involved in its development. In contrast to the proteins of tubulin-based microtubules and actin microfilaments, intermediate filaments are comprised of various tissue-specific protein members. Vimentin, the main protein of endothelial intermediate filaments, is one of the most well-studied of these and belongs to type-III intermediate filaments, commonly found in cells of mesenchymal origin. Vimentin filaments are linked mechanically or by signaling molecules to microfilaments and microtubules by which coordinated cell polarisation and migration are carried out, as well as control over several endotheliocyte functions. Moreover, the soluble vimentin acts as an indicator of the state of the cardiovascular system, and the involvement of vimentin in the development and course of atherosclerosis has been demonstrated. Here we discuss current concepts of the participation of vimentin filaments in the vital activity and functioning of endothelial cells, as well as the role of vimentin in the development of inflammatory processes and atherosclerosis.
Collapse
Affiliation(s)
| | - Irina B. Alieva
- A.N. Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| |
Collapse
|
7
|
Ostrowska-Podhorodecka Z, McCulloch CA. Vimentin regulates the assembly and function of matrix adhesions. Wound Repair Regen 2021; 29:602-612. [PMID: 33887795 DOI: 10.1111/wrr.12920] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022]
Abstract
The intermediate filament protein vimentin is a widely used phenotypic marker for identifying cells of the mesenchymal linkage such as fibroblasts and myofibroblasts, but the full repertoire of vimentin's functional attributes has not been fully explored. Here we consider how vimentin, in addition to its contributions to mechanical stabilization of cell structure, also helps to control the assembly of cell adhesions and migration through collagen matrices. While the assembly and function of matrix adhesions are critical for the differentiation of myofibroblasts and many other types of adherent cells, a potential mechanism that explains how vimentin affects the recruitment and abundance of centrally important proteins in cell adhesions has been elusive. Here we review recent data indicating that vimentin plays a central regulatory role in the assembly of focal adhesions which form in response to the attachment to collagen. We show that in particular, vimentin is a key organizer of the β1 integrin adhesive machinery, which affects cell migration through collagen. This review provides a comprehensive picture of the surprisingly broad array of processes and molecules with which vimentin interacts to affect cell function in the context of fibroblast and myofibroblast adhesion and migration on collagen.
Collapse
|
8
|
Ostrowska-Podhorodecka Z, Ding I, Lee W, Tanic J, Abbasi S, Arora PD, Liu RS, Patteson AE, Janmey PA, McCulloch CA. Vimentin tunes cell migration on collagen by controlling β1 integrin activation and clustering. J Cell Sci 2021; 134:jcs.254359. [PMID: 33558312 DOI: 10.1242/jcs.254359] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Vimentin is a structural protein that is required for mesenchymal cell migration and directly interacts with actin, β1 integrin and paxillin. We examined how these interactions enable vimentin to regulate cell migration on collagen. In fibroblasts, depletion of vimentin increased talin-dependent activation of β1 integrin by more than 2-fold. Loss of vimentin was associated with reduction of β1 integrin clustering by 50% and inhibition of paxillin recruitment to focal adhesions by more than 60%, which was restored by vimentin expression. This reduction of paxillin was associated with 65% lower Cdc42 activation, a 60% reduction of cell extension formation and a greater than 35% decrease in cell migration on collagen. The activation of PAK1, a downstream effector of Cdc42, was required for vimentin phosphorylation and filament maturation. We propose that vimentin tunes cell migration through collagen by acting as an adaptor protein for focal adhesion proteins, thereby regulating β1 integrin activation, resulting in well-organized, mature integrin clusters.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Isabel Ding
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Wilson Lee
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Jelena Tanic
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Sevil Abbasi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Richard S Liu
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Alison E Patteson
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA.,Physics Department, Syracuse University, Syracuse, NY 13244, USA
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA
| | | |
Collapse
|
9
|
Roy-Luzarraga M, Abdel-Fatah T, Reynolds LE, Clear A, Taylor JG, Gribben JG, Chan S, Jones L, Hodivala-Dilke K. Association of Low Tumor Endothelial Cell pY397-Focal Adhesion Kinase Expression With Survival in Patients With Neoadjuvant-Treated Locally Advanced Breast Cancer. JAMA Netw Open 2020; 3:e2019304. [PMID: 33107920 PMCID: PMC7592032 DOI: 10.1001/jamanetworkopen.2020.19304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
