1
|
Seneff S, Kyriakopoulos AM, Nigh G. Is autism a PIN1 deficiency syndrome? A proposed etiological role for glyphosate. J Neurochem 2024; 168:2124-2146. [PMID: 38808598 DOI: 10.1111/jnc.16140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Autism is a neurodevelopmental disorder, the prevalence of which has increased dramatically in the United States over the past two decades. It is characterized by stereotyped behaviors and impairments in social interaction and communication. In this paper, we present evidence that autism can be viewed as a PIN1 deficiency syndrome. Peptidyl-prolyl cis/trans isomerase, NIMA-Interacting 1 (PIN1) is a peptidyl-prolyl cis/trans isomerase, and it has widespread influences in biological organisms. Broadly speaking, PIN1 deficiency is linked to many neurodegenerative diseases, whereas PIN1 over-expression is linked to cancer. Death-associated protein kinase 1 (DAPK1) strongly inhibits PIN1, and the hormone melatonin inhibits DAPK1. Melatonin deficiency is strongly linked to autism. It has recently been shown that glyphosate exposure to rats inhibits melatonin synthesis as a result of increased glutamate release from glial cells and increased expression of metabotropic glutamate receptors. Glyphosate's inhibition of melatonin leads to a reduction in PIN1 availability in neurons. In this paper, we show that PIN1 deficiency can explain many of the unique morphological features of autism, including increased dendritic spine density, missing or thin corpus callosum, and reduced bone density. We show how PIN1 deficiency disrupts the functioning of powerful high-level signaling molecules, such as nuclear factor erythroid 2-related factor 2 (NRF2) and p53. Dysregulation of both of these proteins has been linked to autism. Severe depletion of glutathione in the brain resulting from chronic exposure to oxidative stressors and extracellular glutamate leads to oxidation of the cysteine residue in PIN1, inactivating the protein and further contributing to PIN1 deficiency. Impaired autophagy leads to increased sensitivity of neurons to ferroptosis. It is imperative that further research be conducted to experimentally validate whether the mechanisms described here take place in response to chronic glyphosate exposure and whether this ultimately leads to autism.
Collapse
Affiliation(s)
- Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Greg Nigh
- Immersion Health, Portland, Oregon, USA
| |
Collapse
|
2
|
Rodríguez-Cruz D, Boquet-Pujadas A, López-Muñoz E, Rincón-Heredia R, Paredes-Díaz R, Flores-Fortis M, Olivo-Marin JC, Guillén N, Aguilar-Rojas A. Three-dimensional cell culture conditions promoted the Mesenchymal-Amoeboid Transition in the Triple-Negative Breast Cancer cell line MDA-MB-231. Front Cell Dev Biol 2024; 12:1435708. [PMID: 39156975 PMCID: PMC11327030 DOI: 10.3389/fcell.2024.1435708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Breast cancer (BC) is the leading cause of death among women, primarily due to its potential for metastasis. As BC progresses, the extracellular matrix (ECM) produces more type-I collagen, resulting in increased stiffness. This alteration influences cellular behaviors such as migration, invasion, and metastasis. Specifically, cancer cells undergo changes in gene expression that initially promote an epithelial-to-mesenchymal transition (EMT) and subsequently, a transition from a mesenchymal to an amoeboid (MAT) migration mode. In this way, cancer cells can migrate more easily through the stiffer microenvironment. Despite their importance, understanding MATs remains challenging due to the difficulty of replicating in vitro the conditions for cell migration that are observed in vivo. Methods To address this challenge, we developed a three-dimensional (3D) growth system that replicates the different matrix properties observed during the progression of a breast tumor. We used this model to study the migration and invasion of the Triple-Negative BC (TNBC) cell line MDA-MB-231, which is particularly subject to metastasis. Results Our results indicate that denser collagen matrices present a reduction in porosity, collagen fiber size, and collagen fiber orientation, which are associated with the transition of cells to a rounder morphology with bleb-like protrusions. We quantified how this transition is associated with a more persistent migration, an enhanced invasion capacity, and a reduced secretion of matrix metalloproteinases. Discussion Our findings suggest that the proposed 3D growth conditions (especially those with high collagen concentrations) mimic key features of MATs, providing a new platform to study the physiology of migratory transitions and their role in BC progression.
Collapse
Affiliation(s)
- Daniela Rodríguez-Cruz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Aleix Boquet-Pujadas
- École Polytechnique Fédérale de Lausanne, Biomedical Imaging Group, Lausanne, Switzerland
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Eunice López-Muñoz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Ruth Rincón-Heredia
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Rodolfo Paredes-Díaz
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Mauricio Flores-Fortis
- Cuajimalpa Unit, Engineering and Natural Science Doctoral Program, Metropolitan Autonomous University, Mexico City, Mexico
- Cuajimalpa Unit, Department of Natural Science, Metropolitan Autonomous University, Mexico City, Mexico
| | - Jean-Christophe Olivo-Marin
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Nancy Guillén
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS ERL9195, Paris, France
| | - Arturo Aguilar-Rojas
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
| |
Collapse
|
3
|
Thompson E, Prior S, Brüning-Richardson A. Traditional Plant-Derived Compounds Inhibit Cell Migration and Induce Novel Cytoskeletal Effects in Glioblastoma Cells. J Xenobiot 2024; 14:613-633. [PMID: 38804289 PMCID: PMC11130960 DOI: 10.3390/jox14020036] [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: 03/06/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024] Open
Abstract
Glioblastomas (GBMs) are aggressive and invasive cancers of the brain, associated with high rates of tumour recurrence and poor patient outcomes despite initial treatment. Targeting cell migration is therefore of interest in highly invasive cancers such as GBMs, to prevent tumour dissemination and regrowth. One current aim of GBM research focuses on assessing the anti-migratory properties of novel or repurposed inhibitors, including plant-based drugs which display anti-cancer properties. We investigated the potential anti-migratory activity of plant-based products with known cytotoxic effects in cancers, using a range of two-dimensional (2D) and three-dimensional (3D) migration and invasion assays as well as immunofluorescence microscopy to determine the specific anti-migratory and phenotypic effects of three plant-derived compounds, Turmeric, Indigo and Magnolia bark, on established glioma cell lines. Migrastatic activity was observed in all three drugs, with Turmeric exerting the most inhibitory effect on GBM cell migration into scratches and from the spheroid edge at all the timepoints investigated (p < 0.001). We also observed novel cytoskeletal phenotypes affecting actin and the focal adhesion dynamics. As our in vitro results determined that Turmeric, Indigo and Magnolia are promising migrastatic drugs, we suggest additional experimentation at the whole organism level to further validate these novel findings.
Collapse
Affiliation(s)
| | - Sally Prior
- Correspondence: (S.P.); (A.B.-R.); Tel.: +44-01484-472518 (A.B.-R.)
| | | |
Collapse
|
4
|
Aikemu B, Shao Y, Yang G, Ma J, Zhang S, Yang X, Hong H, Yesseyeva G, Huang L, Jia H, Wang C, Zang L, Sun J, Zheng M. NDRG1 regulates Filopodia-induced Colorectal Cancer invasiveness via modulating CDC42 activity. Int J Biol Sci 2021; 17:1716-1730. [PMID: 33994856 PMCID: PMC8120473 DOI: 10.7150/ijbs.56694] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
N-myc downstream regulated gene-1 (NDRG1) has been identified as a putative metastasis suppressor gene and proved to be a key player in cancer spreading and proliferation in our previous work. However, the effects of NDRG1 on tumor invasion and the mechanisms behind it are rarely understood. Here we provided in silico evidence that NDRG1 plays a crucial role in actin reorganization in colorectal cancer (CRC). Through in vitro experiments, we next observed filopodia formation was altered in NDRG1-modified cell lines, while cell division cycle-42 (CDC42) displayed excessive activation in NDRG1-silenced cells. Mechanistically, NDRG1 loss disrupts the binding between RhoGDIα and CDC42 and triggers the activation of CDC42 and the downstream cascades PAK1/Cofilin, thereby promotes the formation of filopodia and invasiveness of CRC. The knockdown of NDRG1 led to enhanced dissemination of CRC cells in vivo and correlates with active CDC42 expression. Using clinical sample analysis, we found an elevated level of active CDC42 in patients with advanced T stage, and it was negatively related to NDRG1 expression. In sum, these results uncover a mechanism utilized by NDRG1 to regulate CDC42 activity in coordinating cytoskeleton reorganization, which was crucial in cancer invasion.
