101
|
Park KC, Paluncic J, Kovacevic Z, Richardson DR. Pharmacological targeting and the diverse functions of the metastasis suppressor, NDRG1, in cancer. Free Radic Biol Med 2020; 157:154-175. [PMID: 31132412 DOI: 10.1016/j.freeradbiomed.2019.05.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/24/2019] [Accepted: 05/16/2019] [Indexed: 12/18/2022]
Abstract
N-myc downstream regulated gene-1 (NDRG1) is a potent metastasis suppressor that is regulated by hypoxia, metal ions including iron, the free radical nitric oxide (NO.), and various stress stimuli. This intriguing molecule exhibits diverse functions in cancer, inhibiting epithelial-mesenchymal transition (EMT), cell migration and angiogenesis by modulation of a plethora of oncogenes via cellular signaling. Thus, pharmacological targeting of NDRG1 signaling in cancer is a promising therapeutic strategy. Of note, novel anti-tumor agents of the di-2-pyridylketone thiosemicarbazone series, which exert the "double punch" mechanism by binding metal ions to form redox-active complexes, have been demonstrated to markedly up-regulate NDRG1 expression in cancer cells. This review describes the mechanisms underlying NDRG1 modulation by the thiosemicarbazones and the diverse effects NDRG1 exerts in cancer. As a major induction mechanism, iron depletion appears critical, with NO. also inducing NDRG1 through its ability to bind iron and generate dinitrosyl-dithiol iron complexes, which are then effluxed from cells. Apart from its potent anti-metastatic role, several studies have reported a pro-oncogenic role of NDRG1 in a number of cancer-types. Hence, it has been suggested that NDRG1 plays pleiotropic roles depending on the cancer-type. The molecular mechanism(s) underlying NDRG1 pleiotropy remain elusive, but are linked to differential regulation of WNT signaling and potentially differential interaction with the tumor suppressor, PTEN. This review discusses NDRG1 induction mechanisms by metal ions and NO. and both the anti- and possible pro-oncogenic functions of NDRG1 in multiple cancer-types and compares the opposite effects this protein exerts on cancer progression.
Collapse
Affiliation(s)
- Kyung Chan Park
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Jasmina Paluncic
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia.
| |
Collapse
|
102
|
Tang VW. Collagen, stiffness, and adhesion: the evolutionary basis of vertebrate mechanobiology. Mol Biol Cell 2020; 31:1823-1834. [PMID: 32730166 PMCID: PMC7525820 DOI: 10.1091/mbc.e19-12-0709] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023] Open
Abstract
The emergence of collagen I in vertebrates resulted in a dramatic increase in the stiffness of the extracellular environment, supporting long-range force propagation and the development of low-compliant tissues necessary for the development of vertebrate traits including pressurized circulation and renal filtration. Vertebrates have also evolved integrins that can bind to collagens, resulting in the generation of higher tension and more efficient force transmission in the extracellular matrix. The stiffer environment provides an opportunity for the vertebrates to create new structures such as the stress fibers, new cell types such as endothelial cells, new developmental processes such as neural crest delamination, and new tissue organizations such as the blood-brain barrier. Molecular players found only in vertebrates allow the modification of conserved mechanisms as well as the design of novel strategies that can better serve the physiological needs of the vertebrates. These innovations collectively contribute to novel morphogenetic behaviors and unprecedented increases in the complexities of tissue mechanics and functions.
Collapse
Affiliation(s)
- Vivian W. Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana–Champaign, Urbana, IL 61801
| |
Collapse
|
103
|
Schakenraad K, Ernst J, Pomp W, Danen EHJ, Merks RMH, Schmidt T, Giomi L. Mechanical interplay between cell shape and actin cytoskeleton organization. SOFT MATTER 2020; 16:6328-6343. [PMID: 32490503 DOI: 10.1039/d0sm00492h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We investigate the mechanical interplay between the spatial organization of the actin cytoskeleton and the shape of animal cells adhering on micropillar arrays. Using a combination of analytical work, computer simulations and in vitro experiments, we demonstrate that the orientation of the stress fibers strongly influences the geometry of the cell edge. In the presence of a uniformly aligned cytoskeleton, the cell edge can be well approximated by elliptical arcs, whose eccentricity reflects the degree of anisotropy of the cell's internal stresses. Upon modeling the actin cytoskeleton as a nematic liquid crystal, we further show that the geometry of the cell edge feeds back on the organization of the stress fibers by altering the length scale at which these are confined. This feedback mechanism is controlled by a dimensionless number, the anchoring number, representing the relative weight of surface-anchoring and bulk-aligning torques. Our model allows to predict both cellular shape and the internal structure of the actin cytoskeleton and is in good quantitative agreement with experiments on fibroblastoid (GDβ1, GDβ3) and epithelioid (GEβ1, GEβ3) cells.
Collapse
Affiliation(s)
- Koen Schakenraad
- Instituut-Lorentz, Leiden University, P.O. Box 9506, 2300 RA Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
104
|
Actin polymerization downstream of integrins: signaling pathways and mechanotransduction. Biochem J 2020; 477:1-21. [PMID: 31913455 DOI: 10.1042/bcj20170719] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/17/2019] [Accepted: 12/16/2019] [Indexed: 01/04/2023]
Abstract
A cell constantly adapts to its environment. Cell decisions to survive, to proliferate or to migrate are dictated not only by soluble growth factors, but also through the direct interaction of the cell with the surrounding extracellular matrix (ECM). Integrins and their connections to the actin cytoskeleton are crucial for monitoring cell attachment and the physical properties of the substratum. Cell adhesion dynamics are modulated in complex ways by the polymerization of branched and linear actin arrays, which in turn reinforce ECM-cytoskeleton connection. This review describes the major actin regulators, Ena/VASP proteins, formins and Arp2/3 complexes, in the context of signaling pathways downstream of integrins. We focus on the specific signaling pathways that transduce the rigidity of the substrate and which control durotaxis, i.e. directed migration of cells towards increased ECM rigidity. By doing so, we highlight several recent findings on mechanotransduction and put them into a broad integrative perspective that is the result of decades of intense research on the actin cytoskeleton and its regulation.
Collapse
|
105
|
SB772077B (SB77) Alleviated the Aqueous Outflow Resistance Mediated by Cyclic Mechanical Stress in Perfused Human Cadaveric Eyes. Sci Rep 2020; 10:10202. [PMID: 32576873 PMCID: PMC7311383 DOI: 10.1038/s41598-020-67087-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/03/2020] [Indexed: 01/08/2023] Open
Abstract
The intraocular pressure lowering property of a new rho kinase inhibitor, SB772077B (SB77) has been previously demonstrated in perfused human cadaveric eyes. In this study, the efficacy of SB77 in alleviating the aqueous outflow resistance mediated by cyclic mechanical stress in perfused human cadaveric eyes was investigated. A human anterior segment perfusion culture model was used to investigate the effect of cyclic intraocular pressure (IOP) on aqueous outflow facility in presence or absence of SB77. The status of RhoA activation and the downstream effector molecule myosin-light chain phosphorylation (p-MLC) was investigated by Western blot. Cyclic mechanical stress resulted in decrease in aqueous outflow facility (-19.79 ± 4.93%; p = 0.019) in perfused human eyes and treatment with SB77 (50 µM) significantly enhanced outflow facility by 15% (p = 0.05). The increase in outflow facility by SB77 was confirmed with the inactivation of RhoA/ROCK signaling and decreased expression of extracellular matrix markers. SB77 effectively reduced the outflow resistance mediated by cyclic IOP and thus may be a potential clinical candidate for the management of glaucoma.
Collapse
|
106
|
Rat corneal endothelial cell migration during wound repair on the basement membrane depends more on the PI-3K pathway than the cdc-42 pathway or actin stress fibers. Cell Tissue Res 2020; 382:351-366. [DOI: 10.1007/s00441-020-03229-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/01/2020] [Indexed: 12/15/2022]
|
107
|
Keller S, Wörgötter K, Liedek A, Kluger PJ, Bach M, Tovar GEM, Southan A. Azide-Functional Extracellular Matrix Coatings as a Bioactive Platform for Bioconjugation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:26868-26879. [PMID: 32426964 DOI: 10.1021/acsami.0c04579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In recent years, the development and application of decellularized extracellular matrices (ECMs) for use as biomaterials have grown rapidly. These cell-derived matrices (CDMs) represent highly bioactive and biocompatible materials consisting of a complex assembly of biomolecules. Even though CDMs mimic the natural microenvironment of cells in vivo very closely, they still lack specifically addressable functional groups, which are often required to tailor a biomaterial functionality by bioconjugation. To overcome this limitation, metabolic glycoengineering has emerged as a powerful tool to equip CDMs with chemical groups such as azides. These small chemical handles are known for their ability to undergo bioorthogonal click reactions, which represent a desirable reaction type for bioconjugation. However, ECM insolubility makes its processing very challenging. In this contribution, we isolated both the unmodified ECM and azide-modified clickECM by osmotic lysis. In a first step, these matrices were concentrated to remove excessive water from the decellularization step. Next, the hydrogel-like ECM and clickECM films were mechanically fragmentized, resulting in easy to pipette suspensions with fragment sizes ranging from 7.62 to 31.29 μm (as indicated by the mean d90 and d10 values). The biomolecular composition was not impaired as proven by immunohistochemistry. The suspensions were used for the reproducible generation of surface coatings, which proved to be homogeneous in terms of ECM fragment sizes and coating thicknesses (the mean coating thickness was found to be 33.2 ± 7.3 μm). Furthermore, they were stable against fluid-mechanical abrasion in a laminar flow cell. When primary human fibroblasts were cultured on the coated substrates, an increased bioactivity was observed. By conjugating the azides within the clickECM coatings with alkyne-coupled biotin molecules, a bioconjugation platform was obtained, where the biotin-streptavidin interaction could be used. Its applicability was demonstrated by equipping the bioactive clickECM coatings with horseradish peroxidase as a model enzyme.