IMPORTANCE Determining the risk of relapse after neoadjuvant chemotherapy in patients with locally advanced breast cancer is required to offer alternative therapeutic strategies. OBJECTIVE To examine whether endothelial cell phosphorylated-focal adhesion kinase (EC-pY397-FAK) expression in patients with treatment-naive locally advanced breast cancer is a biomarker for chemotherapy sensitivity and is associated with survival after neoadjuvant chemotherapy. DESIGN, SETTING, AND PARTICIPANTS In this prognostic study, expression levels of EC-pY397-FAK and tumor cell (TC)-pY397-FAK were determined by immunohistochemistry in prechemotherapy core biopsies from 82 female patients with locally advanced breast cancer treated with anthracycline-based combination neoadjuvant chemotherapy at Nottingham City Hospital in Nottingham, UK. Median follow-up time was 67 months. The study was conducted from December 1, 2010, to September 28, 2019, and data analysis was performed from October 2, 2019, to March 31, 2020. EXPOSURES All women underwent surgery followed by adjuvant radiotherapy and, if tumors were estrogen receptor-positive, 5-year tamoxifen treatment. MAIN OUTCOMES AND MEASURES Outcomes were pathologic complete response and 5-year relapse-free survival examined using Kaplan-Meier, univariable logistic, multivariable logistic, and Cox proportional hazards models. RESULTS A total of 82 women (age, 29-76 years) with locally advanced breast cancer (stage IIA-IIIC) were included. Of these, 21 women (26%) had high EC-pY397-FAK expression that was associated with estrogen receptor positivity (71% vs 46%; P = .04), progesterone receptor positivity (67% vs 39%; P = .03), high Ki67 (86% vs 41%; P < .001), 4-immunohistochemically stained luminal-B (52% vs 8%; P < .001), higher tumor category (T3/T4 category: 90% vs 59%; P = .01), high lymph node category (N2-3 category: 43% vs 5%; P < .001), and high tumor node metastasis stage (IIIA-IIIC: 90% vs 66%; P = .03). Of 21 patients with high EC-pY397-FAK expression levels, none showed pathologic complete response, compared with 11 of 61 patients with low EC-pY397-FAK expression levels who showed pathologic complete response (odds ratio, 0.70; 95% CI, 0.61-0.82; P = .04). High EC-pY397-FAK expression levels and high blood vessel density (BVD) were associated with shorter 5-year relapse-free survival compared with those with low EC-pY397-FAK expression levels (hazard ratio [HR], 2.21; 95% CI, 1.17-4.20; P = .01) and low BVD (HR, 2.2; 95% CI, 1.15-4.35; P = .02). High TC-pY397-FAK expression levels in 15 of 82 women (18%) were not associated significantly with pathologic complete response or 5-year relapse-free survival. A multivariable Cox regression model for 5-year relapse-free survival indicated that high EC-pY397-FAK expression levels was an independent poor prognostic factor after controlling for other validated prognostic factors (HR, 3.91; 95% CI, 1.42-10.74; P = .01). Combined analysis of EC-pY397-FAK expression levels, TC-pY397-FAK expression levels, and BVD improved prognostic significance over individually tested features. CONCLUSIONS AND RELEVANCE The findings of this study suggest that low EC-pY397-FAK expression levels are associated with chemotherapy sensitivity and improved 5-year relapse-free survival after systemic therapy. Combined analysis of high EC-pY397-FAK expression levels, high TC-pY397-FAK expression levels, and high BVD appeared to identify a high-risk population.
Collapse
Affiliation(s)
- Marina Roy-Luzarraga
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Tarek Abdel-Fatah
- Department of Clinical Oncology, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
- Pathology Department, National Liver Institute, Minoufyia University, Al Minufiyah, Egypt
| | - Louise E. Reynolds
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Andrew Clear
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Joseph G. Taylor
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - John G. Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Stephen Chan
- Department of Clinical Oncology, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| | - Kairbaan Hodivala-Dilke
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, United Kingdom
| |
Collapse
|
10
|
Bott CJ, Winckler B. Intermediate filaments in developing neurons: Beyond structure. Cytoskeleton (Hoboken) 2020; 77:110-128. [PMID: 31970897 DOI: 10.1002/cm.21597] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/20/2022]
Abstract
Neuronal development relies on a highly choreographed progression of dynamic cellular processes by which newborn neurons migrate, extend axons and dendrites, innervate their targets, and make functional synapses. Many of these dynamic processes require coordinated changes in morphology, powered by the cell's cytoskeleton. Intermediate filaments (IFs) are the third major cytoskeletal elements in vertebrate cells, but are rarely considered when it comes to understanding axon and dendrite growth, pathfinding and synapse formation. In this review, we first introduce the many new and exciting concepts of IF function, discovered mostly in non-neuronal cells. These roles include dynamic rearrangements, crosstalk with microtubules and actin filaments, mechano-sensing and -transduction, and regulation of signaling cascades. We then discuss the understudied roles of neuronally expressed IFs, with a particular focus on IFs expressed during development, such as nestin, vimentin and α-internexin. Lastly, we illustrate how signaling modulation by the unconventional IF nestin shapes neuronal morphogenesis in unexpected and novel ways. Even though the first IF knockout mice were made over 20 years ago, the study of the cell biological functions of IFs in the brain still has much room for exciting new discoveries.
Collapse
Affiliation(s)
- Christopher J Bott
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
11
|
Strouhalova K, Přechová M, Gandalovičová A, Brábek J, Gregor M, Rosel D. Vimentin Intermediate Filaments as Potential Target for Cancer Treatment. Cancers (Basel) 2020; 12:E184. [PMID: 31940801 PMCID: PMC7017239 DOI: 10.3390/cancers12010184] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Intermediate filaments constitute the third component of the cellular skeleton. Unlike actin and microtubule cytoskeletons, the intermediate filaments are composed of a wide variety of structurally related proteins showing distinct expression patterns in tissues and cell types. Changes in the expression patterns of intermediate filaments are often associated with cancer progression; in particular with phenotypes leading to increased cellular migration and invasion. In this review we will describe the role of vimentin intermediate filaments in cancer cell migration, cell adhesion structures, and metastasis formation. The potential for targeting vimentin in cancer treatment and the development of drugs targeting vimentin will be reviewed.