Collapse
Affiliation(s)
- Batuer Aikemu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanfei Shao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjun Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hiju Hong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Galiya Yesseyeva
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongtao Jia
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Zang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Prieto EI, Mojares EBA, Cortez JJM, Vasquez MR. Electrospun nanofiber scaffolds for the propagation and analysis of breast cancer stem cells in vitro. Biomed Mater 2021; 16:035004. [PMID: 33634797 DOI: 10.1088/1748-605x/abc3dd] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite advances in cancer treatment, breast cancer remains the second foremost cause of cancer mortality among women, with a high rate of relapse after initial treatment success. A subpopulation of highly malignant cancer cells, known as cancer stem cells (CSCs), is suspected to be linked to metastasis and relapse. Targeting of CSCs may therefore provide a means of addressing cancer-related mortality. However, due to their low population in vivo and a lack of proper culture platform for their propagation, much of the CSC biology remains unknown. Since maintenance of CSCs is heavily influenced by the tumor microenvironment, this study developed a 3D culture platform that mimics the metastatic tumor extracellular matrix (ECM) to effectively increase CSC population in vitro and allow CSC analysis. Through electrospinning, nanofibers that were aligned, porous, and collagen-coated were fabricated from polycaprolactone to recreate the metastatic tumor ECM assemblage. Breast cancer cells seeded onto the nanofiber scaffolds exhibited gross morphology and cytoskeletal phenotype similar to invasive cancer cells. Moreover, the population of breast cancer stem cells increased in nanofiber scaffolds. Analysis of breast cancer cells grown on the nanofiber scaffolds demonstrated an upregulation of mesenchymal markers and an increase in cell invasiveness suggesting the cells have undergone epithelial-mesenchymal transition. These results indicate that the fabricated nanofiber scaffolds effectively mimicked the tumor microenvironment that maintains the cancer stem cell population, offering a platform to enrich and analyze CSCs in vitro.
Collapse
Affiliation(s)
- E I Prieto
- National Institute of Molecular Biology and Biotechnology, College of Science, National Science Complex, University of the Philippines, Diliman, Quezon City 1101, Philippines
| | - E B A Mojares
- National Institute of Molecular Biology and Biotechnology, College of Science, National Science Complex, University of the Philippines, Diliman, Quezon City 1101, Philippines
| | - J J M Cortez
- National Institute of Molecular Biology and Biotechnology, College of Science, National Science Complex, University of the Philippines, Diliman, Quezon City 1101, Philippines
| | - M R Vasquez
- Department of Mining, Metallurgical, and Materials Engineering, College of Engineering, University of the Philippines, Diliman, Quezon City 1101, Philippines
| |
Collapse
|
6
|
Shen D, Liu Y, Liu Y, Wang T, Yuan L, Huang X, Wang Y. Long non-coding RNA EWSAT1 promoted metastasis and actin cytoskeleton changes via miR-24-3p sponging in osteosarcoma. J Cell Mol Med 2020; 25:716-728. [PMID: 33225581 PMCID: PMC7812296 DOI: 10.1111/jcmm.16121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/10/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
Non‐coding RNAs are closely associated with tumorigenesis in multiple malignant tumours, including osteosarcoma (OS). Long non‐coding RNA Ewing sarcoma‐associated transcript 1 (EWSAT1) plays a role in metastasis, and actin cytoskeletal changes in OS remain unclear. In the current study, we showed that EWSAT1 expression was up‐regulated in OS and that an elevation in the EWSAT1 expression level was correlated with poor prognosis in patients with OS. Functionally, we showed that knockdown of EWSAT1 suppressed migration and induced actin stress fibre degradation in MNNG/HOS and 143B cells. Moreover, we found that ROCK1 was a key downstream effector in EWSAT1‐mediated cell migration and actin stress fibre changes. Furthermore, we demonstrated that ROCK1 and EWSAT1 shared a similar microRNA response element of microRNA‐24‐3p (miR‐24‐3p). Moreover, we verified that miR‐24‐3p suppressed ROCK1 and its mediated migration and actin stress fibres change by direct targeting. EWSAT1 promoted ROCK1‐mediated migration and actin stress fibre formation through miR‐24‐3p sponging. Lastly, through an in vivo study, we demonstrated that EWSAT1 promoted lung metastasis in OS. According to the above‐mentioned results, we suggest that EWSAT1 acts as an oncogene and that EWSAT1/miR‐24‐3p/ROCK1 axial could be a new target in the treatment of OS.
Collapse
Affiliation(s)
- Dewei Shen
- 4th Department of Orthopaedic Surgery, Central Hospital affiliated to Shenyang Medical College, Shenyang, China
| | - Yize Liu
- 4th Department of Orthopaedic Surgery, Central Hospital affiliated to Shenyang Medical College, Shenyang, China
| | - Yuexin Liu
- School of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
| | - Tao Wang
- 4th Department of Orthopaedic Surgery, Central Hospital affiliated to Shenyang Medical College, Shenyang, China
| | - Lin Yuan
- 2nd Department of Orthopaedic Surgery, Second Affiliated Hospital of Shenyang Medical College, Shenyang, China
| | - Xuyang Huang
- 2nd Department of Neurology, Central Hospital affiliated to Shenyang Medical College, Shenyang, China
| | - Yong Wang
- 4th Department of Orthopaedic Surgery, Central Hospital affiliated to Shenyang Medical College, Shenyang, China.,Central Laboratory, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| |
Collapse
|
7
|
Shatkin G, Yeoman B, Birmingham K, Katira P, Engler AJ. Computational models of migration modes improve our understanding of metastasis. APL Bioeng 2020; 4:041505. [PMID: 33195959 PMCID: PMC7647620 DOI: 10.1063/5.0023748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/23/2020] [Indexed: 01/07/2023] Open
Abstract
Tumor cells migrate through changing microenvironments of diseased and healthy tissue, making their migration particularly challenging to describe. To better understand this process, computational models have been developed for both the ameboid and mesenchymal modes of cell migration. Here, we review various approaches that have been used to account for the physical environment's effect on cell migration in computational models, with a focus on their application to understanding cancer metastasis and the related phenomenon of durotaxis. We then discuss how mesenchymal migration models typically simulate complex cell–extracellular matrix (ECM) interactions, while ameboid migration models use a cell-focused approach that largely ignores ECM when not acting as a physical barrier. This approach greatly simplifies or ignores the mechanosensing ability of ameboid migrating cells and should be reevaluated in future models. We conclude by describing future model elements that have not been included to date but would enhance model accuracy.
