1
|
Pérez-Sala D, Zorrilla S. Versatility of vimentin assemblies: From filaments to biomolecular condensates and back. Eur J Cell Biol 2025; 104:151487. [PMID: 40194320 DOI: 10.1016/j.ejcb.2025.151487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Cytoskeletal structures shape and confer resistance to cells. The intermediate filament protein vimentin forms versatile structures that play key roles in cytoskeletal crosstalk, in the integration of cellular responses to a variety of external and internal cues, and in the defense against stress. Such multifaceted roles can be fulfilled thanks to the vast variety of vimentin proteoforms, which in turn arise from the combinations of a myriad of tightly regulated posttranslational modifications. Diverse vimentin proteoforms will differentially shape its polymeric assemblies, underlying vimentin ability to organize in filaments, bundles, squiggles, droplets, cell surface-bound and/or various secreted forms. Interestingly, certain vimentin dots or droplets have been lately categorized as biomolecular condensates. Biomolecular condensates are phase-separated membraneless structures that are critical for the organization of cellular components and play important roles in pathophysiology. Recent findings have unveiled the importance of low complexity sequence domains in vimentin filament assembly. Moreover, several oxidants trigger the transition of vimentin filaments into phase-separated biomolecular condensates, a reversible process that may provide clues on the role of condensates as seeds for filament formation. Revisiting previous results in the light of recent knowledge prompts the hypothesis that vimentin condensates could play a role in traffic of filament precursors, cytoskeletal crosstalk and cellular responses to stress. Deciphering the "vimentin posttranslational modification code", that is, the structure-function relationships of vimentin proteoforms, constitutes a major challenge to understand the regulation of vimentin behavior and its multiple personalities. This will contribute to unveil essential cellular mechanisms and foster novel opportunities for drug discovery.
Collapse
Affiliation(s)
- Dolores Pérez-Sala
- Department of Molecular and Cellular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain.
| | - Silvia Zorrilla
- Department of Molecular and Cellular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| |
Collapse
|
2
|
Bodegraven EJ, Sluijs JA, Tan AK, Robe PAJT, Hol EM. New GFAP splice isoform (GFAPµ) differentially expressed in glioma translates into 21 kDa N‐terminal GFAP protein. FASEB J 2021; 35:e21389. [DOI: 10.1096/fj.202001767r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Emma J. Bodegraven
- Department of Translational Neurosciences University Medical Center Utrecht Brain CenterUtrecht University Utrecht The Netherlands
| | - Jacqueline A. Sluijs
- Department of Translational Neurosciences University Medical Center Utrecht Brain CenterUtrecht University Utrecht The Netherlands
| | - A. Katherine Tan
- Department of Translational Neurosciences University Medical Center Utrecht Brain CenterUtrecht University Utrecht The Netherlands
- Department of Neurology and Neurosurgery University Medical Center Utrecht Brain CenterUtrecht University Utrecht The Netherlands
| | - Pierre A. J. T. Robe
- Department of Neurology and Neurosurgery University Medical Center Utrecht Brain CenterUtrecht University Utrecht The Netherlands
| | - Elly M. Hol
- Department of Translational Neurosciences University Medical Center Utrecht Brain CenterUtrecht University Utrecht The Netherlands
| |
Collapse
|
3
|
Cytoskeletal Organization Correlates to Motility and Invasiveness of Malignant Mesothelioma Cells. Cancers (Basel) 2021; 13:cancers13040685. [PMID: 33567673 PMCID: PMC7915464 DOI: 10.3390/cancers13040685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary The cytoskeleton is responsible for maintaining normal tissue homeostasis by a tight regulation of cell morphogenesis and cell migration. This homeostasis is lost in cancer mainly because alterations in cytoskeletal dynamics are leading to an increased migratory and invasive capacity of cancer cells. The organization of the cytoskeleton is by large an unknown factor in malignant mesothelioma; therefore we sought to examine the cytoskeletal dynamics and invasive properties of different malignant mesothelioma cell lines originating from patients. Our data suggest that it is possible to classify malignant mesothelioma cell lines into separate categories using straight forward cell staining and analysis of the morphological and invasive capacity of mesothelioma cells. Early diagnosis and new diagnostic tools are urgently needed to effectively treat patients and we propose that the analyses described in this article could potentially provide diagnostic tools that can be further tested on patients. Abstract Malignant mesothelioma (MM) is a rare but highly aggressive cancer that primarily originates from the pleura, peritoneum or pericardium. There is a well-established link between asbestos exposure and progression of MM. Direct invasion of the surrounding tissues is the main feature of MM, which is dependent on dysregulated communication between the mesothelium and the microenvironment. This communication is dependent on the dynamic organization of the cytoskeleton. We have analyzed the organization and function of key cytoskeletal components in MM cell lines of increasing malignancies measured as migratory and invasive properties, and we show that highly malignant and invasive MM cells have an organization of the actin filament and vimentin systems that is distinct from the less malignant MM cell lines. In addition, the Hippo tumor suppressor pathway was inactivated in the invasive MM cells, which was seen as increased YAP nuclear localization.
Collapse
|
4
|
Reis K, Arbiser JL, Hjerpe A, Dobra K, Aspenström P. Inhibitors of cytoskeletal dynamics in malignant mesothelioma. Oncotarget 2020; 11:4637-4647. [PMID: 33400741 PMCID: PMC7747860 DOI: 10.18632/oncotarget.27843] [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: 10/20/2020] [Accepted: 11/30/2020] [Indexed: 11/25/2022] Open
Abstract
Malignant mesotheliomas (MMs) are highly aggressive mesenchymal tumors that originate from mesothelial cells lining serosal cavities; i.e., the pleura, peritoneum, and pericardium. Classically, there is a well-established link between asbestos exposure, oxidative stress, release of reactive oxygen species, and chronic inflammatory mediators that leads to progression of MMs. MMs have an intermediate phenotype, with co-expression of mesenchymal and epithelial markers and dysregulated communication between the mesothelium and the microenvironment. We have previously shown that the organization and function of key cytoskeletal components can distinguish highly invasive cell lines from those more indolent. Here, we used these tools to study three different types of small-molecule inhibitors, where their common feature is their influence on production of reactive oxygen species. One of these, imipramine blue, was particularly effective in counteracting some key malignant properties of highly invasive MM cells. This opens a new possibility for targeted inhibition of MMs based on well-established molecular mechanisms.
Collapse
Affiliation(s)
- Katarina Reis
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta Veterans Administration Medical Center, Atlanta, GA, USA
| | - Anders Hjerpe
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Katalin Dobra
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Pontus Aspenström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
5
|
Pinto-Costa R, Sousa MM. Profilin as a dual regulator of actin and microtubule dynamics. Cytoskeleton (Hoboken) 2019; 77:76-83. [PMID: 31811707 DOI: 10.1002/cm.21586] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/18/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022]
Abstract
Although originally identified as G-actin sequestering proteins, profilins are emerging as critical regulators of actin dynamics, capable of interacting with multiple acting binding proteins, and being able to link membrane lipids to cytoskeleton components. Recently, in addition to its actin, poly-proline, and phosphatidylinositol binding domains, profilin has been shown to contain residues specialized in microtubule binding. Here we will discuss in a critical perspective the emerging body of data supporting that profilins are central mediators of actin microfilament and microtubule interaction. We will also address the unanswered questions in the field, including the nature of the interaction of profilin with microtubules, and its effect on microtubule dynamics. These recent discoveries deepen our understanding on how different cytoskeleton components are integrated within cells.