Collapse
Affiliation(s)
- Katerina Strouhalova
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Magdalena Přechová
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Aneta Gandalovičová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Daniel Rosel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| |
Collapse
|
12
|
Intermediate Filaments as Effectors of Cancer Development and Metastasis: A Focus on Keratins, Vimentin, and Nestin. Cells 2019; 8:cells8050497. [PMID: 31126068 PMCID: PMC6562751 DOI: 10.3390/cells8050497] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
Intermediate filament (IF) proteins make up the largest family of cytoskeletal proteins in metazoans, and are traditionally known for their roles in fostering structural integrity in cells and tissues. Remarkably, individual IF genes are tightly regulated in a fashion that reflects the type of tissue, its developmental and differentiation stages, and biological context. In cancer, IF proteins serve as diagnostic markers, as tumor cells partially retain their original signature expression of IF proteins. However, there are also characteristic alterations in IF gene expression and protein regulation. The use of high throughput analytics suggests that tumor-associated alterations in IF gene expression have prognostic value. Parallel research is also showing that IF proteins directly and significantly impact several key cellular properties, including proliferation, death, migration, and invasiveness, with a demonstrated impact on the development, progression, and characteristics of various tumors. In this review, we draw from recent studies focused on three IF proteins most associated with cancer (keratins, vimentin, and nestin) to highlight how several “hallmarks of cancer” described by Hanahan and Weinberg are impacted by IF proteins. The evidence already in hand establishes that IF proteins function beyond their classical roles as markers and serve as effectors of tumorigenesis.
Collapse
|
13
|
Dave JM, Mirabella T, Weatherbee SD, Greif DM. Pericyte ALK5/TIMP3 Axis Contributes to Endothelial Morphogenesis in the Developing Brain. Dev Cell 2018; 44:665-678.e6. [PMID: 29456135 DOI: 10.1016/j.devcel.2018.01.018] [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: 02/14/2017] [Revised: 12/22/2017] [Accepted: 01/22/2018] [Indexed: 12/16/2022]
Abstract
The murine embryonic blood-brain barrier (BBB) consists of endothelial cells (ECs), pericytes (PCs), and basement membrane. Although PCs are critical for inducing vascular stability, signaling pathways in PCs that regulate EC morphogenesis during BBB development remain unexplored. Herein, we find that murine embryos lacking the transforming growth factor β (TGF-β) receptor activin receptor-like kinase 5 (Alk5) in brain PCs (mutants) develop gross germinal matrix hemorrhage-intraventricular hemorrhage (GMH-IVH). The germinal matrix (GM) is a highly vascularized structure rich in neuronal and glial precursors. We show that GM microvessels of mutants display abnormal dilation, reduced PC coverage, EC hyperproliferation, reduced basement membrane collagen, and enhanced perivascular matrix metalloproteinase activity. Furthermore, ALK5-depleted PCs downregulate tissue inhibitor of matrix metalloproteinase 3 (TIMP3), and TIMP3 administration to mutants improves endothelial morphogenesis and attenuates GMH-IVH. Overall, our findings reveal a key role for PC ALK5 in regulating brain endothelial morphogenesis and a substantial therapeutic potential for TIMP3 during GMH-IVH.
Collapse
Affiliation(s)
- Jui M Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 773J, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Teodelinda Mirabella
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 773J, New Haven, CT 06511, USA
| | - Scott D Weatherbee
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daniel M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 773J, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
14
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
15
|
BPIFB1 (LPLUNC1) inhibits migration and invasion of nasopharyngeal carcinoma by interacting with VTN and VIM. Br J Cancer 2017; 118:233-247. [PMID: 29123267 PMCID: PMC5785741 DOI: 10.1038/bjc.2017.385] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/21/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Bactericidal/Permeability-increasing-fold-containing family B member 1 (BPIFB1, previously termed LPLUNC1) is highly expressed in the nasopharynx, significantly downregulated in nasopharyngeal carcinoma (NPC), and associated with prognosis in NPC patients. Because metastasis represents the primary cause of NPC-related death, we explored the role of BPIFB1 in NPC migration and invasion. Methods: The role of BPIFB1 in NPC metastasis was investigated in vitro and in vivo. A co-immunoprecipitation assay coupled with mass spectrometry was used to identify BPIFB1-binding proteins. Additionally, western blotting, immunofluorescence, and immunohistochemistry allowed assessment of the molecular mechanisms associated with BPIFB1-specific metastatic inhibition via vitronectin (VTN) and vimentin (VIM) interactions. Results: Our results showed that BPIFB1 expression markedly inhibited NPC cell migration, invasion, and lung-metastatic abilities. Additionally, identification of two BPIFB1-interacting proteins, VTN and VIM, showed that BPIFB1 reduced VTN expression and the formation of a VTN-integrin αV complex in NPC cells, leading to inhibition of the FAK/Src/ERK signalling pathway. Moreover, BPIFB1 attenuated NPC cell migration and invasion by inhibiting VTN- or VIM-induced epithelial–mesenchymal transition. Conclusions: This study represents the first demonstration of BPIFB1 function in NPC migration, invasion, and lung metastasis. Our findings indicate that re-expression of BPIFB1 might represent a useful strategy for preventing and treating NPC.