Collapse
Affiliation(s)
- Gabriel Shatkin
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| | | | - Katherine Birmingham
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
8
|
Estradiol Induces Epithelial to Mesenchymal Transition of Human Glioblastoma Cells. Cells 2020; 9:cells9091930. [PMID: 32825553 PMCID: PMC7564468 DOI: 10.3390/cells9091930] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/02/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
The mesenchymal phenotype of glioblastoma multiforme (GBM), the most frequent and malignant brain tumor, is associated with the worst prognosis. The epithelial–mesenchymal transition (EMT) is a cell plasticity mechanism involved in GBM malignancy. In this study, we determined 17β-estradiol (E2)-induced EMT by changes in cell morphology, expression of EMT markers, and cell migration and invasion assays in human GBM-derived cell lines. E2 (10 nM) modified the shape and size of GBM cells due to a reorganization of actin filaments. We evaluated EMT markers expression by RT-qPCR, Western blot, and immunofluorescence.We found that E2 upregulated the expression of the mesenchymal markers, vimentin, and N-cadherin. Scratch and transwell assays showed that E2 increased migration and invasion of GBM cells. The estrogen receptor-α (ER-α)-selective agonist 4,4’,4’’-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT, 10 nM) affected similarly to E2 in terms of the expression of EMT markers and cell migration, and the treatment with the ER-α antagonist methyl-piperidino-pyrazole (MPP, 1 μM) blocked E2 and PPT effects. ER-β-selective agonist diarylpropionitrile (DNP, 10 nM) and antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazole[1,5-a]pyrimidin-3-yl]phenol (PHTPP, 1 μM) showed no effects on EMT marker expression. These data suggest that E2 induces EMT activation through ER-α in human GBM-derived cells.
Collapse
|
9
|
Matyushenko AM, Levitsky DI. Molecular Mechanisms of Pathologies of Skeletal and Cardiac Muscles Caused by Point Mutations in the Tropomyosin Genes. BIOCHEMISTRY (MOSCOW) 2020; 85:S20-S33. [PMID: 32087052 DOI: 10.1134/s0006297920140023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review is devoted to tropomyosin (Tpm) - actin-binding protein, which plays a crucial role in the regulation of contraction of skeletal and cardiac muscles. Special attention is paid to myopathies and cardiomyopathies - severe hereditary diseases of skeletal and cardiac muscles associated with point mutations in Tpm genes. The current views on the molecular mechanisms of these diseases and the effects of such mutations on the Tpm structure and functions are considered in detail. Besides, some part of the review is devoted to analysis of the properties of Tpm homodimers and heterodimers with myopathic substitutions of amino acid residues in only one of the two chains of the Tpm dimeric molecule.
Collapse
Affiliation(s)
- A M Matyushenko
- Bach Institute of Biochemistry, Federal Research Center on Fundamentals of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia.
| | - D I Levitsky
- Bach Institute of Biochemistry, Federal Research Center on Fundamentals of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
| |
Collapse
|
10
|
Dorland YL, Cornelissen AS, Kuijk C, Tol S, Hoogenboezem M, van Buul JD, Nolte MA, Voermans C, Huveneers S. Nuclear shape, protrusive behaviour and in vivo retention of human bone marrow mesenchymal stromal cells is controlled by Lamin-A/C expression. Sci Rep 2019; 9:14401. [PMID: 31591420 PMCID: PMC6779744 DOI: 10.1038/s41598-019-50955-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Culture expanded mesenchymal stromal cells (MSCs) are being extensively studied for therapeutic applications, including treatment of graft-versus-host disease, osteogenesis imperfecta and for enhancing engraftment of hematopoietic stem cells after transplantation. Thus far, clinical trials have shown that the therapeutic efficiency of MSCs is variable, which may in part be due to inefficient cell migration. Here we demonstrate that human MSCs display remarkable low migratory behaviour compared to other mesodermal-derived primary human cell types. We reveal that specifically in MSCs the nucleus is irregularly shaped and nuclear lamina are prone to wrinkling. In addition, we show that expression of Lamin A/C is relatively high in MSCs. We further demonstrate that in vitro MSC migration through confined pores is limited by their nuclei, a property that might correlate to the therapeutic inefficiency of administered MSC in vivo. Silencing expression of Lamin A/C in MSCs improves nuclear envelope morphology, promotes the protrusive activity of MSCs through confined pores and enhances their retention in the lung after intravenous administration in vivo. Our findings suggest that the intrinsic nuclear lamina properties of MSCs underlie their limited capacity to migrate, and that strategies that target the nuclear lamina might alter MSC-based cellular therapies.
Collapse
Affiliation(s)
- Yvonne L Dorland
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne S Cornelissen
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlijn Kuijk
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Simon Tol
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Hoogenboezem
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martijn A Nolte
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlijn Voermans
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Dietz C, Infanger M, Romswinkel A, Strube F, Kraus A. Apoptosis Induction and Alteration of Cell Adherence in Human Lung Cancer Cells under Simulated Microgravity. Int J Mol Sci 2019; 20:E3601. [PMID: 31340547 PMCID: PMC6678991 DOI: 10.3390/ijms20143601] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Lung cancer cells are known to change proliferation and migration under simulated microgravity. In this study, we sought to evaluate cell adherence, apoptosis, cytoskeleton arrangement, and gene expression under simulated microgravity. METHODS Human lung cancer cells were exposed to simulated microgravity in a random-positioning machine (RPM). Cell morphology and adherence were observed under phase-contrast microscopy, cytoskeleton staining was performed, apoptosis rate was determined, and changes in gene and protein expression were detected by real-time PCR with western blot confirmation. RESULTS Three-dimensional (3D)-spheroid formation was observed under simulated microgravity. Cell viability was not impaired. Actin filaments showed a shift in alignment from longitudinal to spherical. Apoptosis rate was significantly increased in the spheroids compared to the control. TP53, CDKN2A, PTEN, and RB1 gene expression was significantly upregulated in the adherent cells under simulated microgravity with an increase in corresponding protein production for p14 and RB1. SOX2 expression was significantly upregulated in the adherent cells, but protein was not. Gene expressions of AKT3, PIK3CA, and NFE2L2 remained unaltered. CONCLUSION Simulated microgravity induces alteration in cell adherence, increases apoptosis rate, and leads to upregulation of tumor suppressor genes in human lung cancer cells.
Collapse
Affiliation(s)
- Carlo Dietz
- Department of Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University, Leipziger Strasse 44, D-39120 Magdeburg, Germany
| | - Manfred Infanger
- Department of Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University, Leipziger Strasse 44, D-39120 Magdeburg, Germany
| | - Alexander Romswinkel
- Department of Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University, Leipziger Strasse 44, D-39120 Magdeburg, Germany
| | - Florian Strube
- Department of Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University, Leipziger Strasse 44, D-39120 Magdeburg, Germany
| | - Armin Kraus
- Department of Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke-University, Leipziger Strasse 44, D-39120 Magdeburg, Germany.
| |
Collapse
|
12
|
Tropomyosin Tpm 2.1 loss induces glioblastoma spreading in soft brain-like environments. J Neurooncol 2018; 141:303-313. [PMID: 30535593 DOI: 10.1007/s11060-018-03049-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/12/2018] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The brain is a very soft tissue. Glioblastoma (GBM) brain tumours are highly infiltrative into the surrounding healthy brain tissue and invasion mechanisms that have been defined using rigid substrates therefore may not apply to GBM dissemination. GBMs characteristically lose expression of the high molecular weight tropomyosins, a class of actin-associating proteins and essential regulators of the actin stress fibres and focal adhesions that underpin cell migration on rigid substrates. METHODS Here, we investigated how loss of the high molecular weight tropomyosins affects GBM on soft matrices that recapitulate the biomechanical architecture of the brain. RESULTS We find that Tpm 2.1 is down-regulated in GBM grown on soft substrates. We demonstrate that Tpm 2.1 depletion by siRNA induces cell spreading and elongation in soft 3D hydrogels, irrespective of matrix composition. Tpm 1.7, a second high molecular weight tropomyosin is also down-regulated when cells are cultured on soft brain-like surfaces and we show that effects of this isoform are matrix dependent, with Tpm 1.7 inducing cell rounding in 3D collagen gels. Finally, we show that the absence of Tpm 2.1 from primary patient-derived GBMs correlates with elongated, mesenchymal invasion. CONCLUSIONS We propose that Tpm 2.1 down-regulation facilitates GBM colonisation of the soft brain environment. This specialisation of the GBM actin cytoskeleton organisation that is highly suited to the soft brain-like environment may provide novel therapeutic targets for arresting GBM invasion.