Collapse
Affiliation(s)
- Rita Pinto-Costa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Porto, Portugal.,Graduate Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar-ICBAS, University of Porto, Porto, Portugal
| | - Mónica M Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
6
|
Kaschula CH, Tuveri R, Ngarande E, Dzobo K, Barnett C, Kusza DA, Graham LM, Katz AA, Rafudeen MS, Parker MI, Hunter R, Schäfer G. The garlic compound ajoene covalently binds vimentin, disrupts the vimentin network and exerts anti-metastatic activity in cancer cells. BMC Cancer 2019; 19:248. [PMID: 30894168 PMCID: PMC6425727 DOI: 10.1186/s12885-019-5388-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/20/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Garlic has been used for centuries for its flavour and health promoting properties that include protection against cancer. The vinyl disulfide-sulfoxide ajoene is one of the phytochemicals found in crushed cloves, hypothesised to act by S-thiolating reactive cysteines in target proteins. METHODS Using our fluorescently labelled ajoene analogue called dansyl-ajoene, ajoene's protein targets in MDA-MB-231 breast cancer cells were tagged and separated by 2D electrophoresis. A predominant band was identified by MALDI-TOF MS/MS to be vimentin. Target validation experiments were performed using pure recombinant vimentin protein. Computational modelling of vimentin bound to ajoene was performed using Schrödinger and pKa calculations by Epik software. Cytotoxicity of ajoene in MDA-MB-231 and HeLa cells was measured by the MTT assay. The vimentin filament network was visualised in ajoene-treated and non-treated cells by immunofluorescence and vimentin protein expression was determined by immunoblot. The invasion and migration activity was measured by wound healing and transwell assays using wildtype cells and cells in which the vimentin protein had been transiently knocked down by siRNA or overexpressed. RESULTS The dominant protein tagged by dansyl-ajoene was identified to be the 57 kDa protein vimentin. The vimentin target was validated to reveal that ajoene and dansyl-ajoene covalently bind to recombinant vimentin via a disulfide linkage at Cys-328. Computational modelling showed Cys-328 to be exposed at the termini of the vimentin tetramer. Treatment of MDA-MB-231 or HeLa cells with a non-cytotoxic concentration of ajoene caused the vimentin filament network to condense; and to increase vimentin protein expression. Ajoene inhibited the invasion and migration of both cancer cell lines which was found to be dependent on the presence of vimentin. Vimentin overexpression caused cells to become more migratory, an effect that was completely rescued by ajoene. CONCLUSIONS The garlic-derived phytochemical ajoene targets and covalently modifies vimentin in cancer cells by S-thiolating Cys-328. This interaction results in the disruption of the vimentin filament network and contributes to the anti-metastatic activity of ajoene in cancer cells.
Collapse
Affiliation(s)
- Catherine H. Kaschula
- Department of Chemistry and Polymer Science, Stellenbosch University, Stellenbosch, 7600 South Africa
| | - Rosanna Tuveri
- Department of Biomedical Science, University of Cagliari, 09042 Monserrato, Italy
| | - Ellen Ngarande
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Kevin Dzobo
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), UCT Medical Campus, Anzio Rd, Observatory, Cape Town, 7925 South Africa
| | - Christopher Barnett
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - Daniel A. Kusza
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - Lisa M. Graham
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Arieh A. Katz
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Mohamed Suhail Rafudeen
- Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - M. Iqbal Parker
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| | - Roger Hunter
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700 South Africa
| | - Georgia Schäfer
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925 South Africa
| |
Collapse
|
7
|
Walker JL, Bleaken BM, Romisher AR, Alnwibit AA, Menko AS. In wound repair vimentin mediates the transition of mesenchymal leader cells to a myofibroblast phenotype. Mol Biol Cell 2018; 29:1555-1570. [PMID: 29718762 PMCID: PMC6080657 DOI: 10.1091/mbc.e17-06-0364] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Following injury, mesenchymal repair cells are activated to function as leader cells that modulate wound healing. These cells have the potential to differentiate to myofibroblasts, resulting in fibrosis and scarring. The signals underlying these differing pathways are complex and incompletely understood. The ex vivo mock cataract surgery cultures are an attractive model with which to address this question. With this model we study, concurrently, the mechanisms that control mesenchymal leader cell function in injury repair within their native microenvironment and the signals that induce this same cell population to acquire a myofibroblast phenotype when these cells encounter the environment of the adjacent tissue culture platform. Here we show that on injury, the cytoskeletal protein vimentin is released into the extracellular space, binds to the cell surface of the mesenchymal leader cells located at the wound edge in the native matrix environment, and supports wound closure. In profibrotic environments, the extracellular vimentin pool also links specifically to the mesenchymal leader cells and has an essential role in signaling their fate change to a myofibroblast. These findings suggest a novel role for extracellular, cell-surface–associated vimentin in mediating repair-cell function in wound repair and in transitioning these cells to a myofibroblast phenotype.
Collapse
Affiliation(s)
- J L Walker
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - B M Bleaken
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A R Romisher
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A A Alnwibit
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A S Menko
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
8
|
Emerging Role of Nestin as an Angiogenesis and Cancer Stem Cell Marker in Epithelial Ovarian Cancer: Immunohistochemical Study. Appl Immunohistochem Mol Morphol 2018; 25:571-580. [PMID: 26945442 DOI: 10.1097/pai.0000000000000338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the most fatal gynecologic malignancy and the existing second-line treatments have not been confirmed to be effective. Cancer stem cells research has a leading role to explore promising therapeutic applications. Nestin was postulated to reflect cancer stem cell properties in various tumors, correlating with poor prognosis. Furthermore, nestin is proposed as a reliable neovascularization marker. This study aimed to elucidate the status of nestin expression in various epithelial ovarian cancers (EOCs), its neoangiogenic properties, and investigate its potential association with clinicopathologic parameters. A total of 80 primary EOCs (37 serous, 20 Mucinous, 13 endometrioid, and 10 clear cell carcinomas) were immunohistochemically stained with nestin. Staining intensity and automated microvascular density (MVD) were assessed. Positive nestin expression was defined in ≈47.5% of all EOC; more commonly in ≈60% of the serous tumors. It was noticeably expressed in tumor spheroids. Nestin expression significantly correlated with overall tumor grade, lymph node, distant metastasis, and stage. Nestin neoangiogenesis was detectable in all cases (average=60.1). The nestin expression in tumor cells significantly correlated with Nestin/MVD. The average Nestin/MVD was significantly an independent predictor of high tumor stage. As a stem cell marker, nestin is expressed in cells of EOC including those growing as spherules and correlated with poor prognosis. Thus, nestin may be a novel therapeutic target for tumor angiogenesis and a combination therapy that includes nestin-targeting agents may be an effective therapeutic approach. In addition, detection of Nestin/stem cells and Nestin/MVD can be used as predictors of disease.
Collapse
|
9
|
Li SS, Xu LZ, Zhou W, Yao S, Wang CL, Xia JL, Wang HF, Kamran M, Xue XY, Dong L, Wang J, Ding XD, Bella L, Bugeon L, Xu J, Zheng FM, Dallman MJ, Lam EWF, Liu Q. p62/SQSTM1 interacts with vimentin to enhance breast cancer metastasis. Carcinogenesis 2017; 38:1092-1103. [PMID: 28968743 PMCID: PMC5862327 DOI: 10.1093/carcin/bgx099] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 09/11/2017] [Indexed: 11/23/2022] Open
Abstract
The signalling adaptor p62 is frequently overexpressed in numerous cancer types. Here, we found that p62 expression was elevated in metastatic breast cancer and its overexpression correlated with reduced metastasis- and relapse-free survival times. Analysis of p62 expression in breast cancer cell lines demonstrated that high p62 expression was associated with the invasive phenotypes of breast cancer. Indeed, silencing p62 expression attenuated the invasive phenotypes of highly metastatic cells, whereas overexpressing p62 promoted the invasion of non-metastatic cells in in vitro microfluidic model. Moreover, MDA-MB-231 cells with p62 depletion which were grown in a three-dimensional culture system exhibited a loss of invasive protrusions. Consistently, genetic ablation of p62 suppressed breast cancer metastasis in both zebrafish embryo and immunodeficient mouse models, as well as decreased tumourigenicity in vivo. To explore the molecular mechanism by which p62 promotes breast cancer invasion, we performed a co-immunoprecipitation–mass spectrometry analysis and revealed that p62 interacted with vimentin, which mediated the function of p62 in promoting breast cancer invasion. Vimentin protein expression was downregulated upon p62 suppression and upregulated with p62 overexpression in breast cancer cells. Linear regression analysis of clinical breast cancer specimens showed a positive correlation between p62 and vimentin protein expression. Together, our findings provide strong evidence that p62 functions as a tumour metastasis promoter by binding vimentin and promoting its expression. This finding might help to develop novel molecular therapeutic strategies for breast cancer metastasis treatment.
Collapse
Affiliation(s)
- Si-Si Li
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ling-Zhi Xu
- Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Zhou
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shang Yao
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Chun-Li Wang
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiang-Long Xia
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - He-Fei Wang
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Muhammad Kamran
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Yuan Xue
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lin Dong
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing Wang
- Department of Oncology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xu-Dong Ding
- Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Laura Bella
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Laurence Bugeon
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Jie Xu
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fei-Meng Zheng
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Margaret J Dallman
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Eric W F Lam
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Quentin Liu
- Cancer Center, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
10
|
Abstract
A mechanism of intermediate filament disassembly regulation is proposed in which disassembly is regulated by the amount of proteins assembled in networks. It is also hypothesized that a delay might exist between regulation and actual disassembly. Under realistic biological conditions of assembly and disassembly, it is shown that such a delay is harmless and does not destabilize the organization of intermediate filaments in networks. However, for high rates of disassembly, the model predicts that delay can destabilize the organization, with the intermediate filament material oscillating between organizations mainly in networks and in nonfilamentous particles.