Collapse
|
16
|
Liao S, Xiao S, Chen H, Zhang M, Chen Z, Long Y, Gao L, He J, Ge Y, Yi W, Wu M, Li G, Zhou Y. The receptor for activated protein kinase C promotes cell growth, invasion and migration in cervical cancer. Int J Oncol 2017; 51:1497-1507. [PMID: 29048616 PMCID: PMC5642390 DOI: 10.3892/ijo.2017.4137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 09/21/2017] [Indexed: 01/21/2023] Open
Abstract
Cervical cancer is one of the most common malignant tumors in women all over the world. However, the exact etiology of cervical cancer remains unclear. The receptor for activated protein kinase C (RACK1) is reported to be involved in tumorigenesis and tumor progression. Besides, the prognostic value of RACK1 in several kinds of tumors has been identified. However, there are limited studies on the functional role of RACK1 in cervical cancer. In this study, we tested the expression level of RACK1 by immunohistochemistry and western blot technologies and find that it is upregulated in cervical cancer. Colony formation and CCK8 assays indicate that RACK1 promotes cell proliferation in CaSki cervical cancer cells. While the silence of RACK1 decreases the cell proliferation in CCK8 analysis. β-galactosidase staining suggests that RACK1 decreases cell senescence in cervical cancer cells. Invasion and migration assay show that RACK1 promotes the invasion and migration of cervical cancer cells. Also, when RACK1 was silenced, it exerts the opposite result. Furthermore, the mRNA expression levels of MMP‑3, MMP‑9 and MMP‑10 were upregulated in RACK1‑overexpressed CaSki cells by qPCR analysis. RACK1 also induces S phase accumulation in cell cycle analysis and suppresses cell apoptosis in cervical cancer cells. Flow cytometry analysis of mitochondria functions suggests that RACK1 increases the mitochondrial membrane potential (Δψm) levels to prevent mitochondrial apoptosis in cervical cancer cells. To explore the possible mechanism of RACK1, we tested and found that RACK1 upregulates the expression of NF-κB, cyclin D1 and CDK4 and downregulates the expression of p53, p38, p21 and STAT1 in cervical cancer cells. These results suggest that RACK1 promotes cell growth and invasion and inhibits the senescence and apoptosis in cervical cancer cells probably by affecting the p53 pathway.
Collapse
Affiliation(s)
- Shan Liao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health, Xiangya Hospital
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan
| | - Hongxiang Chen
- The Gynecology Department, People's Hospital of Xinjiang
| | - Manying Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Zhifang Chen
- The Gynecology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang, P.R. China
| | - Yuehua Long
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Lu Gao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Junyu He
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Yanshan Ge
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Wei Yi
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health, Xiangya Hospital
| | - Yanhong Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health, Xiangya Hospital
| |
Collapse
|
17
|
Selective regulation of Notch ligands during angiogenesis is mediated by vimentin. Proc Natl Acad Sci U S A 2017; 114:E4574-E4581. [PMID: 28533359 PMCID: PMC5468602 DOI: 10.1073/pnas.1703057114] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Notch signaling is a key regulator of angiogenesis, in which sprouting is regulated by an equilibrium between inhibitory Dll4-Notch signaling and promoting Jagged-Notch signaling. Whereas Fringe proteins modify Notch receptors and strengthen their activation by Dll4 ligands, other mechanisms balancing Jagged and Dll4 signaling are yet to be described. The intermediate filament protein vimentin, which has been previously shown to affect vascular integrity and regenerative signaling, is here shown to regulate ligand-specific Notch signaling. Vimentin interacts with Jagged, impedes basal recycling endocytosis of ligands, but is required for efficient receptor ligand transendocytosis and Notch activation upon receptor binding. Analyses of Notch signal activation by using chimeric ligands with swapped intracellular domains (ICDs), demonstrated that the Jagged ICD binds to vimentin and contributes to signaling strength. Vimentin also suppresses expression of Fringe proteins, whereas depletion of vimentin enhances Fringe levels to promote Dll4 signaling. In line with these data, the vasculature in vimentin knockout (VimKO) embryos and placental tissue is underdeveloped with reduced branching. Disrupted angiogenesis in aortic rings from VimKO mice and in endothelial 3D sprouting assays can be rescued by reactivating Notch signaling by recombinant Jagged ligands. Taken together, we reveal a function of vimentin and demonstrate that vimentin regulates Notch ligand signaling activities during angiogenesis.
Collapse
|
18
|
Duff D, Long A. Roles for RACK1 in cancer cell migration and invasion. Cell Signal 2017; 35:250-255. [PMID: 28336233 DOI: 10.1016/j.cellsig.2017.03.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 01/16/2023]
Abstract
Migration and invasion of cancer cells into surrounding tissue and vasculature is an important initial step in cancer metastasis. Metastasis is the leading cause of cancer related death and thus it is crucial that we improve our understanding of the mechanisms that promote this life-threatening phenomenon. Cell migration involves a complex, multistep process that leads to the actin-driven movement of cells on or through the tissues of the body. The multifunctional scaffolding protein RACK1 plays important roles in nucleating cell signalling hubs, anchoring proteins at specific subcellular locations and regulating protein activity. It is essential for cell migration and accumulating evidence now demonstrates multiple roles for RACK1 in regulating migration and invasion of tumour cells. The possibility of designing drugs that block the migratory and invasive capabilities of cancer cells represents an attractive therapeutic strategy for treating malignant disease with RACK1 being a potential target. In this review we summarize this evidence and examine the mechanisms that underlie the contribution of RACK1 to the various stages of cell migration and invasion.