Collapse
|
13
|
RND1 regulates migration of human glioblastoma stem-like cells according to their anatomical localization and defines a prognostic signature in glioblastoma. Oncotarget 2018; 9:33788-33803. [PMID: 30333910 PMCID: PMC6173464 DOI: 10.18632/oncotarget.26082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/31/2018] [Indexed: 12/21/2022] Open
Abstract
Despite post-operative radio-chemotherapy, glioblastoma systematically locally recurs. Tumors contacting the periventricular zone (PVZ) show earlier and more distant relapses than tumors not contacting the PVZ. Since glioblastoma stem-like cells (GSCs) have been proposed to play a major role in glioblastoma recurrence, we decided to test whether GSC migration properties could be different according to their anatomical location (PVZ+/PVZ–). For that purpose, we established paired cultures of GSCs from the cortical area (CT) and the PVZ of glioblastoma patient tumors. We demonstrated that PVZ GSCs possess higher migration and invasion capacities than CT GSCs. We highlighted specific transcriptomic profiles in PVZ versus CT populations and identified a down-regulation of the RhoGTPase, RND1 in PVZ GSCs compared to CT GSCs. Overexpression of RND1, dramatically inhibited PVZ GSC migration and conversely, downregulation of RND1 increased CT GSC migration. Additionally, transcriptomic analyses also revealed a down-regulation of RND1 in glioblastoma compared to normal brain. Using the glioblastoma TCGA database, low levels of RND1 were also shown to correlate with a decreased overall survival of patients. Finally, based on signaling pathways activated in patients with low levels of RND1, we identified an RND1low signature of six genes (MET, LAMC1, ITGA5, COL5A1, COL3A1, COL1A2) that is an independent prognostic factor in glioblastoma. These findings contribute to explain the shorter time to progression of patients with PVZ involvement and, point out genes that establish the RND1low signature as key targets genes to impede tumor relapse after treatment.
Collapse
|
14
|
Mitchell CB, O'Neill GM. Rac GTPase regulation of 3D invasion in neuroblastomas lacking MYCN amplification. Cell Adh Migr 2016; 11:68-79. [PMID: 27224546 DOI: 10.1080/19336918.2016.1183868] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neuroblastomas are highly invasive tumors that occur in pediatric patients and treatment of invasive disease remains a challenge. The study of cells invading in 3-dimensional (3D) hydrogels has revealed morphologically distinct modes of invasion by which cancer cells adapt to the local tissue environment in order to invade local tissue. Specifically, the small G protein Rac GTPase has been implicated as regulating the elongated/mesenchymal mode of cell invasion. In the present study we demonstrate an inverse association between Rac expression and amplification of MYCN, a well-established prognostic indicator in neuroblastoma. Moreover, the association further tracks with previously described morphological variants of neuroblastoma. Importantly, while MYCN amplification is associated with universally poor prognosis, the clinical course of patients whose tumors lack MYCN amplification are more difficult to predict. Therefore, we analyzed the role that Rac plays in regulating the invasive behavior of neuroblastoma cells lacking MYCN amplification. Using siRNA targeting Rac in single cell suspensions in 3D collagen gels and Rac inhibition of multicellular spheroids (MCS) embedded in collagen gels, we find that the high Rac-expressing lines differ in their morphological response to Rac depletion and inhibition. Live cell imaging of embedded MCS reveals distinct individual and collective modes of invasion between the cell lines. Critically, Rac inhibition blocked both individual and collective invasion in 2 of the 3 high Rac expressing cell lines. Our study suggests that Rac activity may be an important determinant of metastatic capability in subsets of neuroblastoma cells lacking MYCN amplification.
Collapse
Affiliation(s)
- Camilla B Mitchell
- a Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead , Westmead , New South Wales , Australia
| | - Geraldine M O'Neill
- a Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead , Westmead , New South Wales , Australia.,b Discipline of Pediatrics and Child Health, The University of Sydney , Sydney , New South Wales , Australia
| |
Collapse
|
15
|
Wang P, Zhao J, Yang X, Guan S, Feng H, Han D, Lu J, Ou B, Jin R, Sun J, Zong Y, Feng B, Ma J, Lu A, Zheng M. PFDN1, an indicator for colorectal cancer prognosis, enhances tumor cell proliferation and motility through cytoskeletal reorganization. Med Oncol 2015; 32:264. [PMID: 26553318 DOI: 10.1007/s12032-015-0710-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 10/31/2015] [Indexed: 11/24/2022]
Abstract
Prefoldin (PFDN) subunits have been reported upregulated in various tumor types, while the expression and functions of PFDN1 (PFDN subunit 1) in colorectal cancer (CRC) are not well elucidated. The aim of this study was to investigate the use of PFDN1 as a poor prognosis indicator for CRC and explore the functions of PFDN1 in CRC. The relationship between PFDN1 expression and CRC clinical-pathological statistics was detected on the tissue microarray containing 145 cases of CRC. ShRNA was used to silence PFDN1 expression in SW480 and RKO CRC cells, and these transfected cells were analyzed for changes in proliferation, colony formation, cell cycle, migration, and invasion. Immunofluorescence and immunoblot were used to determine the remodeling of the F-actin and α-tubulin. Finally, tumor growth on nude mice was observed and measured. In this study, we found PFDN1 was upregulated in CRC tissues compared with adjacent normal tissues. Also, PFDN1 expression positively correlated with tumor size and tumor invasion. Moreover, after silencing PFDN1 in SW480 and RKO cells, the proliferation and motility of CRC cells were significantly suppressed. The inhibitory effect of PFDN1 on tumor cell growth and motility was partially due to G2/M cell cycle blockage and cytoskeletal deficiency. Finally, in vivo assay showed that downregulation of PFDN1 inhibited tumor growth on nude mice and PFDN1 expression correlated with higher levels of Ki-67 staining. These findings indicate that PFDN1 was involved in the progression of CRC, and provide new insights into PFDN1 as a potential therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Puxiongzhi Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China.
| | - Jingkun Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China
| | - Shaopei Guan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China
| | - Hao Feng
- School of Medicine, University of Munich, Munich, Germany
| | - Dingpei Han
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai, China
| | - Jun Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baochi Ou
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runsen Jin
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China
| | - Yaping Zong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China
| | - Bo Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjun Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Aiguo Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China.