Collapse
Affiliation(s)
- Chengjun Sun
- School of Management and Economics, Kunming University of Science and Technology, Kunming, P. R. China
- Department of Mechanical Engineering, University of Hong Kong, Pokfulam Road, Hong Kong
| | - Julien Arino
- Department of Mathematics, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Stéphanie Portet
- Department of Mathematics, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| |
Collapse
|
11
|
Matsuda Y, Ishiwata T, Yoshimura H, Yamashita S, Ushijima T, Arai T. Systemic Administration of Small Interfering RNA Targeting Human Nestin Inhibits Pancreatic Cancer Cell Proliferation and Metastasis. Pancreas 2016; 45:93-100. [PMID: 26335012 DOI: 10.1097/mpa.0000000000000427] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Nestin, a progenitor/stem cell marker, is expressed in human pancreatic cancer, where its expression correlates positively with invasiveness and metastasis. Here, we investigated the inhibition of nestin expression and the regulation of nestin expression. METHODS We analyzed the effects of small interfering RNA (siRNA) targeting nestin using pancreatic cancer cell lines. RESULTS Nestin siRNA inhibited the growth, migration, invasion, and sphere-forming ability of the pancreatic cancer cell lines. Pancreatic cancer cells cotreated with gemcitabine and nestin siRNA exhibited lower cell viability than cells treated with a control siRNA, gemcitabine alone, or nestin siRNA alone. Cells derived from the metastatic nodules of mice showed higher nestin expression than the parental cells, and nestin expression in pancreatic cancer cells was regulated by methylation of the nestin gene. In an orthotopic implantation model using mice, administration of nestin siRNA significantly decreased primary and metastatic tumor formation by human pancreatic cancer cells compared to tumor formation in control siRNA-treated mice. CONCLUSIONS Nestin plays a key role in pancreatic cancer cell metastasis and stemness and that administration of nestin siRNA may offer a novel therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Yoko Matsuda
- From the *Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Itabashi-ku; †Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, Bunkyo-ku; and ‡Division of Epigenomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
12
|
Bargagna-Mohan P, Lei L, Thompson A, Shaw C, Kasahara K, Inagaki M, Mohan R. Vimentin Phosphorylation Underlies Myofibroblast Sensitivity to Withaferin A In Vitro and during Corneal Fibrosis. PLoS One 2015; 10:e0133399. [PMID: 26186445 PMCID: PMC4506086 DOI: 10.1371/journal.pone.0133399] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 06/26/2015] [Indexed: 12/21/2022] Open
Abstract
Vimentin is a newly recognized target for corneal fibrosis. Using primary rabbit corneal fibroblasts and myofibroblasts we show that myofibroblasts, unlike fibroblasts, display impaired cell spreading and cell polarization, which is associated with increased levels of soluble serine-38 phosphorylated vimentin (pSer38Vim). This pSer38Vim isoform is inefficiently incorporated into growing vimentin intermediate filaments (IFs) of myofibroblasts during cell spreading, and as a result, myofibroblasts maintain higher soluble pSer38Vim levels compared to fibroblasts. Moreover, the soluble vimentin-targeting small molecule and fibrotic inhibitor withaferin A (WFA) causes a potent blockade of cell spreading selectively in myofibroblasts by targeting soluble pSer38Vim for hyperphosphorylation. WFA treatment does not induce vimentin hyperphosphorylation in fibroblasts. This hyperphosphorylated pSer38Vim species in WFA-treated myofibroblasts becomes complexed with adaptor protein filamin A (FlnA), and these complexes appear as short squiggles when displaced from focal adhesions. The extracellular-signal regulated kinase (ERK) is also phosphorylated (pERK) in response to WFA, but surprisingly, pERK does not enter the nucleus but remains bound to pSer38Vim in cytoplasmic complexes. Using a model of corneal alkali injury, we show that fibrotic corneas of wild type mice possess high levels of pERK, whereas injured corneas of vimentin-deficient (Vim KO) mice that heal with reduced fibrosis have highly reduced pERK expression. Finally, WFA treatment causes a decrease in pERK and pSer38Vim expression in healing corneas of wild type mice. Taken together, these findings identify a hereto-unappreciated role for pSer38Vim as an important determinant of myofibroblast sensitivity to WFA.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Ling Lei
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Alexis Thompson
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Camille Shaw
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Kousuke Kasahara
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Masaki Inagaki
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Royce Mohan
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
13
|
Matsuda Y, Ishiwata T, Yoshimura H, Hagio M, Arai T. Inhibition of nestin suppresses stem cell phenotype of glioblastomas through the alteration of post-translational modification of heat shock protein HSPA8/HSC71. Cancer Lett 2015; 357:602-11. [DOI: 10.1016/j.canlet.2014.12.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 12/16/2022]
|
14
|
Al-Zeer MA, Al-Younes HM, Kerr M, Abu-Lubad M, Gonzalez E, Brinkmann V, Meyer TF. Chlamydia trachomatis remodels stable microtubules to coordinate Golgi stack recruitment to the chlamydial inclusion surface. Mol Microbiol 2014; 94:1285-97. [PMID: 25315131 DOI: 10.1111/mmi.12829] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2014] [Indexed: 12/31/2022]
Abstract
Chlamydia trachomatis (Ctr), an obligate intracellular bacterium, survives and replicates within a membrane-bound vacuole, termed the inclusion, which intercepts host exocytic pathways to acquire nutrients. Ctr subverts cellular trafficking pathways from the Golgi by targeting small GTPases, including Rab proteins, to sustain intracellular bacterial replication; however, the precise mechanisms involved remain incompletely understood. Here, we show that Chlamydia infection in human epithelial cells induces microtubule remodeling, in particular the formation of detyrosinated stable MTs, to recruit Golgi ministacks, but not recycling endosomes, to the inclusion. These stable microtubules show increased resistance to chemically induced depolymerization, and their selective depletion results in reduced bacterial infectivity. Rab6 knockdown reversibly prevented not only Golgi ministack formation but also detyrosinated microtubule association with the inclusion. Our data demonstrate that Chlamydia co-opts the function of stable microtubules to support its development.
Collapse
Affiliation(s)
- Munir A Al-Zeer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Charitèplatz 1, 10117, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Trendowski M. Exploiting the cytoskeletal filaments of neoplastic cells to potentiate a novel therapeutic approach. Biochim Biophys Acta Rev Cancer 2014; 1846:599-616. [PMID: 25286320 DOI: 10.1016/j.bbcan.2014.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 02/06/2023]
Abstract
Although cytoskeletal-directed agents have been a mainstay in chemotherapeutic protocols due to their ability to readily interfere with the rapid mitotic progression of neoplastic cells, they are all microtubule-based drugs, and there has yet to be any microfilament- or intermediate filament-directed agents approved for clinical use. There are many inherent differences between the cytoskeletal networks of malignant and normal cells, providing an ideal target to attain preferential damage. Further, numerous microfilament-directed agents, and an intermediate filament-directed agent of particular interest (withaferin A) have demonstrated in vitro and in vivo efficacy, suggesting that cytoskeletal filaments may be exploited to supplement chemotherapeutic approaches currently used in the clinical setting. Therefore, this review is intended to expose academics and clinicians to the tremendous variety of cytoskeletal filament-directed agents that are currently available for further chemotherapeutic evaluation. The mechanisms by which microfilament directed- and intermediate filament-directed agents damage malignant cells are discussed in detail in order to establish how the drugs can be used in combination with each other, or with currently approved chemotherapeutic agents to generate a substantial synergistic attack, potentially establishing a new paradigm of chemotherapeutic agents.