Collapse
Affiliation(s)
- Silke Keller
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569 Stuttgart, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Katharina Wörgötter
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Anke Liedek
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Petra J Kluger
- School of Applied Chemistry, Reutlingen University, Alteburgstraße 150, 72762 Reutlingen, Germany
| | - Monika Bach
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Günter E M Tovar
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569 Stuttgart, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569 Stuttgart, Germany
| |
Collapse
|
108
|
Keller S, Bakker T, Kimmel B, Rebers L, Götz T, Tovar GEM, Kluger PJ, Southan A. Azido-functionalized gelatin via direct conversion of lysine amino groups by diazo transfer as a building block for biofunctional hydrogels. J Biomed Mater Res A 2020; 109:77-91. [PMID: 32421917 DOI: 10.1002/jbm.a.37008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/08/2020] [Accepted: 04/19/2020] [Indexed: 12/25/2022]
Abstract
Gelatin is one of the most prominent biopolymers in biomedical material research and development. It is frequently used in hybrid hydrogels, which combine the advantageous properties of bio-based and synthetic polymers. To prevent the biological component from leaching out of the hydrogel, the biomolecules can be equipped with azides. Those groups can be used to immobilize gelatin covalently in hydrogels by the highly selective and specific azide-alkyne cycloaddition. In this contribution, we functionalized gelatin with azides at its lysine residues by diazo transfer, which offers the great advantage of only minimal side-chain extension. Approximately 84-90% of the amino groups are modified as shown by 1 H-NMR spectroscopy, 2,4,6-trinitrobenzenesulfonic acid assay as well as Fourier-transform infrared spectroscopy, rheology, and the determination of the isoelectric point. Furthermore, the azido-functional gelatin is incorporated into hydrogels based on poly(ethylene glycol) diacrylate (PEG-DA) at different concentrations (0.6, 3.0, and 5.5%). All hydrogels were classified as noncyctotoxic with significantly enhanced cell adhesion of human fibroblasts on their surfaces compared to pure PEG-DA hydrogels. Thus, the new gelatin derivative is found to be a very promising building block for tailoring the bioactivity of materials.
Collapse
Affiliation(s)
- Silke Keller
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Tomke Bakker
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Benjamin Kimmel
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Lisa Rebers
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Tobias Götz
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Günter E M Tovar
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany.,Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB), Stuttgart, Germany
| | - Petra J Kluger
- School of Applied Chemistry, Reutlingen University, Reutlingen, Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
109
|
Liu Y, Xu L, Hu L, Chen D, Yu L, Li X, Chen H, Zhu J, Chen C, Luo Y, Wang B, Li G. Stearic acid methyl ester promotes migration of mesenchymal stem cells and accelerates cartilage defect repair. J Orthop Translat 2020; 22:81-91. [PMID: 32440503 PMCID: PMC7231966 DOI: 10.1016/j.jot.2019.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/22/2019] [Accepted: 09/25/2019] [Indexed: 01/07/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) can be easily expanded without losing the ability of multilineage differentiation, including oesteogenic, chondrogenic and adipogenic differentiation. These characters make MSCs a promising cell resource for cartilage defect repair. MSCs could be recruited by inflammatory stimulation, then home to the injury tissues. However, its capacity of homing is extremely limited. Thus, it has become extremely necessary to develop an agent or a method, which can be used to enhance the efficiency of MSCs homing. This study investigates the effect of stearic acid methyl ester (SAME) on MSCs mobilisation and cartilage regeneration. Methods MSCs were isolated from femurs of Sprague-Dawley (SD) rats. MTT assay was used to detect effect of SAME on viability of MSCs. Transwell assay and wound healing assay were used to detect effect of SAME on migration of MSCs. RNA-seq, quantitative real-time PCR and western blot were performed to analyze the expression of RNAs and proteins. Colony forming assay and flow cytometry were used to evaluate the effect of SAME on MSCs mobilisation in vivo. A rat cartilage defect model was created to evaluate the effect of SAME on cartilage regeneration. Results We found that SAME could promote the migration of MSCs. Interestingly, we found SAME significantly increased the expression levels of Vav1 in MSCs. On the other hand, the enhanced migration ability of MSCs induced by SAME was retarded by Vav1 small interfering RNA (siRNA) and Rho-associated protein kinase 2 (ROCK2) inhibitor. In addition, we also checked the effect of SAME on mobilisation of MSCs in vivo. The results showed that SAME increased the number of MSCs in peripheral blood and enhanced the capacity of colony formation. Finally, using a cartilage defect model in rats, we found SAME could improve cartilage repair. Conclusion Our study demonstrates that SAME can enhance MSCs migration ability mainly through the Vav1/ROCK2 signaling pathway, which could contribute to the accelerated cartilage regeneration. The translational potential of this article These findings provide evidence that SAME could be used as a therapeutic reagent for MSCs mobilisation and cartilage regeneration.
Collapse
Affiliation(s)
- Yamei Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liuchao Hu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Dongfeng Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lijuan Yu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hongtai Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Junlang Zhu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Chen Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yiwen Luo
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Bin Wang
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510240, China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| |
Collapse
|
110
|
Chen C, Ye C, Xia J, Zhou Y, Wu R. Ezrin T567 phosphorylation regulates migration and invasion of ectopic endometrial stromal cells by changing actin cytoskeleton. Life Sci 2020; 254:117681. [PMID: 32380081 DOI: 10.1016/j.lfs.2020.117681] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/31/2020] [Accepted: 04/14/2020] [Indexed: 01/15/2023]
Abstract
AIMS The enhanced ability of endometrial cell migration and invasion is the foundation for formation of ectopic lesions in endometriosis. Ezrin has been reported to regulate cell motility by remodeling the cytoskeleton. However, little is known about the mechanisms through which ezrin remodels the cytoskeleton and cell structure to promote cell motility in endometriosis. METHODS In our study, expression and distribution of ezrin, and Rho pathway were detected through immunohistochemical analysis. The effects of inhibiting ezrin T567 phosphorylation on Rho signaling pathway and cytoskeleton were investigated through western blot, transmission electron microscopy and immunofluorescence analysis. KEY FINDINGS We found that the expression of ezrin and Rho pathway was higher in ectopic endometrium. NSC305787 inhibited the phosphorylation of ezrin T567, resulting in decreased expression of Rho pathway and reduced filopodia formation in ectopic endometrial stromal cells. SIGNIFICANCE Taken together, our study suggested that ezrin T567 phosphorylation modulated migration and invasion of ectopic ESCs through actin reconstructions, which may serve as a novel therapeutic target in ovarian endometriosis.
Collapse
Affiliation(s)
- Chaolu Chen
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Chaoshuang Ye
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Jianmei Xia
- Department of Obstetrics and Gynecology, Hangzhou First People's Hospital, Zhejiang 310006, China
| | - Yong Zhou
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China
| | - Ruijin Wu
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Zhejiang 310006, China.
| |
Collapse
|
111
|
Li T, Liu X, Xu B, Wu W, Zang Y, Li J, Wei L, Qian Y, Xu H, Xie M, Wang Q, Wang L. SKA1 regulates actin cytoskeleton remodelling via activating Cdc42 and influences the migration of pancreatic ductal adenocarcinoma cells. Cell Prolif 2020; 53:e12799. [PMID: 32232899 PMCID: PMC7162805 DOI: 10.1111/cpr.12799] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/08/2020] [Accepted: 03/04/2020] [Indexed: 12/29/2022] Open
Abstract
Objectives Spindle and kinetochore–associated protein 1(SKA1), originally identified as a protein essential for proper chromosome segregation, has been recently linked to multiple malignancies. This study aimed to explore the biological, clinical role and molecular mechanism of SKA1 in pancreatic carcinogenesis. Materials and Methods SKA1 expression was detected in 145 pancreatic ductal adenocarcinoma (PDAC) specimens by immunohistochemistry. Biological behaviour assays were used to determine the role of SKA1 in PDAC progression in vitro and in vivo. Using isobaric tags for relative and absolute quantitation (iTRAQ), SKA1’s downstream proteins were examined. Moreover, cytochalasin B and ZCL278 were used to explore the changes of SKA1‐induced signalling and cell morphology, with further confirmation by immunoblotting and immunofluorescence assays. Results Increased SKA1 expression was significantly correlated with tumour size and cellular differentiation degree in PDAC tissues. Furthermore, elevated levels of SKA1 reflected shorter overall survival (P = .019). As for biological behaviour, SKA1 acted as a tumour promotor in PDAC, overexpression of SKA1 facilitates cell proliferation, migration and invasion in vitro and in vivo. Mechanistically, we demonstrated that SKA1 enhanced pancreatic cancer aggressiveness by inhibiting G2/M arrest and regulating actin cytoskeleton organization via activating Cdc42. Conclusions This study revealed novel roles for SKA1 as an important regulator of actin cytoskeleton organization and an oncogene in PDAC cells, which may provide insights into developing novel therapeutics.
Collapse
Affiliation(s)
- Tong Li
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Liu
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Xu
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wu
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Li
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lumin Wei
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Qian
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Xu
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingping Xie
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifu Wang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
112
|
Pulido Companys P, Norris A, Bischoff M. Coordination of cytoskeletal dynamics and cell behaviour during Drosophila abdominal morphogenesis. J Cell Sci 2020; 133:jcs235325. [PMID: 32229579 PMCID: PMC7132776 DOI: 10.1242/jcs.235325] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/20/2020] [Indexed: 12/20/2022] Open
Abstract
During morphogenesis, cells exhibit various behaviours, such as migration and constriction, which need to be coordinated. How this is achieved remains elusive. During morphogenesis of the Drosophila adult abdominal epidermis, larval epithelial cells (LECs) migrate directedly before constricting apically and undergoing apoptosis. Here, we study the mechanisms underlying the transition from migration to constriction. We show that LECs possess a pulsatile apical actomyosin network, and that a change in network polarity correlates with behavioural change. Exploring the properties of the contractile network, we find that cell contractility, as determined by myosin activity, has an impact on the behaviour of the network, as well as on cytoskeletal architecture and cell behaviour. Pulsed contractions occur only in cells with intermediate levels of contractility. Furthermore, increasing levels of the small Rho GTPase Rho1 disrupts pulsing, leading to cells that cycle between two states, characterised by a junctional cortical and an apicomedial actin network. Our results highlight that behavioural change relies on tightly controlled cellular contractility. Moreover, we show that constriction can occur without pulsing, raising questions why constricting cells pulse in some contexts but not in others.