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai, China
| |
Collapse
|
16
|
Schmeiser C, Winkler C. The flatness of Lamellipodia explained by the interaction between actin dynamics and membrane deformation. J Theor Biol 2015; 380:144-55. [PMID: 26002996 DOI: 10.1016/j.jtbi.2015.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 04/30/2015] [Accepted: 05/05/2015] [Indexed: 11/28/2022]
Abstract
The crawling motility of many cell types relies on lamellipodia, flat protrusions spreading on flat substrates but (on cells in suspension) also growing into three-dimensional space. Lamellipodia consist of a plasma membrane wrapped around an oriented actin filament meshwork. It is well known that the actin density is controlled by coordinated polymerization, branching, and capping processes, but the mechanisms producing the small aspect ratios of lamellipodia (hundreds of nm thickness vs. several μm lateral and inward extension) remain unclear. The main hypothesis of this work is a strong influence of the local geometry of the plasma membrane on the actin dynamics. This is motivated by observations of co-localization of proteins with I-BAR domains (like IRSp53) with polymerization and branching agents along the membrane. The I-BAR domains are known to bind to the membrane and to prefer and promote membrane curvature. This hypothesis is translated into a stochastic mathematical model where branching and capping rates, and polymerization speeds depend on the local membrane geometry and branching directions are influenced by the principal curvature directions. This requires the knowledge of the deformation of the membrane, being described in a quasi-stationary approximation by minimization of a modified Helfrich energy, subject to the actin filaments acting as obstacles. Simulations with this model predict pieces of flat lamellipodia without any prescribed geometric restrictions.
Collapse
Affiliation(s)
- Christian Schmeiser
- Faculty of Mathematics, University of Vienna, Oskar-Morgenstern-Platz 1, 1090 Vienna, Austria; Johann Radon Institute for Computational and Applied Mathematics, Austrian Academy of Sciences, Altenberger Straße 69, 4040 Linz, Austria
| | - Christoph Winkler
- Johann Radon Institute for Computational and Applied Mathematics, Austrian Academy of Sciences, Altenberger Straße 69, 4040 Linz, Austria.
| |
Collapse
|
17
|
O'Neill GM. Scared stiff: Stabilizing the actin cytoskeleton to stop invading cancer cells in their tracks. BIOARCHITECTURE 2014; 1:29-31. [PMID: 21866259 DOI: 10.4161/bioa.1.1.14665] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 12/28/2010] [Indexed: 01/29/2023]
Abstract
In recent years the concept of plasticity between invasion modes used by individual cancer cells has been gaining increasing interest in the field. Individually invading tumour cells can be divided into those that use a mesenchymal invasion mode, those that use "amoeboid" invasion and those that can switch between the two modes. The morphological distinctions between these different modes of invasion suggest that the actin cytoskeleton is likely to be a major contributor to the plasticity of cancer cell invasion mechanisms. We have recently investigated this idea by manipulating expression of Tm5NM1, one isoform of the tropomyosin family of actin-associating proteins. In a novel finding, we discovered that stabilizing the actin cytoskeleton via elevated expression of Tm5NM1 specifically inhibits mesenchymal-type cancer cell invasion, without causing transition to "amoeboid" motility-thus stopping the invading cancer cells in their tracks. The present perspective discusses our recent data in the context of current understanding of invasion plasticity and considers how stabilizing actin filaments may inhibit the mesenchymal invasion mode.
Collapse
Affiliation(s)
- Geraldine M O'Neill
- Children's Cancer Research Unit; Kids Research Institute; The Children's Hospital at Westmead; Discipline of Paediatrics and Child Health; University of Sydney; Sydney, Australia
| |
Collapse
|
18
|
Jones TD, Naimipour H, Sun S, Cho M, Alapati SB. Mechanical changes in human dental pulp stem cells during early odontogenic differentiation. J Endod 2014; 41:50-5. [PMID: 25241024 DOI: 10.1016/j.joen.2014.07.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/18/2014] [Accepted: 07/28/2014] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Cell adhesion and migration in bioactive scaffolds require actin cytoskeleton remodeling and focal adhesion formation. Additionally, human dental pulp stem cells (hDPSCs) undergo several changes in their mechanical properties during odontogenic differentiation. The effect of factors essential for odontogenesis on actin stress fiber elasticity and focal adhesion formation is not known. METHODS Live hDPSCs cultured in odontogenic media were imaged for cytoskeleton changes using an atomic force microscope. The Young's modulus (kPa) of the cytoskeleton was recorded as a function of culture medium for 10 days. Focal adhesion formation was assessed using immunofluorescence. Cultured hDPSCs were incubated with a monoclonal vinculin antibody, and filamentous actins were visualized using 0.5 μmol/L phalloidin. RESULTS Cytoskeletal elasticity significantly increased in response to odontogenic media. Both the number and physical size of focal adhesions in hDPSCs also increased. Up-regulation of vinculin expression was evident. The increase in the formation of focal adhesions was consistent with actin remodeling to stress fibers. CONCLUSIONS Our findings suggest that hDPSCs firmly attach to the glass substrate in response to odontogenic media. Successful regeneration of pulp-dentin tissue using biomimetic scaffolds will likely require cell-extracellular matrix interactions influenced by biochemical induction factors.
Collapse
Affiliation(s)
- Taneka D Jones
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Hamed Naimipour
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Shan Sun
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Michael Cho
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Satish B Alapati
- Department of Endodontics, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
19
|
N-myc downstream-regulated gene 1 downregulates cell proliferation, invasiveness, and tumorigenesis in human oral squamous cell carcinoma. Cancer Lett 2014; 355:242-52. [PMID: 25218595 DOI: 10.1016/j.canlet.2014.08.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 02/06/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common phenotype of oral cancer. N-myc downstream regulated gene 1 (NDRG1) is a modulator for cell proliferation, differentiation, and invasion. The role and function of NDRG1 in OSCC cells remain inconclusive. The (3)H-thymidine incorporation and in vitro matrigel invasion assays revealed NDRG1-knockdown significantly enhanced OSCC cell proliferation and invasion. Overexpressed NDRG1 arrested the cell cycle at the S-phase, thus attenuated cell proliferation in OECM-1 cells. The NDRG1-knockdown enhanced tumorigenesis of OECM-1 cells in the xenograft animal model. Western-blot and zymographic assays revealed that NDRG1 downregulated the gelatinase activities and protein levels of metalloproteinase-2 (MMP-2) and metalloproteinase-9 (MMP-9). NDRG1 modulated epithelial-mesenchymal transition (EMT) through upregulation of the E-cadherin expression, but downregulation of the N-cadherin, Vimentin, Snail-1, and Slug. Immunofluorescence staining indicated knockdown of NDRG1 enhanced F-actin expression and polymerization. Our results indicated NDRG1 attenuated OSCC cell growth in vitro and in vivo. The downregulation of EMT, MMP-2, and MMP-9 may explain the role of anti-invasion of NDRG1 in human OSCC cells. The experiments recognize that NDRG1 is an antitumor gene in OSCC cells.
Collapse
|
20
|
Zhu L, Yang J, Zhang J, Peng B. A Comparative Study of BioAggregate and ProRoot MTA on Adhesion, Migration, and Attachment of Human Dental Pulp Cells. J Endod 2014; 40:1118-23. [DOI: 10.1016/j.joen.2013.12.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/24/2013] [Accepted: 12/27/2013] [Indexed: 01/08/2023]
|
21
|
Exendin-4 induces cell adhesion and differentiation and counteracts the invasive potential of human neuroblastoma cells. PLoS One 2013; 8:e71716. [PMID: 23990978 PMCID: PMC3750033 DOI: 10.1371/journal.pone.0071716] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 07/02/2013] [Indexed: 12/25/2022] Open
Abstract
Exendin-4 is a molecule currently used, in its synthetic form exenatide, for the treatment of type 2 diabetes mellitus. Exendin-4 binds and activates the Glucagon-Like Peptide-1 Receptor (GLP-1R), thus inducing insulin release. More recently, additional biological properties have been associated to molecules that belong to the GLP-1 family. For instance, Peptide YY and Vasoactive Intestinal Peptide have been found to affect cell adhesion and migration and our previous data have shown a considerable actin cytoskeleton rearrangement after exendin-4 treatment. However, no data are currently available on the effects of exendin-4 on tumor cell motility. The aim of this study was to investigate the effects of this molecule on cell adhesion, differentiation and migration in two neuroblastoma cell lines, SH-SY5Y and SK-N-AS. We first demonstrated, by Extra Cellular Matrix cell adhesion arrays, that exendin-4 increased cell adhesion, in particular on a vitronectin substrate. Subsequently, we found that this molecule induced a more differentiated phenotype, as assessed by i) the evaluation of neurite-like protrusions in 3D cell cultures, ii) the analysis of the expression of neuronal markers and iii) electrophysiological studies. Furthermore, we demonstrated that exendin-4 reduced cell migration and counteracted anchorage-independent growth in neuroblastoma cells. Overall, these data indicate for the first time that exendin-4 may have anti-tumoral properties.