Collapse
Affiliation(s)
- Deirdre Duff
- Trinity Translational Medicine Institute, Trinity College Dublin, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
19
|
Duran CL, Kaunas R, Bayless KJ. S1P Synergizes with Wall Shear Stress and Other Angiogenic Factors to Induce Endothelial Cell Sprouting Responses. Methods Mol Biol 2017; 1697:99-115. [PMID: 28456951 DOI: 10.1007/7651_2017_26] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis is the process of new blood vessel growth from pre-existing structures. During sprout initiation, endothelial cells (ECs) are activated by pro-angiogenic factors to degrade the basement membrane, migrate into the surrounding matrix, and form structures that anastomose to connect neighboring vessels. Sphingosine 1-phosphate (S1P) is a biologically active lysosphingolipid that is secreted by platelets and promotes angiogenesis under normal and pathological conditions by acting on ECs. In addition to biochemical factors, the endothelium is continuously subjected to mechanical forces in the form of wall shear stress (WSS) from fluid forces. Here, we describe an in vitro, three-dimensional (3D) endothelial sprouting assay that is significantly enhanced by S1P, WSS, angiogenic growth factors (GFs), and fibronectin. This assay is assembled by seeding primary human endothelial cells onto 3D collagen matrices containing S1P and other pro-angiogenic factors. Once attached, physiological levels of WSS are applied to induce robust sprouting responses. This approach promotes the initiation of angiogenic sprouts stimulated by S1P, and allows the study of 3D sprouting of primary human endothelial cells induced in response to these physiological factors.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77843-1114, USA
| | - Roland Kaunas
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX, 77843-1114, USA.
| |
Collapse
|
20
|
Peng H, Gong PG, Li JB, Cai LM, Yang L, Liu YY, Yao KT, Li X. The important role of the receptor for activated C kinase 1 (RACK1) in nasopharyngeal carcinoma progression. J Transl Med 2016; 14:131. [PMID: 27170279 PMCID: PMC4864934 DOI: 10.1186/s12967-016-0885-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/28/2016] [Indexed: 01/26/2023] Open
Abstract
Background The receptor for activated C kinase 1 (RACK1) is involved in various cancers, but its roles in nasopharyngeal carcinoma (NPC) have not yet been fully elucidated. Methods Initially, RACK1 expression was analyzed by immunohistochemistry in NPC and normal nasopharyngeal (NP) tissues. It was also detected by qPCR and Western blot in NPC cells. Confocal microscope and immunofluorescence were performed to detect the subcellular compartmentalization of RACK1. Subsequently, after up- or down-regulating RACK1 in NPC cells, cell proliferation and migration/invasion were tested using in vitro assays including MTT, EdU, colony formation, Transwell and Boyden assays. Furthermore, several key molecules were detected by Western blot to explore underlying mechanism. Finally, clinical samples were analyzed to confirm the relationship between RACK1 expression and clinical features. Results Receptor for activated C kinase 1 expression was much higher in NPC than NP tissues. And RACK1 was mainly located in the cytoplasm. Overexpression of RACK1 promoted NPC cell proliferation and metastasis/invasion, whereas depletion of this protein suppressed NPC cell proliferation and metastasis/invasion. Mechanistically, RACK1 deprivation obviously suppressed the activation of Akt and FAK, suggesting the PI3K/Akt/FAK pathway as one of functional mechanisms of RACK1 in NPC. Furthermore, clinical sample analysis indicated a positive correlation between in vivo expression of RACK1 with lymph node invasion and clinical stage of NPC. Conclusion Our results demonstrate that RACK1 protein plays an important role in NPC development and progression. The upregulation of RACK1 can promote the proliferation and invasion of NPC by regulating the PI3K/Akt/FAK signal pathway. Thus, this study contributes to the discovery of a potential therapeutic target for NPC. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0885-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Peng
- Department of Otolaryngology-Head and Neck Surgery, The Second People's Hospital of Guangdong Province, Southern Medical University, Guangzhou, 510317, China.
| | - Ping-Gui Gong
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jin-Bang Li
- Department of Pathology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
| | - Long-Mei Cai
- Cancer Research Institute and the Provincial Key Laboratory of Cancer Immunotherapy, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Le Yang
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yun-Yi Liu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Kai-Tai Yao
- Cancer Research Institute and the Provincial Key Laboratory of Cancer Immunotherapy, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xin Li
- Cancer Research Institute and the Provincial Key Laboratory of Cancer Immunotherapy, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
21
|
Xiao Z, Zou Q, Liu Y, Yang X. Genome-wide assessment of differential translations with ribosome profiling data. Nat Commun 2016; 7:11194. [PMID: 27041671 PMCID: PMC4822032 DOI: 10.1038/ncomms11194] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 02/29/2016] [Indexed: 12/29/2022] Open
Abstract
The closely regulated process of mRNA translation is crucial for precise control of protein abundance and quality. Ribosome profiling, a combination of ribosome foot-printing and RNA deep sequencing, has been used in a large variety of studies to quantify genome-wide mRNA translation. Here, we developed Xtail, an analysis pipeline tailored for ribosome profiling data that comprehensively and accurately identifies differentially translated genes in pairwise comparisons. Applied on simulated and real datasets, Xtail exhibits high sensitivity with minimal false-positive rates, outperforming existing methods in the accuracy of quantifying differential translations. With published ribosome profiling datasets, Xtail does not only reveal differentially translated genes that make biological sense, but also uncovers new events of differential translation in human cancer cells on mTOR signalling perturbation and in human primary macrophages on interferon gamma (IFN-γ) treatment. This demonstrates the value of Xtail in providing novel insights into the molecular mechanisms that involve translational dysregulations.