Collapse
Affiliation(s)
- Pau Pulido Companys
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9ST, UK
| | - Anneliese Norris
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9ST, UK
| | - Marcus Bischoff
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9ST, UK
| |
Collapse
|
113
|
Galenkamp KMO, Sosicka P, Jung M, Recouvreux MV, Zhang Y, Moldenhauer MR, Brandi G, Freeze HH, Commisso C. Golgi Acidification by NHE7 Regulates Cytosolic pH Homeostasis in Pancreatic Cancer Cells. Cancer Discov 2020; 10:822-835. [PMID: 32200349 DOI: 10.1158/2159-8290.cd-19-1007] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/07/2020] [Accepted: 03/18/2020] [Indexed: 11/16/2022]
Abstract
Cancer cells reprogram their metabolism to meet elevated energy demands and favor glycolysis for energy production. This boost in glycolytic flux supports proliferation, but also generates acid in the form of hydrogen ions that must be eliminated from the cytoplasm to maintain the alkaline intracellular pH (pHi) associated with transformation. To cope with acid production, tumor cells employ ion transport systems, including the family of sodium-hydrogen exchangers (NHE). Here, we identify NHE7 as a novel regulator of pHi in pancreatic ductal adenocarcinoma (PDAC). We determine that NHE7 suppression causes alkalinization of the Golgi, leading to a buildup of cytosolic acid that diminishes tumor cell fitness mainly through the dysregulation of actin. Importantly, NHE7 knockdown in vivo leads to the abrogation of tumor growth. These results identify Golgi acidification as a mechanism to control pHi and point to the regulation of pHi as a possible therapeutic vulnerability in PDAC. SIGNIFICANCE: NHE7 regulates cytosolic pH through Golgi acidification, which points to the Golgi as a "proton sink" for metabolic acid. Disruption of cytosolic pH homeostasis via NHE7 suppression compromises PDAC cell viability and tumor growth.See related commentary by Ward and DeNicola, p. 768.This article is highlighted in the In This Issue feature, p. 747.
Collapse
Affiliation(s)
- Koen M O Galenkamp
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Paulina Sosicka
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Michael Jung
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - M Victoria Recouvreux
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Yijuan Zhang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Matthew R Moldenhauer
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Giovanni Brandi
- Department of Experimental, Diagnostic, and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Hudson H Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Cosimo Commisso
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| |
Collapse
|
114
|
Bhadoria R, Ping K, Lohk C, Järving I, Starkov P. A phenotypic approach to probing cellular outcomes using heterobivalent constructs. Chem Commun (Camb) 2020; 56:4216-4219. [PMID: 32181457 DOI: 10.1039/c9cc09595k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Various conjugation techniques are used to affect the intracellular delivery of bioactive small molecules. However, the ability to track changes in the phenotype when applying these tools remains poorly studied. We addressed this issue by having prepared a focused library of heterobivalent constructs based on Rho kinase inhibitor HA-100. By comparing the induction of the phenotype of interest, cell viability and cellular uptake, we demonstrate that various conjugates indeed lead to divergent cellular outcomes.
Collapse
Affiliation(s)
- Rohit Bhadoria
- Department of Chemistry & Biotechnology, Tallinn University of Technology, 12618 Tallinn, Estonia.
| | | | | | | | | |
Collapse
|
115
|
Bae J, Kumazoe M, Murata K, Fujimura Y, Tachibana H. Procyanidin C1 Inhibits Melanoma Cell Growth by Activating 67-kDa Laminin Receptor Signaling. Mol Nutr Food Res 2020; 64:e1900986. [PMID: 32103628 DOI: 10.1002/mnfr.201900986] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/06/2020] [Indexed: 12/11/2022]
Abstract
SCOPE Procyanidin C1 (PC1) is an epicatechin trimer found mainly in grapes that is reported to provide several health benefits. However, little is known about the molecular mechanisms underlying these benefits. The aim of this study is to demonstrate the molecular mechanisms by which PC1 operates. METHODS AND RESULTS A 67-kDa laminin receptor (67LR) is identified as a cell surface receptor of PC1, with a Kd value of 2.8 µm. PC1 induces an inhibitory effect on growth, accompanied by dephosphorylation of the C-kinase potentiated protein phosphatase-1 inhibitor protein of 17 kDa (CPI17) and myosin regulatory light chain (MRLC) proteins, followed by actin cytoskeleton remodeling in melanoma cells. These actions are mediated by protein kinase A (PKA) and protein phosphatase 2A (PP2A) activation once PC1 is bound to 67LR. CONCLUSION It is demonstrated that PC1 elicits melanoma cell growth inhibition by activating the 67LR/PKA/PP2A/CPI17/MRLC pathway.
Collapse
Affiliation(s)
- Jaehoon Bae
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Kyosuke Murata
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yoshinori Fujimura
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| |
Collapse
|
116
|
Kawahira N, Ohtsuka D, Kida N, Hironaka KI, Morishita Y. Quantitative Analysis of 3D Tissue Deformation Reveals Key Cellular Mechanism Associated with Initial Heart Looping. Cell Rep 2020; 30:3889-3903.e5. [DOI: 10.1016/j.celrep.2020.02.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 08/01/2019] [Accepted: 02/18/2020] [Indexed: 12/18/2022] Open
|
117
|
Shi Z, Ren M, Rockey DC. Myocardin and myocardin-related transcription factor-A synergistically mediate actin cytoskeletal-dependent inhibition of liver fibrogenesis. Am J Physiol Gastrointest Liver Physiol 2020; 318:G504-G517. [PMID: 31928221 PMCID: PMC7099496 DOI: 10.1152/ajpgi.00302.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Activation of hepatic stellate cells (HSCs), characterized by development of a robust actin cytoskeleton and expression of abundant extracellular matrix (ECM) proteins, such as type 1 collagen (COL.1), is a central cellular and molecular event in liver fibrosis. It has been demonstrated that HSCs express both myocardin and myocardin-related transcription factor-A (MRTF-A). However, the biological effects of myocardin and MRTF-A on HSC activation and liver fibrosis, as well as the molecular mechanism under the process, remain unclear. Here, we report that myocardin and MRTF-A's expression and nuclear accumulation are prominently increased during the HSC activation process, accompanied by robust activation of actin cytoskeleton dynamics. Targeting myocardin and MRTF-A binding and function with a novel small molecule, CCG-203971, led to dose-dependent inhibition of HSC actin cytoskeleton dynamics and abrogated multiple functional features of HSC activation (i.e., HSC contraction, migration and proliferation) and decreased COL.1 expression in vitro and liver fibrosis in vivo. Mechanistically, blocking the myocardin and MRTF-A nuclear translocation pathway with CCG-203971 directly inhibited myocardin/MRTF-A-mediated serum response factor (SRF), and Smad2/3 activation in the COL.1α2 promoter and indirectly abrogated actin cytoskeleton-dependent regulation of Smad2/3 and Erk1/2 phosphorylation and their nuclear accumulation. Finally, there was no effect of CCG-203971 on markers of inflammation, suggesting a direct effect of the compound on HSCs and liver fibrosis. These data reveal that myocardin and MRTF-A are two important cotranscriptional factors in HSCs and represent entirely novel therapeutic pathways that might be targeted to treat liver fibrosis.NEW & NOTEWORTHY Myocardin and myocardin-related transcription factor-A (MRTF-A) are upregulated in activated hepatic stellate cells (HSCs) in vitro and in vivo, closely associated with robustly increased actin cytoskeleton remodeling. Targeting myocardin and MRTF-A by CCG-203971 leads to actin cytoskeleton-dependent inhibition of HSC activation, reduced cell contractility, impeded cell migration and proliferation, and decreased COL.1 expression in vitro and in vivo. Dual expression of myocardin and MRTF-A in HSCs may represent novel therapeutic targets in liver fibrosis.