Collapse
|
22
|
Avellino R, Carrella S, Pirozzi M, Risolino M, Salierno FG, Franco P, Stoppelli P, Verde P, Banfi S, Conte I. miR-204 targeting of Ankrd13A controls both mesenchymal neural crest and lens cell migration. PLoS One 2013; 8:e61099. [PMID: 23620728 PMCID: PMC3631221 DOI: 10.1371/journal.pone.0061099] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 03/05/2013] [Indexed: 12/20/2022] Open
Abstract
Loss of cell adhesion and enhancement of cell motility contribute to epithelial-to-mesenchymal transition during development. These processes are related to a) rearrangement of cell-cell and cell-substrate adhesion molecules; b) cross talk between extra-cellular matrix and internal cytoskeleton through focal adhesion molecules. Focal adhesions are stringently regulated transient structures implicated in cell adhesion, spreading and motility during tissue development. Importantly, despite the extensive elucidation of the molecular composition of focal adhesions, the complex regulation of their dynamics is largely unclear. Here, we demonstrate, using live-imaging in medaka, that the microRNA miR-204 promotes both mesenchymal neural crest and lens cell migration and elongation. Overexpression of miR-204 results in upregulated cell motility, while morpholino-mediated ablation of miR-204 activity causes abnormal lens morphogenesis and neural crest cell mislocalization. Using a variety of in vivo and in vitro approaches, we demonstrate that these actions are mediated by the direct targeting of the Ankrd13A gene, which in turn controls focal cell adhesion formation and distribution. Significantly, in vivo restoration of abnormally elevated levels of Ankrd13A resulting from miR-204 inactivation rescued the aberrant lens phenotype in medaka fish. These data uncover, for the first time in vivo, the role of a microRNA in developmental control of mesenchymal cell migration and highlight miR-204 as a "master regulator" of the molecular networks that regulate lens morphogenesis in vertebrates.
Collapse
Affiliation(s)
| | | | | | | | | | - Paola Franco
- Institute of Genetics and Biophysics, Naples, Italy
| | | | | | - Sandro Banfi
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Medical Genetics, Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Naples, Italy
| |
Collapse
|
23
|
Sun J, Zhang D, Zheng Y, Zhao Q, Zheng M, Kovacevic Z, Richardson DR. Targeting the metastasis suppressor, NDRG1, using novel iron chelators: regulation of stress fiber-mediated tumor cell migration via modulation of the ROCK1/pMLC2 signaling pathway. Mol Pharmacol 2013; 83:454-69. [PMID: 23188716 DOI: 10.1124/mol.112.083097] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The iron-regulated metastasis suppressor, N-myc downstream-regulated gene 1 (NDRG1), is up-regulated by cellular iron depletion mediated by iron chelators and can inhibit cancer cell migration. However, the mechanism of how NDRG1 achieves this effect remains unclear. In this study, we implemented established and newly constructed NDRG1 overexpression and knockdown models using the DU145, HT29, and HCT116 cancer cell lines to investigate the molecular basis by which NDRG1 exerts its inhibitory effect on cell migration. Using these models, we demonstrated that NDRG1 overexpression inhibits cell migration by preventing actin-filament polymerization, stress fiber assembly and formation. In contrast, NDRG1 knockdown had the opposite effect. Moreover, we identified that NDRG1 inhibited an important regulatory pathway mediated by the Rho-associated, coiled-coil containing protein kinase 1 (ROCK1)/phosphorylated myosin light chain 2 (pMLC2) pathway that modulates stress fiber assembly. The phosphorylation of MLC2 is a key process in inducing stress fiber contraction, and this was shown to be markedly decreased or increased by NDRG1 overexpression or knockdown, respectively. The mechanism involved in the inhibition of MLC2 phosphorylation by NDRG1 was mediated by a significant (P < 0.001) decrease in ROCK1 expression that is a key kinase involved in MLC2 phosphorylation. Considering that NDRG1 is up-regulated after cellular iron depletion, novel thiosemicarbazone iron chelators (e.g., di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone) were demonstrated to inhibit ROCK1/pMLC2-modulated actin-filament polymerization, stress fiber assembly, and formation via a mechanism involving NDRG1. These results highlight the role of the ROCK1/pMLC2 pathway in the NDRG1-mediated antimetastatic signaling network and the therapeutic potential of iron chelators at inhibiting metastasis.
Collapse
Affiliation(s)
- Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|
24
|
Chapin LM, Blankman E, Smith MA, Shiu YT, Beckerle MC. Lateral communication between stress fiber sarcomeres facilitates a local remodeling response. Biophys J 2012. [PMID: 23200042 DOI: 10.1016/j.bpj.2012.09.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Actin stress fibers (SFs) are load-bearing and mechanosensitive structures. To our knowledge, the mechanisms that enable SFs to sense and respond to strain have not been fully defined. Acute local strain events can involve a twofold extension of a single SF sarcomere, but how these dramatic local events affect the overall SF architecture is not believed to be understood. Here we have investigated how SF architecture adjusts to episodes of local strain that occur in the cell center. Using fluorescently tagged zyxin to track the borders of sarcomeres, we characterize the dynamics of resting sarcomeres and strain-site sarcomeres. We find that sarcomeres flanking a strain site undergo rapid shortening that directly compensates for the strain-site extension, illustrating lateral communication of mechanical information along the length of a stress fiber. When a strain-site sarcomere extends asymmetrically, its adjacent sarcomeres exhibit a parallel asymmetric shortening response, illustrating that flanking sarcomeres respond to strain magnitude. After extension, strain-site sarcomeres become locations of new sarcomere addition, highlighting mechanical strain as a trigger of sarcomere addition and revealing a, to our knowledge, novel type of SF remodeling. Our findings provide evidence to suggest SF sarcomeres act as strain sensors and are interconnected to support communication of mechanical information.