Collapse
Affiliation(s)
- Matthew Trendowski
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA.
| |
Collapse
|
16
|
Kraemer FB, Khor VK, Shen WJ, Azhar S. Cholesterol ester droplets and steroidogenesis. Mol Cell Endocrinol 2013; 371:15-9. [PMID: 23089211 PMCID: PMC3584206 DOI: 10.1016/j.mce.2012.10.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 10/05/2012] [Accepted: 10/11/2012] [Indexed: 12/12/2022]
Abstract
Intracellular lipid droplets (LDs) are dynamic organelles that contain a number of associated proteins including perilipin (Plin) and vimentin. Cholesteryl ester (CE)-rich LDs normally accumulate in steroidogenic cells and their mobilization is the preferred initial source of cholesterol for steroidogenesis. Plin1a, 1b and 5 were found to preferentially associate with triacylglycerol-rich LDs and Plin1c and Plin4 to associate with CE-rich LDs, but the biological significance of this remains unanswered. Vimentin null mice were found to have decreased ACTH-stimulated corticosterone levels, and decreased progesterone levels in females, but normal hCG-stimulated testosterone levels in males. Smaller LDs were seen in null cells. Lipoprotein cholesterol delivery to adrenals and ovary was normal, as was the expression of steroidogenic genes; however, the movement of cholesterol to mitochondria was reduced in vimentin null mice. These results suggest that vimentin is important in the maintenance of CE-rich LDs and in the movement of cholesterol for steroidogenesis.
Collapse
|
17
|
Bargagna-Mohan P, Deokule SP, Thompson K, Wizeman J, Srinivasan C, Vooturi S, Kompella UB, Mohan R. Withaferin A effectively targets soluble vimentin in the glaucoma filtration surgical model of fibrosis. PLoS One 2013; 8:e63881. [PMID: 23667686 PMCID: PMC3648549 DOI: 10.1371/journal.pone.0063881] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/09/2013] [Indexed: 01/23/2023] Open
Abstract
Withaferin A (WFA) is a natural product that binds to soluble forms of the type III intermediate filament (IF) vimentin. Currently, it is unknown under what pathophysiological contexts vimentin is druggable, as cytoskeltal vimentin-IFs are abundantly expressed. To investigate druggability of vimentin, we exploited rabbit Tenon's capsule fibroblast (RbTCF) cell cultures and the rabbit glaucoma filtration surgical (GFS) model of fibrosis. WFA potently caused G₀/G₁ cell cycle inhibition (IC₅₀ 25 nM) in RbTCFs, downregulating ubiquitin E3 ligase skp2 and inducing p27(Kip1) expression. Transforming growth factor (TGF)-ß-induced myofibroblast transformation caused development of cell spheroids with numerous elongated invadopodia, which WFA blocked potently by downregulating soluble vimentin and α-smooth muscle actin (SMA) expression. In the pilot proof-of-concept study using the GFS model, subconjunctival injections of a low WFA dose reduced skp2 expression in Tenon's capsule and increased p27(Kip1) expression without significant alteration to vimentin-IFs. This treatment maintains significant nanomolar WFA concentrations in anterior segment tissues that correspond to WFA's cell cycle targeting activity. A ten-fold higher WFA dose caused potent downregulation of soluble vimentin and skp2 expression, but as found in cell cultures, no further increase in p27(Kip1) expression was observed. Instead, this high WFA dose potently induced vimentin-IF disruption and downregulated α-SMA expression that mimicked WFA activity in TGF-ß-treated RbTCFs that blocked cell contractile activity at submicromolar concentrations. These findings illuminate that localized WFA injection to ocular tissues exerts pharmacological control over the skp2-p27(Kip1) pathway by targeting of soluble vimentin in a model of surgical fibrosis.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Sunil P. Deokule
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kyle Thompson
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - John Wizeman
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Cidambi Srinivasan
- Statistics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Sunil Vooturi
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Uday B. Kompella
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Royce Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
18
|
Shen WJ, Zaidi SK, Patel S, Cortez Y, Ueno M, Azhar R, Azhar S, Kraemer FB. Ablation of vimentin results in defective steroidogenesis. Endocrinology 2012; 153:3249-57. [PMID: 22535769 PMCID: PMC3380307 DOI: 10.1210/en.2012-1048] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In steroidogenic tissues, cholesterol must be transported to the inner mitochondrial membrane to be converted to pregnenolone as the first step of steroidogenesis. Whereas steroidogenic acute regulatory protein has been shown to be responsible for the transport of cholesterol from the outer to the inner mitochondrial membrane, the process of how cholesterol moves to mitochondria from the cytoplasm is not clearly defined. The involvement of the cytoskeleton has been suggested; however, no specific mechanism has been confirmed. In this paper, using genetic ablation of an intermediate filament protein in mice, we present data demonstrating a marked defect in adrenal and ovarian steroidogenesis in the absence of vimentin. Cosyntropin-stimulated corticosterone production is decreased 35 and 50% in male and female Vimentin null (Vim(-/-)) mice, respectively, whereas progesterone production is decreased 70% in female Vim(-/-) mice after pregnant mare's serum gonadotropin and human chorionic gonadotropin stimulation, but no abnormalities in human chorionic gonadotropin-stimulated testosterone production is observed in male Vim(-/-) mice. These defects in steroid production are also seen in isolated adrenal and granulosa cells in vitro. Further studies show a defect in the movement of cholesterol from the cytosol to mitochondria in Vim(-/-) cells. Because the mobilization of cholesterol from lipid droplets and its transport to mitochondria is a preferred pathway for the initiation of steroid production in the adrenal and ovary but not the testis and vimentin is a droplet-associated protein, our results suggest that vimentin is involved in the movement of cholesterol from its storage in lipid droplets to mitochondria for steroidogenesis.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Division of Endocrinology, Gerontology, and Metabolism, Stanford University, Palo Alto, California 94304, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Dráber P, Sulimenko V, Dráberová E. Cytoskeleton in mast cell signaling. Front Immunol 2012; 3:130. [PMID: 22654883 PMCID: PMC3360219 DOI: 10.3389/fimmu.2012.00130] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Accepted: 05/05/2012] [Indexed: 11/13/2022] Open
Abstract
Mast cell activation mediated by the high affinity receptor for IgE (FcεRI) is a key event in allergic response and inflammation. Other receptors on mast cells, as c-Kit for stem cell factor and G protein-coupled receptors (GPCRs) synergistically enhance the FcεRI-mediated release of inflammatory mediators. Activation of various signaling pathways in mast cells results in changes in cell morphology, adhesion to substrate, exocytosis, and migration. Reorganization of cytoskeleton is pivotal in all these processes. Cytoskeletal proteins also play an important role in initial stages of FcεRI and other surface receptors induced triggering. Highly dynamic microtubules formed by αβ-tubulin dimers as well as microfilaments build up from polymerized actin are affected in activated cells by kinases/phosphatases, Rho GTPases and changes in concentration of cytosolic Ca(2+). Also important are nucleation proteins; the γ-tubulin complexes in case of microtubules or Arp 2/3 complex with its nucleation promoting factors and formins in case of microfilaments. The dynamic nature of microtubules and microfilaments in activated cells depends on many associated/regulatory proteins. Changes in rigidity of activated mast cells reflect changes in intermediate filaments build up from vimentin. This review offers a critical appraisal of current knowledge on the role of cytoskeleton in mast cells signaling.
Collapse
Affiliation(s)
- Pavel Dráber
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech RepublicPrague, Czech Republic
| | - Vadym Sulimenko
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech RepublicPrague, Czech Republic
| | - Eduarda Dráberová
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech RepublicPrague, Czech Republic
| |
Collapse
|
20
|
Lee KY, Liu L, Jin Y, Fu SB, Rosales JL. Cdk5 mediates vimentin Ser56 phosphorylation during GTP-induced secretion by neutrophils. J Cell Physiol 2012; 227:739-50. [PMID: 21465480 DOI: 10.1002/jcp.22782] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Secretion by neutrophils contributes to acute inflammation following injury or infection. Vimentin has been shown to be important for secretion by neutrophils but little is known about its dynamics during secretion, which is regulated by cyclin-dependent kinase 5 (Cdk5). In this study, we sought to examine the vimentin dynamics and its potential regulation by Cdk5 during neutrophil secretion. We show that vimentin is a Cdk5 substrate that is specifically phosphorylated at Ser56. In response to neutrophil stimulation with GTP, vimentin Ser56 was phosphorylated and colocalized with Cdk5 in the cytoplasmic compartment. Vimentin pSer56 and Cdk5 colocalization was consistent with coimmunoprecipitation from stimulated cells. Vimentin Ser56 phosphorylation occurred immediately after stimulation, and a remarkable increase in phosphorylation was noted later in the secretory process. Decreased GTP-induced vimentin Ser56 phosphorylation and secretion resulted from inhibition of Cdk5 activity by roscovitine or olomoucine or by depletion of Cdk5 by siRNA, suggesting that GTP-induced Cdk5-mediated vimentin Ser56 phosphorylation may be related to GTP-induced Cdk5-mediated secretion by neutrophils. Indeed, inhibition of vimentin Ser56 phosphorylation led to a corresponding inhibition of GTP-induced secretion, indicating a link between these two events. While fMLP also induced vimentin Ser56 phosphorylation, such phosphorylation was unaffected by roscovitine, which nonetheless, inhibited secretion, suggesting that Cdk5 regulates fMLP-induced secretion via a mechanism independent of Cdk5-mediated vimentin Ser56 phosphorylation. These findings demonstrate the distinct involvement of Cdk5 in GTP- and fMLP-induced secretion by neutrophils, and support the notion that specific targeting of Cdk5 may serve to inhibit the neutrophil secretory process.