Collapse
Affiliation(s)
- Zengdun Shi
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Mudan Ren
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Don C. Rockey
- Department of Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
118
|
Zhang Y, Hu Y, Li M, Wang J, Guo G, Li F, Yu B, Kou J. The Traditional Chinese Medicine Compound, GRS, Alleviates Blood-Brain Barrier Dysfunction. Drug Des Devel Ther 2020; 14:933-947. [PMID: 32184562 PMCID: PMC7053822 DOI: 10.2147/dddt.s229302] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/10/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Traditional Chinese medicine (TCM) provides unique advantages for treatment of ischemic stroke, an aging-related vascular disease. Shengmai powder (GRS) is composed of three active components, specifically, ginsenoside Rb1, ruscogenin and schisandrin A, at a ratio of 6:0.75:6. The main objective of this study was to evaluate the effects of GRS on blood–brain barrier (BBB) dysfunction under conditions of middle cerebral artery occlusion/reperfusion (MCAO/R). Methods C57BL/6J mice subjected to MCAO/R were used as a model to assess the protective effects of varying doses of GRS (6.4, 12.8, and 19.2 mg/kg) on BBB dysfunction. Results GRS reduced cerebral infarct volume and degree of brain tissue damage, improved behavioral scores, decreased water content and BBB permeability, and restored cerebral blood flow. Moreover, GRS promoted expression of zona occludens-1 (ZO-1) and claudin-5 while inhibiting matrix metalloproteinase 2/9 (MMP-2/9) expression and myosin light chain (MLC) phosphorylation. In vitro, GRS (1, 10, and 100 ng/mL) enhanced the viability of bEnd.3 cells subjected to oxygen glucose deprivation/reoxygenation (OGD/R) and decreased sodium fluorescein permeability. Conclusion Consistent with in vivo findings, ZO-1 and claudin-5 were significantly upregulated by GRS in bEnd.3 cells under OGD/R and MMP-2/9 levels and MLC phosphorylation reduced through the Rho-associated coil-forming protein kinase (ROCK)/cofilin signaling pathway. Based on the collective findings, we propose that the TCM compound, GRS, plays a protective role against I/R-induced BBB dysfunction.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yang Hu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Min Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jieman Wang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Gengshuo Guo
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
119
|
Werner M, Kurniawan NA, Bouten CVC. Cellular Geometry Sensing at Different Length Scales and its Implications for Scaffold Design. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E963. [PMID: 32098110 PMCID: PMC7078773 DOI: 10.3390/ma13040963] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
Geometrical cues provided by the intrinsic architecture of tissues and implanted biomaterials have a high relevance in controlling cellular behavior. Knowledge of how cells sense and subsequently respond to complex geometrical cues of various sizes and origins is needed to understand the role of the architecture of the extracellular environment as a cell-instructive parameter. This is of particular interest in the field of tissue engineering, where the success of scaffold-guided tissue regeneration largely depends on the formation of new tissue in a native-like organization in order to ensure proper tissue function. A well-considered internal scaffold design (i.e., the inner architecture of the porous structure) can largely contribute to the desired cell and tissue organization. Advances in scaffold production techniques for tissue engineering purposes in the last years have provided the possibility to accurately create scaffolds with defined macroscale external and microscale internal architectures. Using the knowledge of how cells sense geometrical cues of different size ranges can drive the rational design of scaffolds that control cellular and tissue architecture. This concise review addresses the recently gained knowledge of the sensory mechanisms of cells towards geometrical cues of different sizes (from the nanometer to millimeter scale) and points out how this insight can contribute to informed architectural scaffold designs.
Collapse
Affiliation(s)
- Maike Werner
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Nicholas A. Kurniawan
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Carlijn V. C. Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| |
Collapse
|
120
|
Khong ZJ, Lai SK, Koh CG, Geifman-Shochat S, Li HY. A novel function of AAA-ATPase p97/VCP in the regulation of cell motility. Oncotarget 2020; 11:74-85. [PMID: 32002125 PMCID: PMC6967774 DOI: 10.18632/oncotarget.27419] [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: 08/19/2019] [Accepted: 12/21/2019] [Indexed: 11/25/2022] Open
Abstract
High level of the multifunctional AAA-ATPase p97/VCP is often correlated to the development of cancer; however, the underlying mechanism is not understood completely. Here, we report a novel function of p97/VCP in actin regulation and cell motility. We found that loss of p97/VCP promotes stabilization of F-actin, which cannot be reversed by actin-destabilizing agent, Cytochalasin D. Live-cell imaging demonstrated reduced actin dynamics in p97/VCP-knockdown cells, leading to compromised cell motility. We further examined the underlying mechanism and found elevated RhoA protein levels along with increased phosphorylation of its downstream effectors, ROCK, LIMK, and MLC upon the knockdown of p97/VCP. Since p97/VCP is indispensable in the ubiquitination-dependent protein degradation pathway, we investigated if the loss of p97/VCP hinders the protein degradation of RhoA. Knockdown of p97/VCP resulted in a higher amount of ubiquitinated RhoA, suggesting p97/VCP involvement in the proteasome-dependent protein degradation pathway. Finally, we found that p97/VCP interacts with FBXL19, a molecular chaperone known to guide ubiquitinated RhoA for proteasomal degradation. Reduction of p97/VCP may result in the accumulation of RhoA which, in turn, enhances cytoplasmic F-actin formation. In summary, our study uncovered a novel function of p97/VCP in actin regulation and cell motility via the Rho-ROCK dependent pathway which provides fundamental insights into how p97/VCP is involved in cancer development.
Collapse
Affiliation(s)
- Zi-Jia Khong
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Soak-Kuan Lai
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Cheng-Gee Koh
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Susana Geifman-Shochat
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| | - Hoi-Yeung Li
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
121
|
Kwan SH, Aziz NHKA, Ismail MN. Bioactive Proteins in Channa striata Promote Wound Healing through Angiogenesis and Cell Proliferation. Protein Pept Lett 2020; 27:48-59. [PMID: 31362651 PMCID: PMC6978642 DOI: 10.2174/0929866526666190730121711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/17/2019] [Accepted: 06/25/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Channa striata are speculated to contain bioactive proteins with the ability to enhancing wound healing. It is commonly consumed after surgery for a faster recovery of the wound. OBJECTIVE To identify the bioactive proteins and evaluate their ability in cell proliferation and angiogenesis promotion. MATERIAL AND METHODS Freeze-Dried Water Extracts (FDWE) and Spray-Dried Water Extracts (SDWE) of C. striata were tested with MTT assay using EA.hy926 endothelial cell line and ex-vivo aortic ring assay. Later the proteins were fractionated and analysed using an LC-QTOF mass spectrometer. The data generated were matched with human gene database for protein similarity and pathway identification. RESULTS Both samples have shown positive cell proliferation and pro-angiogenic activity. Four essential proteins/genes were identified, which are collagen type XI, actin 1, myosin light chain and myosin heavy chain. The pathways discovered that related to these proteins are integrin pathway, Slit-Robo signalling pathway and immune response C-C Chemokine Receptor-3 signalling pathway in eosinophils, which contribute towards wound healing mechanism. CONCLUSIONS The results presented have demonstrated that C. striata FDWE and SDWE protein fractions contain bioactive proteins that are highly similar to human proteins and thus could be involved in the wound healing process via specific biological pathways.
Collapse
Affiliation(s)
| | | | - Mohd Nazri Ismail
- Address correspondence to this author at the Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800, USM, Penang,
Malaysia; Tel: +604-6532694; E-mail:
| |
Collapse
|
122
|
Zhao S, Cai J, Zhang X, Cui J, Jiu Y. Different formins restrict localization of distinct tropomyosins on dorsal stress fibers in osteosarcoma cells. Cytoskeleton (Hoboken) 2019; 77:16-24. [PMID: 31820591 DOI: 10.1002/cm.21588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/01/2019] [Accepted: 12/04/2019] [Indexed: 12/25/2022]
Abstract
Formins and tropomyosins (Tpms) are two central components of the microfilaments. Formins are involved in the nucleation and polymerization of actin filaments, and Tpms form along the actin stress fibers to regulate their dynamics. However, the correlation between formins and Tpms remains unclear. Here, we elucidated the function of distinct formins and their specific regulation to the subcellular-localization of Tpm isoforms on dorsal stress fibers in human osteosarcoma cells. Knockdown of individual formin isoform led to varied defects in actin stress fiber network, but did not affect the expression level of other formin isoforms and Tpms. Further investigation showed that different formins regulated distinct Tpm isoforms in decorating dorsal stress fibers. Specifically, DAAM1 and FHOD1 restricted the distal end expression of Tpm3.1; INF2 controlled the approximate localization of Tpm4.2; and Dia1 partially modulated the dorsal localization of Tpm1.6. Taken together, these data provide microscopy experimental evidence that different formins restrict the localization of distinct Tpm isoforms on dorsal actin stress fibers.
Collapse
Affiliation(s)
- Shuangshuang Zhao
- The Joint Center for Infection and Immunity between Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center (Guangzhou, 510623, China) and Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jinping Cai
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Xin Zhang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Jie Cui
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yaming Jiu
- The Joint Center for Infection and Immunity between Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center (Guangzhou, 510623, China) and Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
123
|
Nakuluri K, Nishad R, Mukhi D, Kumar S, Nakka VP, Kolligundla LP, Narne P, Natuva SSK, Phanithi PB, Pasupulati AK. Cerebral ischemia induces TRPC6 via HIF1α/ZEB2 axis in the glomerular podocytes and contributes to proteinuria. Sci Rep 2019; 9:17897. [PMID: 31784544 PMCID: PMC6884642 DOI: 10.1038/s41598-019-52872-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022] Open
Abstract
Podocytes are specialized cells of the glomerulus and key component of the glomerular filtration apparatus (GFA). GFA regulates the permselectivity and ultrafiltration of blood. The mechanism by which the integrity of the GFA is compromised and manifest in proteinuria during ischemic stroke remains enigmatic. We investigated the mechanism of ischemic hypoxia-induced proteinuria in a middle cerebral artery occlusion (MCAO) model. Ischemic hypoxia resulted in the accumulation of HIF1α in the podocytes that resulted in the increased expression of ZEB2 (Zinc finger E-box-binding homeobox 2). ZEB2, in turn, induced TRPC6 (transient receptor potential cation channel, subfamily C, member 6), which has increased selectivity for calcium. Elevated expression of TRPC6 elicited increased calcium influx and aberrant activation of focal adhesion kinase (FAK) in podocytes. FAK activation resulted in the stress fibers reorganization and podocyte foot process effacement. Our study suggests overactive HIF1α/ZEB2 axis during ischemic-hypoxia raises intracellular calcium levels via TRPC6 and consequently altered podocyte structure and function thus contributes to proteinuria.