Collapse
Affiliation(s)
- Laura M Chapin
- Huntsman Cancer Institute, Departments of Biology and Oncological Sciences, Salt Lake City, UT, USA
| | | | | | | | | |
Collapse
|
25
|
Eitaki M, Yamamori T, Meike S, Yasui H, Inanami O. Vincristine enhances amoeboid-like motility via GEF-H1/RhoA/ROCK/Myosin light chain signaling in MKN45 cells. BMC Cancer 2012; 12:469. [PMID: 23057787 PMCID: PMC3522013 DOI: 10.1186/1471-2407-12-469] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 10/09/2012] [Indexed: 12/22/2022] Open
Abstract
Background Anti-cancer drugs are widely used in cancer treatment frequently combined with surgical therapy and/or radiation therapy. Although surgery and radiation have been suggested to facilitate invasion and metastasis of tumor cells in some cases, there is so far little information about the effect of anti-cancer drugs on cellular invasive ability and metastasis. In this study, using four different anti-cancer drugs (vincristine, paclitaxel, cisplatin and etoposide), we examined whether these drugs influence the invasive ability of tumor cells. Methods Human gastric adenocarcinoma MKN45 cells were used to evaluate the effect of anti-cancer drugs. After drug treatment, cellular invasive ability was assessed using the Matrigel invasion chamber. Cytoskeletal changes after treatment were examined microscopically with F-actin staining. In addition, we monitored cellular motility in 3D matrigel environment by time-lapse microscopic analysis. The drug-induced activation of RhoA and ROCK was evaluated by pull-down assay and Western blotting using an antibody against phosphorylated myosin light chain (MLC), respectively. Where necessary, a ROCK inhibitor Y27632 and siRNA for guanine nucleotide exchange factor-H1 (GEF-H1) were applied. Results Among all drugs tested, only vincristine stimulated the invasive ability of MKN45 cells. Microscopic analysis revealed that vincristine induced the formation of non-apoptotic membrane blebs and amoeboid-like motility. Vincristine significantly enhanced RhoA activity and MLC phosphorylation, suggesting the involvement of RhoA/ROCK pathway in the vincristine-induced cytoskeletal reorganization and cellular invasion. Furthermore, we found that Y27632 as well as the siRNA for GEF-H1, a RhoA-specific activator, attenuated MLC phosphorylation, the formation of membrane blebs and the invasive ability after vincristine treatment. Conclusions These results indicate that vincristine activates GEF-H1/RhoA/ROCK/MLC signaling, thereby promoting amoeboid-like motility and the invasive ability of MKN45 cells.
Collapse
Affiliation(s)
- Masato Eitaki
- Department of Environmental Veterinary Sciences, Laboratory of Radiation Biology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | |
Collapse
|
26
|
Mironava T, Hadjiargyrou M, Simon M, Rafailovich MH. The effects of UV emission from compact fluorescent light exposure on human dermal fibroblasts and keratinocytes in vitro. Photochem Photobiol 2012; 88:1497-506. [PMID: 22724459 DOI: 10.1111/j.1751-1097.2012.01192.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Compact fluorescent light (CFL) bulbs can provide the same amount of lumens as incandescent light bulbs, using one quarter of the energy. Recently, CFL exposure was found to exacerbate existing skin conditions; however, the effects of CFL exposure on healthy skin tissue have not been thoroughly investigated. In this study, we studied the effects of exposure to CFL illumination on healthy human skin tissue cells (fibroblasts and keratinocytes). Cells exposed to CFLs exhibited a decrease in the proliferation rate, a significant increase in the production of reactive oxygen species, and a decrease in their ability to contract collagen. Measurements of UV emissions from these bulbs found significant levels of UVC and UVA (mercury [Hg] emission lines), which appeared to originate from cracks in the phosphor coatings, present in all bulbs studied. The response of the cells to the CFLs was consistent with damage from UV radiation, which was further enhanced when low dosages of TiO(2) nanoparticles (NPs), normally used for UV absorption, were added prior to exposure. No effect on cells, with or without TiO(2) NPs, was observed when they were exposed to incandescent light of the same intensity.
Collapse
Affiliation(s)
- Tatsiana Mironava
- Department of Materials Science and Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA.
| | | | | | | |
Collapse
|
27
|
Zhong J, Baquiran JB, Bonakdar N, Lees J, Ching YW, Pugacheva E, Fabry B, O'Neill GM. NEDD9 stabilizes focal adhesions, increases binding to the extra-cellular matrix and differentially effects 2D versus 3D cell migration. PLoS One 2012; 7:e35058. [PMID: 22509381 PMCID: PMC3324407 DOI: 10.1371/journal.pone.0035058] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 03/08/2012] [Indexed: 12/30/2022] Open
Abstract
The speed of cell migration on 2-dimensional (2D) surfaces is determined by the rate of assembly and disassembly of clustered integrin receptors known as focal adhesions. Different modes of cell migration that have been described in 3D environments are distinguished by their dependence on integrin-mediated interactions with the extra-cellular matrix. In particular, the mesenchymal invasion mode is the most dependent on focal adhesion dynamics. The focal adhesion protein NEDD9 is a key signalling intermediary in mesenchymal cell migration, however whether NEDD9 plays a role in regulating focal adhesion dynamics has not previously been reported. As NEDD9 effects on 2D migration speed appear to depend on the cell type examined, in the present study we have used mouse embryo fibroblasts (MEFs) from mice in which the NEDD9 gene has been depleted (NEDD9 -/- MEFs). This allows comparison with effects of other focal adhesion proteins that have previously been demonstrated using MEFs. We show that focal adhesion disassembly rates are increased in the absence of NEDD9 expression and this is correlated with increased paxillin phosphorylation at focal adhesions. NEDD9-/- MEFs have increased rates of migration on 2D surfaces, but conversely, migration of these cells is significantly reduced in 3D collagen gels. Importantly we show that myosin light chain kinase is activated in 3D in the absence of NEDD9 and is conversely inhibited in 2D cultures. Measurement of adhesion strength reveals that NEDD9-/- MEFs have decreased adhesion to fibronectin, despite upregulated α5β1 fibronectin receptor expression. We find that β1 integrin activation is significantly suppressed in the NEDD9-/-, suggesting that in the absence of NEDD9 there is decreased integrin receptor activation. Collectively our data suggest that NEDD9 may promote 3D cell migration by slowing focal adhesion disassembly, promoting integrin receptor activation and increasing adhesion force to the ECM.
Collapse
Affiliation(s)
- Jessie Zhong
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jaime B. Baquiran
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Navid Bonakdar
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Justin Lees
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Yu Wooi Ching
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Elena Pugacheva
- Mary Babb Randolph Cancer Center (MBRCC), West Virginia University, Morgantown, West Virginia, United States of America
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Geraldine M. O'Neill
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
28
|
Bradbury P, Mahmassani M, Zhong J, Turner K, Paul A, Verrills NM, O'Neill GM. PP2A phosphatase suppresses function of the mesenchymal invasion regulator NEDD9. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1823:290-7. [PMID: 22061964 DOI: 10.1016/j.bbamcr.2011.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 10/18/2011] [Accepted: 10/18/2011] [Indexed: 12/26/2022]
Abstract
The mesenchymal mode of cancer cell invasion characterized by active adhesion turnover and a polarized actin cytoskeleton, is critically regulated by the adaptor protein NEDD9/HEF1/Cas-L. While it is known that NEDD9 is subject to extensive phosphorylation modification, the molecules that determine NEDD9 phosphorylation to stimulate adhesion turnover and mesenchymal cell morphologies are currently unknown. Earlier studies have suggested that the serine/threonine phosphatase PP2A regulates interconversion between a low molecular mass NEDD9 phosphoform and higher molecular mass phosphoforms. However, previous studies have used chemical inhibitors to block PP2A activity. In the present study we therefore aimed to specifically inhibit PP2A activity via siRNA and dominant negative approaches to investigate the effect of PP2A on interconversion between 115 kDa and 105 kDa NEDD9 and determine the functional consequence of PP2A activity for NEDD9 function. Strikingly, we find that while the phosphatase inhibitor Calyculin A indeed abrogates detachment-induced dephosphorylation of the 115 kDa NEDD9 phosphoform, PP2A depletion does not inhibit 115 kDa to 105 kDa interconversion. Our data suggest instead that PP2A targets discrete NEDD9 phosphorylation modifications separate to the events that mediate interconversion between the two forms. Functionally, PP2A depletion increases NEDD9 mediated cell spreading and mutation of S369 in the serine-rich region of NEDD9 to aspartate mimics this effect. Importantly, mutation of S369 to alanine abrogates the ability of dominant negative PP2A to increase NEDD9-mediated cell spreading. Collectively, our data reveal that the tumour suppressor PP2A may act via S369 to regulated NEDD9-mediated cell spreading.