Collapse
Affiliation(s)
- Ki-Young Lee
- Department of Cell Biology & Anatomy, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
21
|
Bargagna-Mohan P, Paranthan RR, Hamza A, Zhan CG, Lee DM, Kim KB, Lau DL, Srinivasan C, Nakayama K, Nakayama KI, Herrmann H, Mohan R. Corneal antifibrotic switch identified in genetic and pharmacological deficiency of vimentin. J Biol Chem 2011; 287:989-1006. [PMID: 22117063 DOI: 10.1074/jbc.m111.297150] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The type III intermediate filaments (IFs) are essential cytoskeletal elements of mechanosignal transduction and serve critical roles in tissue repair. Mice genetically deficient for the IF protein vimentin (Vim(-/-)) have impaired wound healing from deficits in myofibroblast development. We report a surprising finding made in Vim(-/-) mice that corneas are protected from fibrosis and instead promote regenerative healing after traumatic alkali injury. This reparative phenotype in Vim(-/-) corneas is strikingly recapitulated by the pharmacological agent withaferin A (WFA), a small molecule that binds to vimentin and down-regulates its injury-induced expression. Attenuation of corneal fibrosis by WFA is mediated by down-regulation of ubiquitin-conjugating E3 ligase Skp2 and up-regulation of cyclin-dependent kinase inhibitors p27(Kip1) and p21(Cip1). In cell culture models, WFA exerts G(2)/M cell cycle arrest in a p27(Kip1)- and Skp2-dependent manner. Finally, by developing a highly sensitive imaging method to measure corneal opacity, we identify a novel role for desmin overexpression in corneal haze. We demonstrate that desmin down-regulation by WFA via targeting the conserved WFA-ligand binding site shared among type III IFs promotes further improvement of corneal transparency without affecting cyclin-dependent kinase inhibitor levels in Vim(-/-) mice. This dissociates a direct role for desmin in corneal cell proliferation. Taken together, our findings illuminate a previously unappreciated pathogenic role for type III IF overexpression in corneal fibrotic conditions and also validate WFA as a powerful drug lead toward anti-fibrosis therapeutic development.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Verma SK, Ganesan TS, Kishore U, Parker PJ. The tumor suppressor RASSF1A is a novel effector of small G protein Rap1A. Protein Cell 2011; 2:237-49. [PMID: 21468893 DOI: 10.1007/s13238-011-1028-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 03/13/2011] [Indexed: 10/25/2022] Open
Abstract
Rap1A is a small G protein implicated in a spectrum of biological processes such as cell proliferation, adhesion, differentiation, and embryogenesis. The downstream effectors through which Rap1A mediates its diverse effects are largely unknown. Here we show that Rap1A, but not the related small G proteins Rap2 or Ras, binds the tumor suppressor Ras association domain family 1A (RASSF1A) in a manner that is regulated by phosphorylation of RASSF1A. Interaction with Rap1A is shown to influence the effect of RASSF1A on microtubule behavior.
Collapse
Affiliation(s)
- Sunil K Verma
- Department of Medical Oncology, Medical Sciences Division, The University of Oxford, Oxford, UK
| | | | | | | |
Collapse
|
23
|
Bayless KJ, Johnson GA. Role of the cytoskeleton in formation and maintenance of angiogenic sprouts. J Vasc Res 2011; 48:369-85. [PMID: 21464572 DOI: 10.1159/000324751] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 01/10/2011] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is the formation of new blood vessels from pre-existing structures, and is a key step in tissue and organ development, wound healing and pathological events. Changes in cell shape orchestrated by the cytoskeleton are integral to accomplishing the various steps of angiogenesis, and an intact cytoskeleton is also critical for maintaining newly formed structures. This review focuses on how the 3 main cytoskeletal elements--microfilaments, microtubules, and intermediate filaments--regulate the formation and maintenance of angiogenic sprouts. Multiple classes of compounds target microtubules and microfilaments, revealing much about the role of actin and tubulin and their associated molecules in angiogenic sprout formation and maintenance. In contrast, intermediate filaments are much less studied, yet intriguing evidence suggests a vital, but unresolved, role in angiogenic sprouting. This review discusses evidence for regulatory molecules and pharmacological compounds that affect actin, microtubule and intermediate filament dynamics to alter various steps of angiogenesis, including endothelial sprout formation and maintenance.
Collapse
Affiliation(s)
- Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, USA.
| | | |
Collapse
|
24
|
Matsuda Y, Naito Z, Kawahara K, Nakazawa N, Korc M, Ishiwata T. Nestin is a novel target for suppressing pancreatic cancer cell migration, invasion and metastasis. Cancer Biol Ther 2011; 11:512-23. [PMID: 21258211 DOI: 10.4161/cbt.11.5.14673] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nestin, is a class VI intermediate filament (IF) that is expressed in 30% of pancreatic ductal adenocarcinoma (PDAC) cases, and its expression in PDAC positively correlates with peripancreatic invasion. An expression vector carrying a short hairpin RNA (shRNA) targeting nestin was stably transfected into PANC-1 and PK-45H human pancreatic cancer cells, which express high nestin levels. Alterations in morphology and alignment of actin filaments and α-tubulin were examined by phase-contrast and immunocytochemistry. Effects on cell growth, migration in scratch and Boyden chamber assays, invasion, cell adhesion, and in vivo growth were determined. Differences in mRNA levels were examined by arrays. Nestin shRNA-transfected cells exhibited decreased nestin expression, a sheet-like appearance with tight cell-cell adhesion, increased expression of filamentous F-actin and E-cadherin, and attenuated migration and invasion, both of which were enhanced following nestin re-expression. Expression of α-tubulin, and in vitro cell growth and adhesion were not altered by nestin down-regulation, whereas hepatic metastases were decreased. Thus, nestin plays important roles in pancreatic cancer cell migration, invasion and metastasis by selectively modulating the expression of actin and cell adhesion molecules, and may therefore be a novel therapeutic target in PDAC.
Collapse
Affiliation(s)
- Yoko Matsuda
- Department of Pathology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Cellular cholesterol delivery, intracellular processing and utilization for biosynthesis of steroid hormones. Nutr Metab (Lond) 2010; 7:47. [PMID: 20515451 PMCID: PMC2890697 DOI: 10.1186/1743-7075-7-47] [Citation(s) in RCA: 321] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 06/01/2010] [Indexed: 11/28/2022] Open
Abstract
Steroid hormones regulate diverse physiological functions such as reproduction, blood salt balance, maintenance of secondary sexual characteristics, response to stress, neuronal function and various metabolic processes. They are synthesized from cholesterol mainly in the adrenal gland and gonads in response to tissue-specific tropic hormones. These steroidogenic tissues are unique in that they require cholesterol not only for membrane biogenesis, maintenance of membrane fluidity and cell signaling, but also as the starting material for the biosynthesis of steroid hormones. It is not surprising, then, that cells of steroidogenic tissues have evolved with multiple pathways to assure the constant supply of cholesterol needed to maintain optimum steroid synthesis. The cholesterol utilized for steroidogenesis is derived from a combination of sources: 1) de novo synthesis in the endoplasmic reticulum (ER); 2) the mobilization of cholesteryl esters (CEs) stored in lipid droplets through cholesteryl ester hydrolase; 3) plasma lipoprotein-derived CEs obtained by either LDL receptor-mediated endocytic and/or SR-BI-mediated selective uptake; and 4) in some cultured cell systems from plasma membrane-associated free cholesterol. Here, we focus on recent insights into the molecules and cellular processes that mediate the uptake of plasma lipoprotein-derived cholesterol, events connected with the intracellular cholesterol processing and the role of crucial proteins that mediate cholesterol transport to mitochondria for its utilization for steroid hormone production. In particular, we discuss the structure and function of SR-BI, the importance of the selective cholesterol transport pathway in providing cholesterol substrate for steroid biosynthesis and the role of two key proteins, StAR and PBR/TSO in facilitating cholesterol delivery to inner mitochondrial membrane sites, where P450scc (CYP11A) is localized and where the conversion of cholesterol to pregnenolone (the common steroid precursor) takes place.