Collapse
Affiliation(s)
| | - Rajkishor Nishad
- Department of Biochemistry, University of Hyderabad, Hyderabad, 500046, India
| | - Dhanunjay Mukhi
- Department of Biochemistry, University of Hyderabad, Hyderabad, 500046, India
| | - Sireesh Kumar
- Department of Biotechnology & Bioinformatics, University of Hyderabad, Hyderabad, 500046, India
| | - Venkata P Nakka
- Department of Biochemistry, Acharya Nagarjuna University, Guntur, 522510, India
| | | | - Parimala Narne
- Department of Biotechnology & Bioinformatics, University of Hyderabad, Hyderabad, 500046, India
| | | | - Prakash Babu Phanithi
- Department of Biotechnology & Bioinformatics, University of Hyderabad, Hyderabad, 500046, India.
| | - Anil K Pasupulati
- Department of Biochemistry, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
124
|
Contour Models of Cellular Adhesion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31612451 DOI: 10.1007/978-3-030-17593-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
The development of traction-force microscopy, in the past two decades, has created the unprecedented opportunity of performing direct mechanical measurements on living cells as they adhere or crawl on uniform or micro-patterned substrates. Simultaneously, this has created the demand for a theoretical framework able to decipher the experimental observations, shed light on the complex biomechanical processes that govern the interaction between the cell and the extracellular matrix and offer testable predictions. Contour models of cellular adhesion, represent one of the simplest and yet most insightful approach in this problem. Rooted in the paradigm of active matter, these models allow to explicitly determine the shape of the cell edge and calculate the traction forces experienced by the substrate, starting from the internal and peripheral contractile stresses as well as the passive restoring forces and bending moments arising within the actin cortex and the plasma membrane. In this chapter I provide a general overview of contour models of cellular adhesion and review the specific cases of cells equipped with isotropic and anisotropic actin cytoskeleton as well as the role of bending elasticity.
Collapse
|
125
|
Zaoui K, Rajadurai CV, Duhamel S, Park M. Arf6 regulates RhoB subcellular localization to control cancer cell invasion. J Cell Biol 2019; 218:3812-3826. [PMID: 31591185 PMCID: PMC6829653 DOI: 10.1083/jcb.201806111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 12/21/2018] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
The ADP-ribosylation factor 6 (Arf6) is a small GTPase that regulates endocytic recycling processes in concert with various effectors. Arf6 controls cytoskeletal organization and membrane trafficking; however, the detailed mechanisms of regulation remain poorly understood. Here, we report that Arf6 forms a complex with RhoB. The interaction between RhoB and Arf6 is mediated by the GCI (glycine, cysteine, and isoleucine) residues (188-190) of RhoB. Specific targeting of Arf6 to plasma membrane or mitochondrial membranes promotes recruitment and colocalization of RhoB to these membrane microdomains. Arf6 depletion promotes the loss of RhoB from endosomal membranes and leads to RhoB degradation through an endolysosomal pathway. This results in defective actin and focal adhesion dynamics and increased 3D cell migration upon activation of the Met receptor tyrosine kinase. Our findings identify a novel regulatory mechanism for RhoB localization and stability by Arf6 and establish the strict requirement of Arf6 for RhoB-specific subcellular targeting to endosomes and biological functions.
Collapse
Affiliation(s)
- Kossay Zaoui
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Charles V Rajadurai
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Stéphanie Duhamel
- Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Morag Park
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada .,Rosalind and Morris Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
126
|
Casalou C, Faustino A, Silva F, Ferreira IC, Vaqueirinho D, Ferreira A, Castanheira P, Barona T, Ramalho JS, Serpa J, Félix A, Barral DC. Arl13b Regulates Breast Cancer Cell Migration and Invasion by Controlling Integrin-Mediated Signaling. Cancers (Basel) 2019; 11:cancers11101461. [PMID: 31569511 PMCID: PMC6826833 DOI: 10.3390/cancers11101461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/13/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the first cause of cancer-related mortality among women worldwide, according to the most recent estimates. This mortality is mainly caused by the tumors' ability to form metastases. Cancer cell migration and invasion are essential for metastasis and rely on the interplay between actin cytoskeleton remodeling and cell adhesion. Therefore, understanding the mechanisms by which cancer cell invasion is controlled may provide new strategies to impair cancer progression. We investigated the role of the ADP-ribosylation factor (Arf)-like (Arl) protein Arl13b in breast cancer cell migration and invasion in vitro, using breast cancer cell lines and in vivo, using mouse orthotopic models. We show that Arl13b silencing inhibits breast cancer cell migration and invasion in vitro, as well as cancer progression in vivo. We also observed that Arl13b is upregulated in breast cancer cell lines and patient tissue samples. Moreover, we found that Arl13b localizes to focal adhesions (FAs) and interacts with β3-integrin. Upon Arl13b silencing, β3-integrin cell surface levels and FA size are increased and integrin-mediated signaling is inhibited. Therefore, we uncover a role for Arl13b in breast cancer cell migration and invasion and provide a new mechanism for how ARL13B can function as an oncogene, through the modulation of integrin-mediated signaling.
Collapse
Affiliation(s)
- Cristina Casalou
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
| | - Alexandra Faustino
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
- ProRegeM-PhD Program in Mechanisms of Disease and Regenerative Medicine, 1169-056 Lisboa, Portugal.
| | - Fernanda Silva
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), 1099-023 Lisboa, Portugal.
| | - Inês C Ferreira
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
| | - Daniela Vaqueirinho
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
- Institute for Research in Biomedicine, CH-6500 Bellinzona, Switzerland.
| | - Andreia Ferreira
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
| | - Pedro Castanheira
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
- ProRegeM-PhD Program in Mechanisms of Disease and Regenerative Medicine, 1169-056 Lisboa, Portugal.
| | - Teresa Barona
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
| | - José S Ramalho
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
| | - Jacinta Serpa
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), 1099-023 Lisboa, Portugal.
| | - Ana Félix
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), 1099-023 Lisboa, Portugal.
| | - Duarte C Barral
- CEDOC, NOVA Medical School| Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-82 Lisboa, Portugal.
| |
Collapse
|
127
|
Mohammed M, Thurgood P, Gilliam C, Nguyen N, Pirogova E, Peter K, Khoshmanesh K, Baratchi S. Studying the Response of Aortic Endothelial Cells under Pulsatile Flow Using a Compact Microfluidic System. Anal Chem 2019; 91:12077-12084. [DOI: 10.1021/acs.analchem.9b03247] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Mokhaled Mohammed
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| | | | - Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | | | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| |
Collapse
|
128
|
Taale M, Krüger D, Ossei-Wusu E, Schütt F, Rehman MAU, Mishra YK, Marx J, Stock N, Fiedler B, Boccaccini AR, Willumeit-Römer R, Adelung R, Selhuber-Unkel C. Systematically Designed Periodic Electrophoretic Deposition for Decorating 3D Carbon-Based Scaffolds with Bioactive Nanoparticles. ACS Biomater Sci Eng 2019; 5:4393-4404. [PMID: 33438405 DOI: 10.1021/acsbiomaterials.9b00102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The coating of porous scaffolds with nanoparticles is crucial in many applications, for example to generate scaffolds for catalysis or to make scaffolds bioactive. A standard and well-established method for coating surfaces with charged nanoparticles is electrophoresis, but when used on porous scaffolds, this method often leads to a blockage of the pores so that only the outermost layers of the scaffolds are coated. In this study, the electrophoretic coating process is monitored in situ and the kinetics of nanoparticle deposition are investigated. This concept can be extended to design a periodic electrophoretic deposition (PEPD) strategy, thus avoiding the typical blockage of surface pores. In the present work we demonstrate successful and homogeneous electrophoretic deposition of hydroxyapatite nanoparticles (HAn, diameter ≤200 nm) on a fibrous graphitic 3D structure (ultralightweight aerographite) using the PEPD strategy. The microfilaments of the resulting scaffold are covered with HAn both internally and on the surface. Furthermore, protein adsorption assays and cell proliferation assays were carried out and revealed that the HAn-decorated aerographite scaffolds are biocompatible. The HAn decoration of the scaffolds also significantly increases the alkaline phosphatase activity of osteoblast cells, showing that the scaffolds are able to promote their osteoblastic activity.