Collapse
Affiliation(s)
- Zhengtao Xiao
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Beijing 100084, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China.,School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qin Zou
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China.,School of Life Sciences, Tsinghua University, Beijing 100084, China.,Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Science, Tsinghua University, Beijing 100084, China
| | - Yu Liu
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Beijing 100084, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China.,School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xuerui Yang
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Beijing 100084, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China.,School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
22
|
Dave JM, Abbey CA, Duran CL, Seo H, Johnson GA, Bayless KJ. Hic-5 mediates the initiation of endothelial sprouting by regulating a key surface metalloproteinase. J Cell Sci 2016; 129:743-56. [PMID: 26769900 DOI: 10.1242/jcs.170571] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 01/05/2016] [Indexed: 12/16/2022] Open
Abstract
During angiogenesis, endothelial cells must coordinate matrix proteolysis with migration. Here, we tested whether the focal adhesion scaffold protein Hic-5 (also known as TGFB1I1) regulated endothelial sprouting in three dimensions. Hic-5 silencing reduced endothelial sprouting and lumen formation, and sprouting defects were rescued by the return of Hic-5 expression. Pro-angiogenic factors enhanced colocalization and complex formation between membrane type-1 matrix metalloproteinase (MT1-MMP, also known as MMP14) and Hic-5, but not between paxillin and MT1-MMP. The LIM2 and LIM3 domains of Hic-5 were necessary and sufficient for Hic-5 to form a complex with MT1-MMP. The degree of interaction between MT1-MMP and Hic-5 and the localization of the complex within detergent-resistant membrane fractions were enhanced during endothelial sprouting, and Hic-5 depletion lowered the surface levels of MT1-MMP. In addition, we observed that loss of Hic-5 partially reduced complex formation between MT1-MMP and focal adhesion kinase (FAK, also known as PTK2), suggesting that Hic-5 bridges MT1-MMP and FAK. Finally, Hic-5 LIM2-LIM3 deletion mutants reduced sprout initiation. Hic-5, MT1-MMP and FAK colocalized in angiogenic vessels during porcine pregnancy, supporting that this complex assembles during angiogenesis in vivo. Collectively, Hic-5 appears to enhance complex formation between MT1-MMP and FAK in activated endothelial cells, which likely coordinates matrix proteolysis and cell motility.
Collapse
Affiliation(s)
- Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Colette A Abbey
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Heewon Seo
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Gregory A Johnson
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
23
|
Robert A, Hookway C, Gelfand VI. Intermediate filament dynamics: What we can see now and why it matters. Bioessays 2016; 38:232-43. [PMID: 26763143 DOI: 10.1002/bies.201500142] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mechanical properties of vertebrate cells are largely defined by the system of intermediate filaments (IF). As part of a dense network, IF polymers are constantly rearranged and relocalized in the cell to fulfill their duty as cells change shape, migrate, or divide. With the development of new imaging technologies, such as photoconvertible proteins and super-resolution microscopy, a new appreciation for the complexity of IF dynamics has emerged. This review highlights new findings about the transport of IF, the remodeling of filaments by a process of severing and re-annealing, and the subunit exchange that occurs between filament precursors and a soluble pool of IF. We will also discuss the unique dynamic features of the keratin IF network. Finally, we will speculate about how the dynamic properties of IF are related to their functions.
Collapse
Affiliation(s)
- Amélie Robert
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Caroline Hookway
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
24
|
Gallo S, Manfrini N. Working hard at the nexus between cell signaling and the ribosomal machinery: An insight into the roles of RACK1 in translational regulation. ACTA ACUST UNITED AC 2015; 3:e1120382. [PMID: 26824030 DOI: 10.1080/21690731.2015.1120382] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/19/2015] [Accepted: 11/09/2015] [Indexed: 02/08/2023]
Abstract
RACK1 is a ribosome-associated protein which functions as a receptor for activated PKCs. It also acts as a scaffold for many other proteins involved in diverse signaling pathways, e.g. Src, JNK, PDE4D and FAK signaling. With such a broad interactome, RACK1 has been suggested to function as a linker between cell signaling and the translation machinery. Accordingly, RACK1 modulates translation at different levels in several model organisms. For instance, it regulates ribosome stalling and mRNA quality control in yeasts and promotes translation efficiency downstream of specific cellular stimuli in mammals. However, the molecular mechanism by which RACK1 exerts these roles is widely uncharacterized. Moreover, the full list of ribosome-recruited RACK1 interactors still needs characterization. Here we discuss in vivo and in vitro findings to better delineate the roles of RACK1 in regulating ribosome function and translation.