Collapse
|
26
|
Lahat G, Zhu QS, Huang KL, Wang S, Bolshakov S, Liu J, Torres K, Langley RR, Lazar AJ, Hung MC, Lev D. Vimentin is a novel anti-cancer therapeutic target; insights from in vitro and in vivo mice xenograft studies. PLoS One 2010; 5:e10105. [PMID: 20419128 PMCID: PMC2855704 DOI: 10.1371/journal.pone.0010105] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2009] [Accepted: 03/03/2010] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Vimentin is a ubiquitous mesenchymal intermediate filament supporting mechano-structural integrity of quiescent cells while participating in adhesion, migration, survival, and cell signaling processes via dynamic assembly/disassembly in activated cells. Soft tissue sarcomas and some epithelial cancers exhibiting "epithelial to mesenchymal transition" phenotypes express vimentin. Withaferin-A, a naturally derived bioactive compound, may molecularly target vimentin, so we sought to evaluate its effects on tumor growth in vitro and in vivo thereby elucidating the role of vimentin in drug-induced responses. METHODS AND FINDINGS Withaferin-A elicited marked apoptosis and vimentin cleavage in vimentin-expressing tumor cells but significantly less in normal mesenchymal cells. This proapoptotic response was abrogated after vimentin knockdown or by blockade of caspase-induced vimentin degradation via caspase inhibitors or overexpression of mutated caspase-resistant vimentin. Pronounced anti-angiogenic effects of Withaferin-A were demonstrated, with only minimal effects seen in non-proliferating endothelial cells. Moreover, Withaferin-A significantly blocked soft tissue sarcoma growth, local recurrence, and metastasis in a panel of soft tissue sarcoma xenograft experiments. Apoptosis, decreased angiogenesis, and vimentin degradation were all seen in Withaferin-A treated specimens. CONCLUSIONS In light of these findings, evaluation of Withaferin-A, its analogs, or other anti-vimentin therapeutic approaches in soft tissue sarcoma and "epithelial to mesenchymal transition" clinical contexts is warranted.
Collapse
Affiliation(s)
- Guy Lahat
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Quan-Sheng Zhu
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Kai-Lieh Huang
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Suizhao Wang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Svetlana Bolshakov
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Jeffery Liu
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Keila Torres
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Robert R. Langley
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Alexander J. Lazar
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Mien Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Dina Lev
- Sarcoma Research Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
27
|
Shen WJ, Patel S, Eriksson JE, Kraemer FB. Vimentin is a functional partner of hormone sensitive lipase and facilitates lipolysis. J Proteome Res 2010; 9:1786-94. [PMID: 20143880 PMCID: PMC2849902 DOI: 10.1021/pr900909t] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lipolysis involves a number of components including signaling pathways, droplet-associated proteins, and lipases such as hormone-sensitive lipase (HSL). We used surface enhanced laser desorption/ionization time-of-flight mass spectroscopy to identify cellular proteins that might interact with HSL and potentially influence lipolysis. Using recombinant HSL as bait on protein chips, clusters of proteins of 14.7-18.9, 25.8-26.8, 36.1, 44.3-49.1, and 53.7 kDa were identified that interact with HSL, particularly when lysates were examined from beta-agonist treated mouse adipocytes. The ability to detect these interacting proteins was markedly diminished when the adipocytes were treated with insulin. A very similar pattern of proteins was identified when anti-HSL IgG was used as the bait. Following immunocapture, the identification of the prominent 53.7 kDa protein was carried out by tryptic digestion and MS analysis and determined to be vimentin. The interaction of HSL with vimentin, and its hormonal dependence, was confirmed by coimmunoprecipitation. beta-Agonist stimulated lipolysis and the rate of HSL translocation were impaired in vimentin null adipocytes, even though normal amounts of lipases and droplet-associated proteins are expressed. The current studies provide evidence that vimentin participates in lipolysis through direct, hormonally regulated interactions with HSL.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Division of Endocrinology, Stanford University and VA Palo Alto Health Care System, Palo Alto, CA 94304
| | - Shailja Patel
- Division of Endocrinology, Stanford University and VA Palo Alto Health Care System, Palo Alto, CA 94304
| | | | - Fredric B. Kraemer
- Division of Endocrinology, Stanford University and VA Palo Alto Health Care System, Palo Alto, CA 94304
| |
Collapse
|
28
|
Liang Y, Niederstrasser H, Edwards M, Jackson CE, Cooper JA. Distinct roles for CARMIL isoforms in cell migration. Mol Biol Cell 2010; 20:5290-305. [PMID: 19846667 DOI: 10.1091/mbc.e08-10-1071] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Molecular mechanisms for cell migration, especially how signaling and cytoskeletal systems are integrated, are not understood well. Here, we examined the role of CARMIL (capping protein, Arp2/3, and Myosin-I linker) family proteins in migrating cells. Vertebrates express three conserved genes for CARMIL, and we examined the functions of the two CARMIL genes expressed in migrating human cultured cells. Both isoforms, CARMIL1 and 2, were necessary for cell migration, but for different reasons. CARMIL1 localized to lamellipodia and macropinosomes, and loss of its function caused loss of lamellipodial actin, along with defects in protrusion, ruffling, and macropinocytosis. CARMIL1-knockdown cells showed loss of activation of Rac1, and CARMIL1 was biochemically associated with the GEF Trio. CARMIL2, in contrast, colocalized with vimentin intermediate filaments, and loss of its function caused a distinctive multipolar phenotype. Loss of CARMIL2 also caused decreased levels of myosin-IIB, which may contribute to the polarity phenotype. Expression of one CARMIL isoform was not able to rescue the knockdown phenotypes of the other. Thus, the two isoforms are both important for cell migration, but they have distinct functions.
Collapse
Affiliation(s)
- Yun Liang
- Department of Cell Biology and Physiology, Washington University, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
29
|
Particle tracking of intracellular trafficking of octaarginine-modified liposomes: a comparative study with adenovirus. Mol Ther 2010; 18:955-64. [PMID: 20216528 DOI: 10.1038/mt.2010.33] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is previously reported that octaarginine (R8)-modified liposome (R8-Lip) was taken up via macropinocytosis, and subsequently delivered to the nuclear periphery. In the present study, we investigated the mechanism for the cytoplasmic transport of R8-Lips, comparing with that for adenovirus. Treatment with microtubule-disruption reagent (nocodazole) inhibited the transfection activity of plasmid DNA (pDNA)-encapsulating R8-Lip more extensively than that of adenovirus. The directional transport of R8-Lips along green fluorescent protein (GFP)-tagged microtubules was observed; however, the velocity was slower than those for adenovirus or endosomes that were devoid of R8-Lips. These directional motions were abrogated in R8-Lips by nocodazole treatment, whereas adenovirus continued to undergo random motion. This finding suggests that the nuclear access of R8-Lip predominantly involves microtubule-dependent transport, whereas an apparent diffusive motion is also operative in nuclear access of adenovirus. Furthermore, quantum dot-labeled pDNA underwent directional motion concomitantly with rhodamine-labeled lipid envelopes, indicating that the R8-Lips were subject to microtubule-dependent transport in the intact form. Dual particle tracking of carriers and endosomes revealed that R8-Lip was directionally transported, associated with endosomes, whereas this occurs after endosomal escape in adenovirus. Collectively, the findings reported herein indicate that vesicular transport is a key factor in the cytoplasmic transport of R8-Lips.
Collapse
|
30
|
Johnsson AK, Karlsson R. Microtubule-dependent localization of profilin I mRNA to actin polymerization sites in serum-stimulated cells. Eur J Cell Biol 2010; 89:394-401. [PMID: 20129697 DOI: 10.1016/j.ejcb.2009.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 10/29/2009] [Accepted: 10/29/2009] [Indexed: 12/12/2022] Open
Abstract
Specific localization of messenger RNA (mRNA) appears to be a general mechanism to accumulate certain proteins to subcellular compartments for participation in local processes, thereby maintaining cell polarity under strict spatiotemporal control. Transportation of mRNA with associated protein components (RNP granules) by the actin microfilament or the microtubule systems is one important mechanism to achieve this locally distributed protein production. Here we provide evidence for a microtubule-dependent localization of mRNA encoding the actin regulatory protein profilin to sites in mouse embryonic fibroblasts, which express enhanced actin polymerization.