Collapse
Affiliation(s)
- Mohammadreza Taale
- Biocompatible Nanomaterials, Institute for Materials Science, Kiel University, Kaiserstr. 2, D-24143 Kiel, Germany
| | - Diana Krüger
- Helmholtz-Zentrum Geesthacht, Institute of Materials Research, Division Metallic Biomaterials, Max-Planck-Str. 1, D-21502 Geesthacht, Germany
| | - Emmanuel Ossei-Wusu
- Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiserstr. 2, D-24143 Kiel, Germany
| | - Fabian Schütt
- Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiserstr. 2, D-24143 Kiel, Germany
| | - Muhammad Atiq Ur Rehman
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany.,Department of Materials Science and Engineering, Institute of Space Technology Islamabad, 1, Islamabad Highway, Islamabad 44000, Pakistan
| | - Yogendra Kumar Mishra
- Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiserstr. 2, D-24143 Kiel, Germany
| | - Janik Marx
- Institute of Polymer and Composites, Hamburg University of Technology, Denickestr. 15, D-21073 Hamburg, Germany
| | - Norbert Stock
- Institute of Inorganic Chemistry, Kiel University, Max-Eyth Straße 2, D-24118 Kiel, Germany
| | - Bodo Fiedler
- Institute of Polymer and Composites, Hamburg University of Technology, Denickestr. 15, D-21073 Hamburg, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Regine Willumeit-Römer
- Helmholtz-Zentrum Geesthacht, Institute of Materials Research, Division Metallic Biomaterials, Max-Planck-Str. 1, D-21502 Geesthacht, Germany
| | - Rainer Adelung
- Functional Nanomaterials, Institute for Materials Science, Kiel University, Kaiserstr. 2, D-24143 Kiel, Germany
| | - Christine Selhuber-Unkel
- Biocompatible Nanomaterials, Institute for Materials Science, Kiel University, Kaiserstr. 2, D-24143 Kiel, Germany
| |
Collapse
|
129
|
Zhovmer AS, Tabdanov ED, Miao H, Wen H, Chen J, Luo X, Ma X, Provenzano PP, Adelstein RS. The role of nonmuscle myosin 2A and 2B in the regulation of mesenchymal cell contact guidance. Mol Biol Cell 2019; 30:1961-1973. [PMID: 31318315 PMCID: PMC6727766 DOI: 10.1091/mbc.e19-01-0071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Contact guidance refers to the ability of cells to sense the geometrical features of the microenvironment and respond by changing their shape and adopting the appropriate orientation. Inhibition and ablation of nonmuscle myosin 2 (NM2) paralogues have demonstrated their importance for contact guidance. However, the specific roles of the NM2 paralogues have not been systematically studied. In this work we use micropatterned substrates to examine the roles of NM2A and NM2B and to elucidate the relationship of the microenvironment, actomyosin, and microtubules in contact guidance. We show that contact guidance is preserved following loss of NM2B and that expression of NM2A alone is sufficient to establish an appropriate orientation of the cells. Loss of NM2B and overexpression of NM2A result in a prominent cell polarization that is found to be linked to the increased alignment of microtubules with the actomyosin scaffold. Suppression of actomyosin with blebbistatin reduces cell polarity on a flat surface, but not on a surface with contact guidance cues. This indicates that the lost microtubule-actomyosin interactions are compensated for by microtubule-microenvironment interactions, which are sufficient to establish cell polarity through contact guidance.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Erdem D Tabdanov
- Laboratory for Engineering in Oncology, University of Minnesota, Minneapolis, MN 55455
| | - Houxun Miao
- Imaging Physics Laboratory, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Han Wen
- Imaging Physics Laboratory, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Jinqiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, Bethesda, MD 20892
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, Bethesda, MD 20892
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Paolo P Provenzano
- Laboratory for Engineering in Oncology, University of Minnesota, Minneapolis, MN 55455
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| |
Collapse
|
130
|
Shams H, Hoffman BD, Mofrad MRK. The "Stressful" Life of Cell Adhesion Molecules: On the Mechanosensitivity of Integrin Adhesome. J Biomech Eng 2019; 140:2667887. [PMID: 29272321 DOI: 10.1115/1.4038812] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Indexed: 02/06/2023]
Abstract
Cells have evolved into complex sensory machines that communicate with their microenvironment via mechanochemical signaling. Extracellular mechanical cues trigger complex biochemical pathways in the cell, which regulate various cellular processes. Integrin-mediated focal adhesions (FAs) are large multiprotein complexes, also known as the integrin adhesome, that link the extracellular matrix (ECM) to the actin cytoskeleton, and are part of powerful intracellular machinery orchestrating mechanotransduction pathways. As forces are transmitted across FAs, individual proteins undergo structural and functional changes that involve a conversion of chemical to mechanical energy. The local composition of early adhesions likely defines the regional stress levels and determines the type of newly recruited proteins, which in turn modify the local stress distribution. Various approaches have been used for detecting and exploring molecular mechanisms through which FAs are spatiotemporally regulated, however, many aspects are yet to be understood. Current knowledge on the molecular mechanisms of mechanosensitivity in adhesion proteins is discussed herein along with important questions yet to be addressed, are discussed.
Collapse
Affiliation(s)
- Hengameh Shams
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA 94720-1762
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, 208A Stanley Hall #1762, Berkeley, CA 94720-1762.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, CA 94720 e-mail:
| |
Collapse
|
131
|
Chandrasekaran A, Upadhyaya A, Papoian GA. Remarkable structural transformations of actin bundles are driven by their initial polarity, motor activity, crosslinking, and filament treadmilling. PLoS Comput Biol 2019; 15:e1007156. [PMID: 31287817 PMCID: PMC6615854 DOI: 10.1371/journal.pcbi.1007156] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022] Open
Abstract
Bundled actin structures play a key role in maintaining cellular shape, in aiding force transmission to and from extracellular substrates, and in affecting cellular motility. Recent studies have also brought to light new details on stress generation, force transmission and contractility of actin bundles. In this work, we are primarily interested in the question of what determines the stability of actin bundles and what network geometries do unstable bundles eventually transition to. To address this problem, we used the MEDYAN mechano-chemical force field, modeling several micron-long actin bundles in 3D, while accounting for a comprehensive set of chemical, mechanical and transport processes. We developed a hierarchical clustering algorithm for classification of the different long time scale morphologies in our study. Our main finding is that initially unipolar bundles are significantly more stable compared with an apolar initial configuration. Filaments within the latter bundles slide easily with respect to each other due to myosin activity, producing a loose network that can be subsequently severely distorted. At high myosin concentrations, a morphological transition to aster-like geometries was observed. We also investigated how actin treadmilling rates influence bundle dynamics, and found that enhanced treadmilling leads to network fragmentation and disintegration, while this process is opposed by myosin and crosslinking activities. Interestingly, treadmilling bundles with an initial apolar geometry eventually evolve to a whole gamut of network morphologies based on relative positions of filament ends, such as sarcomere-like organization. We found that apolar bundles show a remarkable sensitivity to environmental conditions, which may be important in enabling rapid cytoskeletal structural reorganization and adaptation in response to intracellular and extracellular cues.
Collapse
Affiliation(s)
- Aravind Chandrasekaran
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, United States of America
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, United States of America
| | - Arpita Upadhyaya
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, United States of America
- Department of Physics, University of Maryland, College Park, United States of America
| | - Garegin A. Papoian
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, United States of America
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, United States of America
| |
Collapse
|
132
|
Hu S, Grobe H, Guo Z, Wang YH, Doss BL, Pan M, Ladoux B, Bershadsky AD, Zaidel-Bar R. Reciprocal regulation of actomyosin organization and contractility in nonmuscle cells by tropomyosins and alpha-actinins. Mol Biol Cell 2019; 30:2025-2036. [PMID: 31216217 PMCID: PMC6727768 DOI: 10.1091/mbc.e19-02-0082] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Contractile arrays of actin and myosin II filaments drive many essential processes in nonmuscle cells, including migration and adhesion. Sequential organization of actin and myosin along one dimension is followed by expansion into a two-dimensional network of parallel actomyosin fibers, in which myosin filaments are aligned to form stacks. The process of stack formation has been studied in detail. However, factors that oppose myosin stack formation have not yet been described. Here, we show that tropomyosins act as negative regulators of myosin stack formation. Knockdown of any or all tropomyosin isoforms in rat embryonic fibroblasts resulted in longer and more numerous myosin stacks and a highly ordered actomyosin organization. The molecular basis for this, we found, is the competition between tropomyosin and alpha-actinin for binding actin. Surprisingly, excessive order in the actomyosin network resulted in smaller focal adhesions, lower tension within the network, and smaller traction forces. Conversely, disordered actomyosin bundles induced by alpha-actinin knockdown led to higher than normal tension and traction forces. Thus, tropomyosin acts as a check on alpha-actinin to achieve intermediate levels of myosin stacks matching the force requirements of the cell.
Collapse
Affiliation(s)
- Shiqiong Hu
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Hanna Grobe
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Yafo 6997801, Israel
| | - Zhenhuan Guo
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Yu-Hsiu Wang
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Bryant L Doss
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Meng Pan
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Benoit Ladoux
- Institut Jacques Monod, Université de Paris and CNRS, 75205 Paris CEDEX 13, France
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore 117411.,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronen Zaidel-Bar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Yafo 6997801, Israel
| |
Collapse
|
133
|
Abstract
The interactions of cytoskeletal actin filaments with myosin family motors are essential for the integrity and function of eukaryotic cells. They support a wide range of force-dependent functions. These include mechano-transduction, directed transcellular transport processes, barrier functions, cytokinesis, and cell migration. Despite the indispensable role of tropomyosins in the generation and maintenance of discrete actomyosin-based structures, the contribution of individual cytoskeletal tropomyosin isoforms to the structural and functional diversification of the actin cytoskeleton remains a work in progress. Here, we review processes that contribute to the dynamic sorting and targeted distribution of tropomyosin isoforms in the formation of discrete actomyosin-based structures in animal cells and their effects on actin-based motility and contractility.
Collapse
|
134
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
135
|
Tovar-Lopez F, Thurgood P, Gilliam C, Nguyen N, Pirogova E, Khoshmanesh K, Baratchi S. A Microfluidic System for Studying the Effects of Disturbed Flow on Endothelial Cells. Front Bioeng Biotechnol 2019; 7:81. [PMID: 31111027 PMCID: PMC6499196 DOI: 10.3389/fbioe.2019.00081] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
Arterial endothelium experience physical stress associated with blood flow and play a central role in maintaining vascular integrity and homeostasis in response to hemodynamic forces. Blood flow within vessels is generally laminar and streamlined. However, abrupt changes in the vessel geometry due to branching, sharp turns or stenosis can disturb the laminar blood flow, causing secondary flows in the form of vortices. Such disturbed flow patterns activate pro-inflammatory phenotypes in endothelial cells, damaging the endothelial layer and can lead to atherosclerosis and thrombosis. Here, we report a microfluidic system with integrated ridge-shaped obstacles for generating controllable disturbed flow patterns. This system is used to study the effect of disturbed flow on the cytoskeleton remodeling and nuclear shape and size of cultured human aortic endothelial cells. Our results demonstrate that the generated disturbed flow changes the orientation angle of actin stress fibers and reduces the nuclear size while increases the nuclear circularity.
Collapse
Affiliation(s)
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | | | - Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, VIC, Australia
| | | | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
136
|
MKL1-actin pathway restricts chromatin accessibility and prevents mature pluripotency activation. Nat Commun 2019; 10:1695. [PMID: 30979898 PMCID: PMC6461646 DOI: 10.1038/s41467-019-09636-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
Actin cytoskeleton is well-known for providing structural/mechanical support, but whether and how it regulates chromatin and cell fate reprogramming is far less clear. Here, we report that MKL1, the key transcriptional co-activator of many actin cytoskeletal genes, regulates genomic accessibility and cell fate reprogramming. The MKL1-actin pathway weakens during somatic cell reprogramming by pluripotency transcription factors. Cells that reprogram efficiently display low endogenous MKL1 and inhibition of actin polymerization promotes mature pluripotency activation. Sustained MKL1 expression at a level seen in typical fibroblasts yields excessive actin cytoskeleton, decreases nuclear volume and reduces global chromatin accessibility, stalling cells on their trajectory toward mature pluripotency. In addition, the MKL1-actin imposed block of pluripotency can be bypassed, at least partially, when the Sun2-containing linker of the nucleoskeleton and cytoskeleton (LINC) complex is inhibited. Thus, we unveil a previously unappreciated aspect of control on chromatin and cell fate reprogramming exerted by the MKL1-actin pathway. MKL1 is a key transcriptional co-activator of actin cytoskeleton genes. Here the authors show that MKL1 activation in somatic cells reduces chromatin accessibility and hinders full reprogramming to pluripotency. Reduction of MKL1, disruption of actin cytoskeleton and its links to the nucleus relieve this repression.