Collapse
Affiliation(s)
- Simone Gallo
- Molecular Histology and Cell Growth Unit; National Institute of Molecular Genetics - INGM "Romeo and Enrica Invernizzi" ; Milan, Italy
| | - Nicola Manfrini
- Molecular Histology and Cell Growth Unit; National Institute of Molecular Genetics - INGM "Romeo and Enrica Invernizzi" ; Milan, Italy
| |
Collapse
|
25
|
Hyder CL, Kemppainen K, Isoniemi KO, Imanishi SY, Goto H, Inagaki M, Fazeli E, Eriksson JE, Törnquist K. Sphingolipids inhibit vimentin-dependent cell migration. J Cell Sci 2015; 128:2057-69. [PMID: 25908861 DOI: 10.1242/jcs.160341] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 04/20/2015] [Indexed: 12/15/2022] Open
Abstract
The sphingolipids, sphingosine 1-phosphate (S1P) and sphingosylphosphorylcholine (SPC), can induce or inhibit cellular migration. The intermediate filament protein vimentin is an inducer of migration and a marker for epithelial-mesenchymal transition. Given that keratin intermediate filaments are regulated by SPC, with consequences for cell motility, we wanted to determine whether vimentin is also regulated by sphingolipid signalling and whether it is a determinant for sphingolipid-mediated functions. In cancer cells where S1P and SPC inhibited migration, we observed that S1P and SPC induced phosphorylation of vimentin on S71, leading to a corresponding reorganization of vimentin filaments. These effects were sphingolipid-signalling-dependent, because inhibition of either the S1P2 receptor (also known as S1PR2) or its downstream effector Rho-associated kinase (ROCK, for which there are two isoforms ROCK1 and ROCK2) nullified the sphingolipid-induced effects on vimentin organization and S71 phosphorylation. Furthermore, the anti-migratory effect of S1P and SPC could be prevented by expressing S71-phosphorylation-deficient vimentin. In addition, we demonstrated, by using wild-type and vimentin-knockout mouse embryonic fibroblasts, that the sphingolipid-mediated inhibition of migration is dependent on vimentin. These results imply that this newly discovered sphingolipid-vimentin signalling axis exerts brake-and-throttle functions in the regulation of cell migration.
Collapse
Affiliation(s)
- Claire L Hyder
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, POB 123, FIN-20521, Turku, Finland Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Kati Kemppainen
- Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Kimmo O Isoniemi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, POB 123, FIN-20521, Turku, Finland Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Susumu Y Imanishi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, POB 123, FIN-20521, Turku, Finland Environmental Science Lab, Faculty of Pharmacy, Meijo University, Yagotoyama 150, Tempaku. Nagoya 468-8503, Japan
| | - Hidemasa Goto
- Division of Biochemistry, Aichi Cancer Center Research Institute, Kanokoden, Chikusa-Ku, Nagoya 464-8681, Japan Department of Cellular Oncology, Graduate School of Medicine, Nagoya University, Showa-Ku, Nagoya 466-8550, Japan
| | - Masaki Inagaki
- Division of Biochemistry, Aichi Cancer Center Research Institute, Kanokoden, Chikusa-Ku, Nagoya 464-8681, Japan Department of Cellular Oncology, Graduate School of Medicine, Nagoya University, Showa-Ku, Nagoya 466-8550, Japan
| | - Elnaz Fazeli
- Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland
| | - John E Eriksson
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, POB 123, FIN-20521, Turku, Finland Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland
| | - Kid Törnquist
- Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, FI-20520, Turku, Finland Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Tukholmankatu 8, 00290 Helsinki, Finland
| |
Collapse
|
26
|
Dave JM, Bayless KJ. Vimentin as an integral regulator of cell adhesion and endothelial sprouting. Microcirculation 2015; 21:333-44. [PMID: 24387004 DOI: 10.1111/micc.12111] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/30/2013] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a multistep process that requires intricate changes in cell shape to generate new blood vessels. IF are a large family of proteins that play an important structural and functional role in forming and regulating the cytoskeleton. Vimentin, a major type III intermediate filament protein is expressed in endothelial and other mesenchymal cells. The structure of vimentin is conserved in mammals and shows dynamic expression profiles in various cell types and different developmental stages. Although initial studies with vimentin-deficient mice demonstrated a virtually normal phenotype, subsequent studies have revealed several defects in cell attachment, migration, signaling, neurite extension, and vascularization. Regulation of vimentin is highly complex and is driven by posttranslational modifications such as phosphorylation and cleavage by intracellular proteases. This review discusses various novel functions which are now known to be mediated by vimentin, summarizing structure, regulation and roles of vimentin in cell adhesion, migration, angiogenesis, neurite extension, and cancer. We specifically highlight a pathway involving growth factor-mediated calpain activation, vimentin cleavage, and MT1-MMP membrane translocation that is required for endothelial cell invasion in 3D environments. This pathway may also regulate the analogous processes of neurite extension and tumor cell invasion.
Collapse
Affiliation(s)
- Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | | |
Collapse
|
27
|
Leduc C, Etienne-Manneville S. Intermediate filaments in cell migration and invasion: the unusual suspects. Curr Opin Cell Biol 2015; 32:102-12. [PMID: 25660489 DOI: 10.1016/j.ceb.2015.01.005] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 01/15/2015] [Accepted: 01/21/2015] [Indexed: 12/22/2022]
Abstract
Cell migration is a multistep process which relies on the coordination of cytoskeletal structures in space and time. While the roles of actin and microtubules have been investigated in great details, the lack of inhibitors and visualizing tools and the large number of proteins forming intermediate filaments (IFs) have delayed the characterization of IF functions during migration. However, a large body of evidence has progressively pointed to changes in IF composition as an important parameter in the regulation of cell migratory properties both during development and tumor invasion. More recent in-depth analyses show that IFs are dynamically reorganized to participate, together with microfilaments and microtubules, to the key steps leading to cell migration.