Collapse
Affiliation(s)
- Anna-Karin Johnsson
- Department of Cell Biology, WGI, Stockholm University, SE-106 91 Stockholm, Sweden
| | | |
Collapse
|
31
|
Intermediate filaments take the heat as stress proteins. Trends Cell Biol 2010; 20:79-91. [PMID: 20045331 DOI: 10.1016/j.tcb.2009.11.004] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 11/13/2009] [Accepted: 11/17/2009] [Indexed: 11/18/2022]
Abstract
Intermediate filament (IF) proteins and heat shock proteins (HSPs) are large multimember families that share several features, including protein abundance, significant upregulation in response to a variety of stresses, cytoprotective functions, and the phenocopying of several human diseases after IF protein or HSP mutation. We are now coming to understand that these common elements point to IFs as important cellular stress proteins with some roles akin to those already well-characterized for HSPs. Unique functional roles for IFs include protection from mechanical stress, whereas HSPs are characteristically involved in protein folding and as chaperones. Shared IF and HSP cytoprotective roles include inhibition of apoptosis, organelle homeostasis, and scaffolding. In this report, we review data that corroborate the view that IFs function as highly specialized cytoskeletal stress proteins that promote cellular organization and homeostasis.
Collapse
|
32
|
Chou YH, Kuo WL, Rosner MR, Tang WJ, Goldman RD. Structural changes in intermediate filament networks alter the activity of insulin-degrading enzyme. FASEB J 2009; 23:3734-42. [PMID: 19584300 PMCID: PMC2775012 DOI: 10.1096/fj.09-137455] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 06/18/2009] [Indexed: 12/13/2022]
Abstract
The intermediate filament (IF) protein nestin coassembles with vimentin and promotes the disassembly of these copolymers when vimentin is hyperphosphorylated during mitosis. The aim of this study is to determine the function of these nonfilamentous particles by identifying their interacting partners. In this study, we report that these disassembled vimentin/nestin complexes interact with insulin degrading enzyme (IDE). Both vimentin and nestin interact with IDE in vitro, but vimentin binds IDE with a higher affinity than nestin. Although the interaction between vimentin and IDE is enhanced by vimentin phosphorylation at Ser-55, the interaction between nestin and IDE is phosphorylation independent. Further analyses show that phosphorylated vimentin plays the dominant role in targeting IDE to the vimentin/nestin particles in vivo, while the requirement for nestin is related to its ability to promote vimentin IF disassembly. The binding of IDE to either nestin or phosphorylated vimentin regulates IDE activity differently, depending on the substrate. The insulin degradation activity of IDE is suppressed approximately 50% by either nestin or phosphorylated vimentin, while the cleavage of bradykinin-mimetic peptide by IDE is increased 2- to 3-fold. Taken together, our data demonstrate that the nestin-mediated disassembly of vimentin IFs generates a structure capable of sequestering and modulating the activity of IDE.
Collapse
Affiliation(s)
- Ying-Hao Chou
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Ave., Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
33
|
Kim H, Nakamura F, Lee W, Shifrin Y, Arora P, McCulloch CA. Filamin A is required for vimentin-mediated cell adhesion and spreading. Am J Physiol Cell Physiol 2009; 298:C221-36. [PMID: 19776392 DOI: 10.1152/ajpcell.00323.2009] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cell adhesion and spreading are regulated by complex interactions involving the cytoskeleton and extracellular matrix proteins. We examined the interaction of the intermediate filament protein vimentin with the actin cross-linking protein filamin A in regulation of spreading in HEK-293 and 3T3 cells. Filamin A and vimentin-expressing cells were well spread on collagen and exhibited numerous cell extensions enriched with filamin A and vimentin. By contrast, cells treated with small interfering RNA (siRNA) to knock down filamin A or vimentin were poorly spread; both of these cell populations exhibited >50% reductions of cell adhesion, cell surface beta1 integrin expression, and beta1 integrin activation. Knockdown of filamin A reduced vimentin phosphorylation and blocked recruitment of vimentin to cell extensions, whereas knockdown of filamin and/or vimentin inhibited the formation of cell extensions. Reduced vimentin phosphorylation, cell spreading, and beta1 integrin surface expression, and activation were phenocopied in cells treated with the protein kinase C inhibitor bisindolylmaleimide; cell spreading was also reduced by siRNA knockdown of protein kinase C-epsilon. By immunoprecipitation of cell lysates and by pull-down assays using purified proteins, we found an association between filamin A and vimentin. Filamin A also associated with protein kinase C-epsilon, which was enriched in cell extensions. These data indicate that filamin A associates with vimentin and to protein kinase C-epsilon, thereby enabling vimentin phosphorylation, which is important for beta1 integrin activation and cell spreading on collagen.
Collapse
Affiliation(s)
- Hugh Kim
- Canadian Institutes of Health Group in Matrix Dynamics, University of Toronto, Toronto, Ontario, M5S 3E2, Canada.
| | | | | | | | | | | |
Collapse
|
34
|
Gabriele S, Benoliel AM, Bongrand P, Théodoly O. Microfluidic investigation reveals distinct roles for actin cytoskeleton and myosin II activity in capillary leukocyte trafficking. Biophys J 2009; 96:4308-18. [PMID: 19450501 DOI: 10.1016/j.bpj.2009.02.037] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 02/03/2009] [Accepted: 02/17/2009] [Indexed: 02/03/2023] Open
Abstract
Circulating leukocyte sequestration in pulmonary capillaries is arguably the initiating event of lung injury in acute respiratory distress syndrome. We present a microfluidic investigation of the roles of actin organization and myosin II activity during the different stages of leukocyte trafficking through narrow capillaries (entry, transit and shape relaxation) using specific drugs (latrunculin A, jasplakinolide, and blebbistatin). The deformation rate during entry reveals that cell stiffness depends strongly on F-actin organization and hardly on myosin II activity, supporting a microfilament role in leukocyte sequestration. In the transit stage, cell friction is influenced by stiffness, demonstrating that the actin network is not completely broken after a forced entry into a capillary. Conversely, membrane unfolding was independent of leukocyte stiffness. The surface area of sequestered leukocytes increased by up to 160% in the absence of myosin II activity, showing the major role of molecular motors in microvilli wrinkling and zipping. Finally, cell shape relaxation was largely independent of both actin organization and myosin II activity, whereas a deformed state was required for normal trafficking through capillary segments.
Collapse
Affiliation(s)
- Sylvain Gabriele
- Université de la Méditerranée, Institut National de la Santé et de la Recherche Médicale INSERM U600-Centre National de la Recherche Scientifique CNRS UMR6212, Marseille, France
| | | | | | | |
Collapse
|
35
|
Chang L, Barlan K, Chou YH, Grin B, Lakonishok M, Serpinskaya AS, Shumaker DK, Herrmann H, Gelfand VI, Goldman RD. The dynamic properties of intermediate filaments during organelle transport. J Cell Sci 2009; 122:2914-23. [PMID: 19638410 DOI: 10.1242/jcs.046789] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Intermediate filament (IF) dynamics during organelle transport and their role in organelle movement were studied using Xenopus laevis melanophores. In these cells, pigment granules (melanosomes) move along microtubules and microfilaments, toward and away from the cell periphery in response to alpha-melanocyte stimulating hormone (alpha-MSH) and melatonin, respectively. In this study we show that melanophores possess a complex network of vimentin IFs which interact with melanosomes. IFs form an intricate, honeycomb-like network that form cages surrounding individual and small clusters of melanosomes, both when they are aggregated and dispersed. Purified melanosome preparations contain a substantial amount of vimentin, suggesting that melanosomes bind to IFs. Analyses of individual melanosome movements in cells with disrupted IF networks show increased movement of granules in both anterograde and retrograde directions, further supporting the notion of a melanosome-IF interaction. Live imaging reveals that IFs, in turn, become highly flexible as melanosomes disperse in response to alpha-MSH. During the height of dispersion there is a marked increase in the rate of fluorescence recovery after photobleaching of GFP-vimentin IFs and an increase in vimentin solubility. These results reveal a dynamic interaction between membrane bound pigment granules and IFs and suggest a role for IFs as modulators of granule movement.
Collapse
Affiliation(s)
- Lynne Chang
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Harvard University, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Leterrier J, Janmey P, Eyer J. Microtubule-independent regulation of neurofilament interactions in vitro by neurofilament-bound ATPase activities. Biochem Biophys Res Commun 2009; 384:37-42. [PMID: 19379708 PMCID: PMC3118462 DOI: 10.1016/j.bbrc.2009.04.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Accepted: 04/11/2009] [Indexed: 12/21/2022]
Abstract
Neurofilaments (NFs), the major neuronal intermediate filaments, form networks in vitro that mimic the axonal NF bundles. This report presents evidence for previously unknown regulation of the interactions between NFs by NF-associated ATPases. Two opposite effects on NF gelation in vitro occur at low and high ATP concentration. These findings support the hypothesis that NF bundles in situ are dynamic structures, and raise the possibility that ATP-hydrolyzing mechanoenzymes regulate their organization.