Collapse
|
137
|
Regulation of Actin Dynamics in the C. elegans Somatic Gonad. J Dev Biol 2019; 7:jdb7010006. [PMID: 30897735 PMCID: PMC6473838 DOI: 10.3390/jdb7010006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/13/2019] [Accepted: 03/15/2019] [Indexed: 11/25/2022] Open
Abstract
The reproductive system of the hermaphroditic nematode C. elegans consists of a series of contractile cell types—including the gonadal sheath cells, the spermathecal cells and the spermatheca–uterine valve—that contract in a coordinated manner to regulate oocyte entry and exit of the fertilized embryo into the uterus. Contraction is driven by acto-myosin contraction and relies on the development and maintenance of specialized acto-myosin networks in each cell type. Study of this system has revealed insights into the regulation of acto-myosin network assembly and contractility in vivo.
Collapse
|
138
|
Temporal-controlled bioactive molecules releasing core-shell nano-system for tissue engineering strategies in endodontics. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:11-20. [PMID: 30844574 DOI: 10.1016/j.nano.2019.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 01/09/2023]
Abstract
Temporal-controlled release of bioactive molecules is of key importance in the clinical translation of tissue engineering techniques. We engineered a core-shell nano-system (TD-NS) that sequentially released transforming growth factor-β1 (TGF-β1), a chemotactic/proliferating growth factor and dexamethasone (Dex), an osteo/odontogenic agent in a temporal-controlled manner. In stage-1, there was a rapid release of TGF-β1, reaching a concentration of 2 ng/mL of TGF-β1 in 7 days to 14 days, which tapers subsequently. In stage-2, Dex was released linearly from 9 days to 28 days. The TD-NS group showed a significantly higher (P < 0.05) osteo/odontogenic differentiation compared to the control and free TGF-β1 group (Free-TD) that was further corroborated with animal models/histochemical examination. The findings from this study highlighted the potential of temporal-controlled delivery of TGF-β1 and Dex from a single nano-carrier to direct spatial and temporal-control for a cell-free tissue engineering strategy in the treatment of apical periodontitis.
Collapse
|
139
|
Duciel L, Anezo O, Mandal K, Laurent C, Planque N, Coquelle FM, Gentien D, Manneville JB, Saule S. Protein tyrosine phosphatase 4A3 (PTP4A3/PRL-3) promotes the aggressiveness of human uveal melanoma through dephosphorylation of CRMP2. Sci Rep 2019; 9:2990. [PMID: 30816227 PMCID: PMC6395723 DOI: 10.1038/s41598-019-39643-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
Uveal melanoma (UM) is an aggressive tumor in which approximately 50% of patients develop metastasis. Expression of the PTP4A3 gene, encoding a phosphatase, is predictive of poor patient survival. PTP4A3 expression in UM cells increases their migration in vitro and invasiveness in vivo. Here, we show that CRMP2 is mostly dephosphorylated on T514 in PTP4A3 expressing cells. We also demonstrate that inhibition of CRMP2 expression in UM cells expressing PTP4A3 increases their migration in vitro and invasiveness in vivo. This phenotype is accompanied by modifications of the actin microfilament network, with shortened filaments, whereas cells with a inactive mutant of the phosphatase do not show the same behavior. In addition, we showed that the cell cytoplasm becomes stiffer when CRMP2 is downregulated or PTP4A3 is expressed. Our results suggest that PTP4A3 acts upstream of CRMP2 in UM cells to enhance their migration and invasiveness and that a low level of CRMP2 in tumors is predictive of poor patient survival.
Collapse
Affiliation(s)
- Laura Duciel
- Institut Curie, PSL Research University, CNRS, INSERM, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS, INSERM, Orsay, France
| | - Océane Anezo
- Institut Curie, PSL Research University, CNRS, INSERM, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS, INSERM, Orsay, France
| | - Kalpana Mandal
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, 19104, USA
| | | | - Nathalie Planque
- Institut Curie, PSL Research University, CNRS, INSERM, Orsay, France.,Université Paris Diderot, Sorbonne Paris Cité, France
| | - Frédéric M Coquelle
- Institut Curie, PSL Research University, CNRS, INSERM, Orsay, France.,Université Paris Sud, Université Paris-Saclay, CNRS, INSERM, Orsay, France
| | - David Gentien
- Institut Curie, PSL Research University, Translational Research Departement, Genomics Platform, Paris, France
| | | | - Simon Saule
- Institut Curie, PSL Research University, CNRS, INSERM, Orsay, France. .,Université Paris Sud, Université Paris-Saclay, CNRS, INSERM, Orsay, France.
| |
Collapse
|
140
|
Schierbaum N, Rheinlaender J, Schäffer TE. Combined atomic force microscopy (AFM) and traction force microscopy (TFM) reveals a correlation between viscoelastic material properties and contractile prestress of living cells. SOFT MATTER 2019; 15:1721-1729. [PMID: 30657157 DOI: 10.1039/c8sm01585f] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Living cells exhibit a complex mechanical behavior, whose underlying mechanisms are still largely unknown. Emerging from the molecular structure and dynamics of the cytoskeleton, the mechanical behavior comprises "passive" viscoelastic material properties and "active" contractile prestress. To directly investigate the connection between these quantities at the single-cell level, we here present the combination of atomic force microscopy (AFM) with traction force microscopy (TFM). With this combination, we simultaneously measure viscoelastic material parameters (stiffness, fluidity) and contractile prestress of adherent fibroblast and epithelial cells. Although stiffness, fluidity, and contractile prestress greatly vary within a cell population, they are highly correlated: stiffer cells have a lower fluidity and a larger prestress than softer cells. We show that viscoelastic material properties and contractile prestress are both governed by the activity of the actomyosin machinery. Our results underline the connection between a cell's viscoelastic material properties and its contractile prestress and their importance in cell mechanics.
Collapse
Affiliation(s)
- Nicolas Schierbaum
- Institute of Applied Physics, University of Tübingen, Auf der Morgenstelle 10, 72076 Tübingen, Germany.
| | | | | |
Collapse
|
141
|
McEvoy E, Deshpande VS, McGarry P. Transient active force generation and stress fibre remodelling in cells under cyclic loading. Biomech Model Mechanobiol 2019; 18:921-937. [PMID: 30783833 DOI: 10.1007/s10237-019-01121-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/21/2019] [Indexed: 12/27/2022]
Abstract
The active cytoskeleton is known to play an important mechanistic role in cellular structure, spreading, and contractility. Contractility is actively generated by stress fibres (SF), which continuously remodel in response to physiological dynamic loading conditions. The influence of actin-myosin cross-bridge cycling on SF remodelling under dynamic loading conditions has not previously been uncovered. In this study, a novel SF cross-bridge cycling model is developed to predict transient active force generation in cells subjected to dynamic loading. Rates of formation of cross-bridges within SFs are governed by the chemical potentials of attached and unattached myosin heads. This transient cross-bridge cycling model is coupled with a thermodynamically motivated framework for SF remodelling to analyse the influence of transient force generation on cytoskeletal evolution. A 1D implementation of the model is shown to correctly predict complex patterns of active cell force generation under a range of dynamic loading conditions, as reported in previous experimental studies.
Collapse
Affiliation(s)
- Eoin McEvoy
- Discipline of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | | | - Patrick McGarry
- Discipline of Biomedical Engineering, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
142
|
Meiring JCM, Bryce NS, Cagigas ML, Benda A, Whan RM, Ariotti N, Parton RG, Stear JH, Hardeman EC, Gunning PW. Colocation of Tpm3.1 and myosin IIa heads defines a discrete subdomain in stress fibres. J Cell Sci 2019; 132:jcs.228916. [DOI: 10.1242/jcs.228916] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/06/2019] [Indexed: 01/06/2023] Open
Abstract
Co-polymers of tropomyosin and actin make up a major fraction of the actin cytoskeleton. Tropomyosin isoforms determine the function of an actin filament by selectively enhancing or inhibiting the association of other actin binding proteins, altering the stability of an actin filament and regulating myosin activity in an isoform specific manner. Previous work has implicated specific roles for at least 5 different tropomyosin isoforms in stress fibres, as depletion of any of these 5 isoforms results in a loss of stress fibres. Despite this, most models of stress fibres continue to exclude tropomyosins. In this study we investigate tropomyosin organisation in stress fibres using super resolution light microscopy and electron microscopy with genetically tagged, endogenous tropomyosin. We show that tropomyosin isoforms are organised in subdomains within the overall domain of stress fibres. Tpm3.1/3.2 co-localises with non-muscle myosin IIa/IIb heads and are in register but do not overlap with non-muscle myosin IIa/IIb tails. Furthermore, perturbation of Tpm3.1/3.2 results in decreased myosin IIa in stress fibres, which is consistent with a role for Tpm3.1 in maintaining myosin IIa localisation in stress fibres.