Collapse
Affiliation(s)
- Cécile Leduc
- Institut Pasteur - CNRS UMR 3691, Cell Polarity, Migration and Cancer Unit, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Sandrine Etienne-Manneville
- Institut Pasteur - CNRS UMR 3691, Cell Polarity, Migration and Cancer Unit, 25 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
28
|
Intermediate filaments and the regulation of focal adhesion. Curr Opin Cell Biol 2014; 32:13-20. [PMID: 25460777 DOI: 10.1016/j.ceb.2014.09.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/19/2022]
Abstract
Focal adhesions are localized actin filament-anchoring signalling centres at the cell-extracellular matrix interface. The currently emerging view is that they fulfil an all-embracing coordinating function for the entire cytoskeleton. This review highlights the tight relationship between focal adhesions and the intermediate filament cytoskeleton. We summarize the accumulating evidence for direct binding of intermediate filaments to focal adhesion components and their mutual cross-talk through signalling molecules. Examples are presented to emphasize the high degree of complexity of these interactions equipping cells with a precisely controlled machinery for context-dependent adjustment of their biomechanical properties.
Collapse
|
29
|
Abstract
Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. FAK promotes tumour progression and metastasis through effects on cancer cells, as well as stromal cells of the tumour microenvironment. The kinase-dependent and kinase-independent functions of FAK control cell movement, invasion, survival, gene expression and cancer stem cell self-renewal. Small molecule FAK inhibitors decrease tumour growth and metastasis in several preclinical models and have initial clinical activity in patients with limited adverse events. In this Review, we discuss FAK signalling effects on both tumour and stromal cell biology that provide rationale and support for future therapeutic opportunities.
Collapse
Affiliation(s)
- Florian J. Sulzmaier
- Department of Reproductive Medicine, UCSD Moores Cancer Center, La Jolla, CA 92093
| | - Christine Jean
- Department of Reproductive Medicine, UCSD Moores Cancer Center, La Jolla, CA 92093
| | - David D. Schlaepfer
- Department of Reproductive Medicine, UCSD Moores Cancer Center, La Jolla, CA 92093
- Address correspondence to: David D. Schlaepfer, Ph.D., University of California San Diego, Moores Cancer Center, Department of Reproductive Medicine, 3855 Health Sciences Dr., MC0803, La Jolla, CA 92093,
| |
Collapse
|
30
|
RACK1, a versatile hub in cancer. Oncogene 2014; 34:1890-8. [PMID: 24882575 DOI: 10.1038/onc.2014.127] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/07/2014] [Accepted: 04/10/2014] [Indexed: 01/15/2023]
Abstract
RACK1 is a highly conserved intracellular adaptor protein with significant homology to Gβ and was originally identified as the anchoring protein for activated protein kinase C. In the past 20 years, the number of binding partners and validated cellular functions for RACK1 has increased, which facilitates clarification of its involvement in different biological events. In this review, we will focus on its role in cancer, summarizing its aberrant expression, pro- or anti-oncogenic effects and the underlying mechanisms in various cancers.
Collapse
|
31
|
Robert A, Herrmann H, Davidson MW, Gelfand VI. Microtubule-dependent transport of vimentin filament precursors is regulated by actin and by the concerted action of Rho- and p21-activated kinases. FASEB J 2014; 28:2879-90. [PMID: 24652946 DOI: 10.1096/fj.14-250019] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Intermediate filaments (IFs) form a dense and dynamic network that is functionally associated with microtubules and actin filaments. We used the GFP-tagged vimentin mutant Y117L to study vimentin-cytoskeletal interactions and transport of vimentin filament precursors. This mutant preserves vimentin interaction with other components of the cytoskeleton, but its assembly is blocked at the unit-length filament (ULF) stage. ULFs are easy to track, and they allow a reliable and quantifiable analysis of movement. Our results show that in cultured human vimentin-negative SW13 cells, 2% of vimentin-ULFs move along microtubules bidirectionally, while the majority are stationary and tightly associated with actin filaments. Rapid motor-dependent transport of ULFs along microtubules is enhanced ≥ 5-fold by depolymerization of actin cytoskeleton with latrunculin B. The microtubule-dependent transport of vimentin ULFs is further regulated by Rho-kinase (ROCK) and p21-activated kinase (PAK): ROCK inhibits ULF transport, while PAK stimulates it. Both kinases act on microtubule transport independently of their effects on actin cytoskeleton. Our study demonstrates the importance of the actin cytoskeleton to restrict IF transport and reveals a new role for PAK and ROCK in the regulation of IF precursor transport.-Robert, A., Herrmann, H., Davidson, M. W., and Gelfand, V. I. Microtubule-dependent transport of vimentin filament precursors is regulated by actin and by the concerted action of Rho- and p21-activated kinases.
Collapse
Affiliation(s)
- Amélie Robert
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Harald Herrmann
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany; and
| | - Michael W Davidson
- National High Magnetic Field Laboratory and Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA;
| |
Collapse
|