Collapse
Affiliation(s)
- J.F. Leterrier
- UMR CNRS 6187, Dpt. Neurosciences, P.B.S., Universite de Poitiers, 40 av. Recteur Pineau, 86022 Poitiers Cedex, France
| | - P.A. Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, 1010 Vagelos Laboratories, 3340 Smith Walk, Philadelphia, PA 19104, USA
| | - J. Eyer
- Laboratoire de Neurobiologie et Transgénèse, UPRES-EA3143, INSERM IFR132, Equipe constituante de Génopole Ouest, Batiment Monteclair, CHU, 49033 Angers, France
| |
Collapse
|
37
|
Izmiryan A, Franco CA, Paulin D, Li Z, Xue Z. Synemin isoforms during mouse development: Multiplicity of partners in vascular and neuronal systems. Exp Cell Res 2009; 315:769-83. [DOI: 10.1016/j.yexcr.2008.12.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 11/21/2008] [Accepted: 12/07/2008] [Indexed: 11/25/2022]
|
38
|
Flitney EW, Kuczmarski ER, Adam SA, Goldman RD. Insights into the mechanical properties of epithelial cells: the effects of shear stress on the assembly and remodeling of keratin intermediate filaments. FASEB J 2009; 23:2110-9. [PMID: 19246484 DOI: 10.1096/fj.08-124453] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The effects of shear stress on the keratin intermediate filament (KIF) cytoskeleton of cultured human alveolar epithelial (A549) cells have been investigated. Under normal culture conditions, immunofluorescence revealed a delicate network of fine tonofibrils containing KIFs, together with many nonfilamentous, keratin-containing "particles," mostly containing either keratin 8 (K8) or 18 (K18), but not both. Triton X-100 extracted approximately 10% of the cellular keratin, and this was accompanied by a loss of the particles but not the KIFs. Shear stress dramatically reduced the soluble keratin component and transformed the fine bundles of KIFs into thicker, "wavy" tonofibrils. Both effects were accompanied by the disappearance of most keratin particles and by increased phosphorylation of K8 and K18 on serine residues 73 and 33, respectively. The particles that remained after shearing were phosphorylated and were closely associated with KIFs. We suggest that keratin particles constitute a reservoir of protein that can be recruited into KIFs under flow, creating a more robust cytoskeleton able to withstand shear forces more effectively.
Collapse
Affiliation(s)
- Eric W Flitney
- Department of Cell and Molecular Biology, Feinberg School of Medicine of Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
39
|
Spurny R, Gregor M, Castañón MJ, Wiche G. Plectin deficiency affects precursor formation and dynamics of vimentin networks. Exp Cell Res 2008; 314:3570-80. [PMID: 18848541 DOI: 10.1016/j.yexcr.2008.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 09/04/2008] [Accepted: 09/07/2008] [Indexed: 10/21/2022]
Abstract
Plectin is a typical cytolinker protein that connects intermediate filaments to the other cytoskeletal filament systems and anchors them at membrane-associated junctional sites. One of the most important binding partners of plectin in fibroblasts is the intermediate filament subunit protein vimentin. Previous studies have demonstrated that vimentin networks are highly dynamic structures whose assembly and disassembly is accomplished stepwise via several intermediates. The precursor forms as well as polymerized (filamentous) vimentin are found in the cells in a dynamic equilibrium characterized by the turnover of the subunits within the polymer and the movement of the smaller precursors. To examine whether plectin plays a role in intermediate filament dynamics, we studied vimentin filament formation in plectin-deficient compared to wild-type fibroblasts using GFP-tagged vimentin. Monitoring vimentin and plectin in spreading and dividing cells, we demonstrate that plectin is associated with vimentin from the early stages of assembly and is required for vimentin motility as well as for the stepwise formation of stable filaments. Furthermore, plectin prevents vimentin networks from complete disassembly during mitosis, facilitating the rebuilding of the intermediate filament network in daughter cells.
Collapse
Affiliation(s)
- Radovan Spurny
- Department of Molecular Cell Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter, A-1030 Vienna, Austria
| | | | | | | |
Collapse
|
40
|
Bagchi S, Tomenius H, Belova LM, Ausmees N. Intermediate filament-like proteins in bacteria and a cytoskeletal function in Streptomyces. Mol Microbiol 2008; 70:1037-50. [PMID: 18976278 PMCID: PMC2680258 DOI: 10.1111/j.1365-2958.2008.06473.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Actin and tubulin cytoskeletons are conserved and widespread in bacteria. A strikingly intermediate filament (IF)-like cytoskeleton, composed of crescentin, is also present in Caulobacter crescentus and determines its specific cell shape. However, the broader significance of this finding remained obscure, because crescentin appeared to be unique to Caulobacter. Here we demonstrate that IF-like function is probably a more widespread phenomenon in bacteria. First, we show that 21 genomes of 26 phylogenetically diverse species encoded uncharacterized proteins with a central segmented coiled coil rod domain, which we regarded as a key structural feature of IF proteins and crescentin. Experimental studies of three in silico predicted candidates from Mycobacterium and other actinomycetes revealed a common IF-like property to spontaneously assemble into filaments in vitro. Furthermore, the IF-like protein FilP formed cytoskeletal structures in the model actinomycete Streptomyces coelicolor and was needed for normal growth and morphogenesis. Atomic force microscopy of living cells revealed that the FilP cytoskeleton contributed to mechanical fitness of the hyphae, thus closely resembling the function of metazoan IF. Together, the bioinformatic and experimental data suggest that an IF-like protein architecture is a versatile design that is generally present in bacteria and utilized to perform diverse cytoskeletal tasks.
Collapse
Affiliation(s)
- Sonchita Bagchi
- Department of Cell and Molecular Biology, Uppsala University, Box 596, 75124 Uppsala, Sweden
| | | | | | | |
Collapse
|
41
|
Pan Y, Jing R, Pitre A, Williams BJ, Skalli O. Intermediate filament protein synemin contributes to the migratory properties of astrocytoma cells by influencing the dynamics of the actin cytoskeleton. FASEB J 2008; 22:3196-206. [PMID: 18509200 DOI: 10.1096/fj.08-106187] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have shown previously that, in astrocytoma cells, synemin is present at the leading edge, an unusual localization for an intermediate filament (IF) protein. Here, we report that synemin down-regulation with specific small hairpin RNAs (shRNAs) sharply decreased the migration of astrocytoma cells. The presence of synemin at the leading edge also correlated with a high migratory potential, as shown by comparing astrocytoma cells to carcinoma cells without synemin at the leading edge. Synemin-silenced astrocytoma cells were smaller and spread more slowly than controls. In addition, synemin silencing reduced proliferation without increasing apoptosis. The adhesion to substratum and distribution of vinculin in focal contacts of synemin-silenced astrocytoma cells were similar to those of controls. Synemin-silenced cells, however, exhibited a reduction in the amount of filamentous (F) -actin and of alpha-actinin, but not of vinculin, associated with F-actin. Altogether, these results demonstrate that synemin is important for the malignant behavior of astrocytoma cells and that it contributes to the high motility of these cells by modulating the dynamics of alpha-actinin and actin.
Collapse
Affiliation(s)
- Yihang Pan
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Hwy., Shreveport, LA 71130, USA
| | | | | | | | | |
Collapse
|
42
|
Intermediate filament assembly: dynamics to disease. Trends Cell Biol 2008; 18:28-37. [PMID: 18083519 DOI: 10.1016/j.tcb.2007.11.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 10/31/2007] [Accepted: 11/01/2007] [Indexed: 11/21/2022]
Abstract
Intermediate filament (IF) proteins belong to a large and diverse gene family with broad representation in vertebrate tissues. Although considered the 'toughest' cytoskeletal fibers, studies in cultured cells have revealed that IF can be surprisingly dynamic and highly regulated. This review examines the diversity of IF assembly behaviors, and considers the ideas that IF proteins are co- or post-translationally assembled into oligomeric precursors, which can be delivered to different subcellular compartments by microtubules or actomyosin and associated motor proteins. Their interaction with other cellular elements via IF associated proteins (IFAPs) affects IF dynamics and also results in cellular networks with properties that transcend those of individual components. We end by discussing how mutations leading to defects in IF assembly, network formation or IF-IFAP association compromise in vivo functions of IF as protectors against environmental stress.
Collapse
|