Collapse
Affiliation(s)
- Peta Bradbury
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, NSW, 2145 Australia
| | | | | | | | | | | | | |
Collapse
|
29
|
Lück S, Fichtl A, Sailer M, Joos H, Brenner RE, Walther P, Schmidt V. Statistical analysis of the intermediate filament network in cells of mesenchymal lineage by greyvalue-oriented image segmentation. Comput Stat 2011. [DOI: 10.1007/s00180-011-0265-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
30
|
Lee S, Terry D, Hurst DR, Welch DR, Sang QXA. Protein Signatures in Human MDA-MB-231 Breast Cancer Cells Indicating a More Invasive Phenotype Following Knockdown of Human Endometase/Matrilysin-2 by siRNA. J Cancer 2011; 2:165-76. [PMID: 21475635 PMCID: PMC3069352 DOI: 10.7150/jca.2.165] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 03/14/2011] [Indexed: 12/16/2022] Open
Abstract
Human matrix metalloproteinase-26 (MMP-26/endometase/matrilysin-2) is a putative biomarker for carcinomas of breast, prostate, and other cancers of epithelial origin. MMP-26 expression was silenced using small interfering RNA (siRNA) in the human breast cancer cell line MDA-MB-231. Immunological and proteomics approaches, including two-dimensional gel electrophoresis and matrix assisted laser desorption/ionization time-of-flight mass spectrometry, were employed to identify differential protein expression in MMP-26 knockdown cells. A comparison of the protein expression profiles of control and MMP-26 knockdown cells revealed nine differentially regulated proteins. Five of the proteins (heat shock protein 90, glucose-regulated protein 78 (GRP78), annexin V, tropomyosin, and peroxiredoxin II) were up-regulated, while alpha-tubulin, cystatin SA-III, breast cancer metastasis suppressor 1 (BRMS1) and beta-actin were down-regulated. This decrease of BRMS1 expression is concomitant with an increase of invasion through matrix-coated membranes. These results suggest an important role for MMP-26 in the regulation of proteins involved in invasive and metastatic breast cancers.
Collapse
Affiliation(s)
- Seakwoo Lee
- 1. Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4390, USA
| | | | | | | | | |
Collapse
|
31
|
Lees JG, Bach CTT, O'Neill GM. Interior decoration: tropomyosin in actin dynamics and cell migration. Cell Adh Migr 2011; 5:181-6. [PMID: 21173575 DOI: 10.4161/cam.5.2.14438] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cell migration and invasion requires the precise temporal and spatial orchestration of a variety of biological processes. Filaments of polymerized actin are critical players in these diverse processes, including the regulation of cell anchorage points (both cell-cell and cell-extracellular matrix), the uptake and delivery of molecules via endocytic pathways and the generation of force for both membrane protrusion and retraction. How the actin filaments are specialized for each of these discrete functions is yet to be comprehensively elucidated. The cytoskeletal tropomyosins are a family of actin associating proteins that form head-to-tail polymers which lay in the major groove of polymerized actin filaments. In the present review we summarize the emerging isoform-specific functions of tropomyosins in cell migration and invasion and discuss their potential roles in the specialization of actin filaments for the diverse cellular processes that together regulate cell migration and invasion.
Collapse
Affiliation(s)
- Justin G Lees
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | | | | |
Collapse
|
32
|
The actin-associating protein Tm5NM1 blocks mesenchymal motility without transition to amoeboid motility. Oncogene 2010; 30:1241-51. [PMID: 21076470 DOI: 10.1038/onc.2010.516] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cell migration is an integral component of metastatic disease. The ability of cells to transit between mesenchymal and amoeboid modes of migration has complicated the development of successful therapies designed to target cell migration as a means of inhibiting metastasis. Therefore, investigations of the mechanisms that regulate cell migration and render cells stationary are necessary. Tropomyosins are actin-associating proteins that regulate the activity of several effectors of actin filament dynamics. Previously, we have shown that the tropomyosin isoform Tm5NM1 stabilizes actin filaments and inhibits cell migration in a two-dimensional culture system. Here, we show that Tm5NM1 inhibits the mesenchymal migration of multiple cell lines in an isoform-specific manner. Tm5NM1 stimulates the downregulation of Src kinase activity and a rounded or elliptical morphology in three-dimensional collagen gels, and cells have dramatically reduced capacity to form pseudopodia. Importantly, we find that Tm5NM1 inhibits both the mesenchymal to amoeboid and amoeboid to mesenchymal transitions. Collectively, our data suggest that mimicking the action of Tm5NM1 overexpression represents an approach for effectively inhibiting the mesenchymal mode of migration.
Collapse
|
33
|
Mesenchymal migration as a therapeutic target in glioblastoma. JOURNAL OF ONCOLOGY 2010; 2010:430142. [PMID: 20652056 PMCID: PMC2905941 DOI: 10.1155/2010/430142] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 04/28/2010] [Indexed: 12/29/2022]
Abstract
Extensive infiltration of the surrounding healthy brain tissue is a cardinal feature of glioblastomas, highly lethal brain tumors. Deep infiltration by the glioblastoma cells renders complete surgical excision difficult and contemporary adjuvant therapies have had little impact on long-term survival. Thus, deep infiltration and resistance to irradiation and chemotherapy remain a major cause of patient mortality. Modern therapies specifically targeted to this unique aspect of glioblastoma cell biology hold significant promise to substantially improve survival rates for glioblastoma patients. In the present paper, we focus on the role of adhesion signaling molecules and the actin cytoskeleton in the mesenchymal mode of motility that characterizes invading glioblastoma cells. We then review current approaches to targeting these elements of the glioblastoma cell migration machinery and discuss other aspects of cell migration that may improve the treatment of infiltrating glioblastoma.
Collapse
|
34
|
Bach CTT, Schevzov G, Bryce NS, Gunning PW, O'Neill GM. Tropomyosin isoform modulation of focal adhesion structure and cell migration. Cell Adh Migr 2010; 4:226-34. [PMID: 20305380 DOI: 10.4161/cam.4.2.10888] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Orderly cell migration is essential for embryonic development, efficient wound healing and a functioning immune system and the dysregulation of this process leads to a number of pathologies. The speed and direction of cell migration is critically dependent on the structural organization of focal adhesions in the cell. While it is well established that contractile forces derived from the acto-myosin filaments control the structure and growth of focal adhesions, how this may be modulated to give different outcomes for speed and persistence is not well understood. The tropomyosin family of actin-associating proteins are emerging as important modulators of the contractile nature of associated actin filaments. The multiple non-muscle tropomyosin isoforms are differentially expressed between tissues and across development and are thought to be major regulators of actin filament functional specialization. In the present study we have investigated the effects of two splice variant isoforms from the same alpha-tropomyosin gene, TmBr1 and TmBr3, on focal adhesion structure and parameters of cell migration. These isoforms are normally switched on in neuronal cells during differentiation and we find that exogenous expression of the two isoforms in undifferentiated neuronal cells has discrete effects on cell migration parameters. While both isoforms cause reduced focal adhesion size and cell migration speed, they differentially effect actin filament phenotypes and migration persistence. Our data suggests that differential expression of tropomyosin isoforms may coordinate acto-myosin contractility and focal adhesion structure to modulate cell speed and persistence.
Collapse
Affiliation(s)
- Cuc T T Bach
- Focal Adhesion Biology, Kids Research Institute, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | | | | | | | | |
Collapse
|