Collapse
Affiliation(s)
- Joyce C. M. Meiring
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nicole S. Bryce
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maria Lastra Cagigas
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Aleš Benda
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Renee M. Whan
- Biomedical Imaging Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nicholas Ariotti
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Robert G. Parton
- Cell Biology and Molecular Medicine Division, Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD 4072, Australia
- Centre for Microscopy and Microanalysis, University of Queensland, St Lucia, QLD 4072, Australia
| | - Jeffrey H. Stear
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Edna C. Hardeman
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter W. Gunning
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
143
|
Cao Y, Lei Y, Luo Y, Tan T, Du B, Zheng Y, Sun L, Liang Q. The actomyosin network is influenced by NMHC IIA and regulated by Crp F46, which is involved in controlling cell migration. Exp Cell Res 2018; 373:119-131. [PMID: 30336116 DOI: 10.1016/j.yexcr.2018.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 10/28/2022]
Abstract
When a cell migrates, the centrosome positions between the nucleus and the leading edge of migration via the microtubule system. The protein CrpF46 (centrosome-related protein F46) has a known role during mitosis and centrosome duplication. However, how CrpF46 efficiently regulates centrosome-related cell migration is unclear. Here, we report that knockdown of CrpF46 resulted in the disruption of microtubule arrangement, with impaired centrosomal reorientation, and slowed down cell migration. In cells that express low levels of CrpF46, stress fibers were weakened, which could be rescued by recovering Flag-CrpF46. We also found that CrpF46 interacted with non-muscle myosin high chain IIA (NMHC IIA) and that its three coiled-coil domains are pivotal for its binding to NMHC IIA. Additionally, analyses of phosphorylation of NMHC IIA and RLC (regulatory light chain) demonstrated that CrpF46 was associated with myosin IIA during filament formation. Indirect immunofluorescence images indicated that NM IIA filaments were inhibited when CrpF46 was under-expressed. Thus, CrpF46 regulates cell migration by centrosomal reorientation and altering the function of the actomyosin network by controlling specific phosphorylation of myosin.
Collapse
Affiliation(s)
- Yang Cao
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Yan Lei
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Yang Luo
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Tan Tan
- School of Pharmacology and Biology, University of South China, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, Hengyang 421001, PR China
| | - Baochen Du
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Yanbo Zheng
- The Institute of Medical Biotechnology (IMB) of the Chinese Academy of Medical Sciences, Beijing 100050, PR China
| | - Le Sun
- AbMax Biotechnology Co., Beijing 101111, PR China
| | - Qianjin Liang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China.
| |
Collapse
|
144
|
Schmitt F, Gold M, Rothemund M, Andronache I, Biersack B, Schobert R, Mueller T. New naphthopyran analogues of LY290181 as potential tumor vascular-disrupting agents. Eur J Med Chem 2018; 163:160-168. [PMID: 30503940 DOI: 10.1016/j.ejmech.2018.11.055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 10/04/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
A series of 19 analogues of the antiproliferative naphthopyran LY290181 were prepared for structure-activity relationship studies. We found the best activities for test compounds bearing small substituents at the meta position of the phenyl ring. The mode of action of LY290181 and eight new analogues was studied in detail. The compounds were highly anti-proliferative with IC50 values in the sub-nanomolar to triple-digit nanomolar range. The new analogues led to G2/M arrest due to interruption of the microtubule dynamics. In 518A2 melanoma cells they caused a mitotic catastrophe which eventually led to apoptosis. The naphthopyrans also induced a disruption of the vasculature in the chorioallantoic membrane (CAM) of fertilized chicken eggs as well as in xenograft tumors in mice. In a preliminary therapy trial, the difluoro derivative 2b retarded the growth of resistant xenograft tumors in mice.
Collapse
Affiliation(s)
- Florian Schmitt
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Madeleine Gold
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Matthias Rothemund
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Ion Andronache
- University of Bucharest, Research Center for Integrated Analysis and Territorial Management, 4-12, Regina Elisabeta Avenue, Bucharest, 3rd District, 030018, Romania
| | - Bernhard Biersack
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany
| | - Rainer Schobert
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440, Bayreuth, Germany.
| | - Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, 06120, Halle, Germany
| |
Collapse
|
145
|
Chen YA, Lu IL, Tsai JW. Contactin-1/F3 Regulates Neuronal Migration and Morphogenesis Through Modulating RhoA Activity. Front Mol Neurosci 2018; 11:422. [PMID: 30515076 PMCID: PMC6255823 DOI: 10.3389/fnmol.2018.00422] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/30/2018] [Indexed: 01/06/2023] Open
Abstract
During neocortical development, newborn neurons migrate along radial fibers from the germinal ventricular zone (VZ) toward the cortical plate (CP) to populate the cerebral cortex. This radial migration requires adhesion activities between neurons and radial fibers; however, past research has identified only a limited number of adhesion molecules involved in this process. Contactin-1/F3 (Cntn1), a cell adhesion molecule expressed in the developing nervous system is essential for many key developmental events including neural cell adhesion, neurite outgrowth, axon guidance and myelination. However, the potential role of Cntn1 in neuronal migration during cortical development has not been investigated. Here we used in utero electroporation to introduce short hairpin RNA (shRNA) to knock down (KD) Cntn1 in neural stem cells in vivo. We found that Cntn1 KD led to a delay in neuronal migration. The arrested cells presented abnormal morphology in their leading process and more multipolar cells were observed in the deep layers of the brain, suggestive of dysregulation in process formation. Intriguingly, Cntn1 KD also resulted in upregulation of RhoA, a negative regulator for neuronal migration. Interference of RhoA by expression of the dominant-negative RhoAN19 partially rescued the neuronal migration defects caused by Cntn1 KD. Our results showed that Cntn1 is a novel adhesion protein that is essential for neuronal migration and regulates process formation of newborn cortical neurons through modulating RhoA signaling pathway.
Collapse
Affiliation(s)
- Yi-An Chen
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - I-Ling Lu
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Jin-Wu Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan.,Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
146
|
Schaks M, Singh SP, Kage F, Thomason P, Klünemann T, Steffen A, Blankenfeldt W, Stradal TE, Insall RH, Rottner K. Distinct Interaction Sites of Rac GTPase with WAVE Regulatory Complex Have Non-redundant Functions in Vivo. Curr Biol 2018; 28:3674-3684.e6. [PMID: 30393033 PMCID: PMC6264382 DOI: 10.1016/j.cub.2018.10.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/30/2018] [Accepted: 10/01/2018] [Indexed: 12/31/2022]
Abstract
Cell migration often involves the formation of sheet-like lamellipodia generated by branched actin filaments. The branches are initiated when Arp2/3 complex [1] is activated by WAVE regulatory complex (WRC) downstream of small GTPases of the Rac family [2]. Recent structural studies defined two independent Rac binding sites on WRC within the Sra-1/PIR121 subunit of the pentameric WRC [3, 4], but the functions of these sites in vivo have remained unknown. Here we dissect the mechanism of WRC activation and the in vivo relevance of distinct Rac binding sites on Sra-1, using CRISPR/Cas9-mediated gene disruption of Sra-1 and its paralog PIR121 in murine B16-F1 cells combined with Sra-1 mutant rescue. We show that the A site, positioned adjacent to the binding region of WAVE-WCA mediating actin and Arp2/3 complex binding, is the main site for allosteric activation of WRC. In contrast, the D site toward the C terminus is dispensable for WRC activation but required for optimal lamellipodium morphology and function. These results were confirmed in evolutionarily distant Dictyostelium cells. Moreover, the phenotype seen in D site mutants was recapitulated in Rac1 E31 and F37 mutants; we conclude these residues are important for Rac-D site interaction. Finally, constitutively activated WRC was able to induce lamellipodia even after both Rac interaction sites were lost, showing that Rac interaction is not essential for membrane recruitment. Our data establish that physical interaction with Rac is required for WRC activation, in particular through the A site, but is not mandatory for WRC accumulation in the lamellipodium.
Collapse
Affiliation(s)
- Matthias Schaks
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany; Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Shashi P Singh
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK; University of Glasgow Institute of Cancer Sciences, Switchback Road, Glasgow G61 1BD, UK
| | - Frieda Kage
- Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Peter Thomason
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK; University of Glasgow Institute of Cancer Sciences, Switchback Road, Glasgow G61 1BD, UK
| | - Thomas Klünemann
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Anika Steffen
- Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Wulf Blankenfeldt
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Theresia E Stradal
- Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Robert H Insall
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK; University of Glasgow Institute of Cancer Sciences, Switchback Road, Glasgow G61 1BD, UK.
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany; Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.
| |
Collapse
|
147
|
Parreno J, Fowler VM. Multifunctional roles of tropomodulin-3 in regulating actin dynamics. Biophys Rev 2018; 10:1605-1615. [PMID: 30430457 DOI: 10.1007/s12551-018-0481-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022] Open
Abstract
Tropomodulins (Tmods) are proteins that cap the slow-growing (pointed) ends of actin filaments (F-actin). The basis for our current understanding of Tmod function comes from studies in cells with relatively stable and highly organized F-actin networks, leading to the view that Tmod capping functions principally to preserve F-actin stability. However, not only is Tmod capping dynamic, but it also can play major roles in regulating diverse cellular processes involving F-actin remodeling. Here, we highlight the multifunctional roles of Tmod with a focus on Tmod3. Like other Tmods, Tmod3 binds tropomyosin (Tpm) and actin, capping pure F-actin at submicromolar and Tpm-coated F-actin at nanomolar concentrations. Unlike other Tmods, Tmod3 can also bind actin monomers and its ability to bind actin is inhibited by phosphorylation of Tmod3 by Akt2. Tmod3 is ubiquitously expressed and is present in a diverse array of cytoskeletal structures, including contractile structures such as sarcomere-like units of actomyosin stress fibers and in the F-actin network encompassing adherens junctions. Tmod3 participates in F-actin network remodeling in lamellipodia during cell migration and in the assembly of specialized F-actin networks during exocytosis. Furthermore, Tmod3 is required for development, regulating F-actin mesh formation during meiosis I of mouse oocytes, erythroblast enucleation in definitive erythropoiesis, and megakaryocyte morphogenesis in the mouse fetal liver. Thus, Tmod3 plays vital roles in dynamic and stable F-actin networks in cell physiology and development, with further research required to delineate the mechanistic details of Tmod3 regulation in the aforementioned processes, or in other yet to be discovered processes.
Collapse
Affiliation(s)
- Justin Parreno
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Velia M Fowler
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
148
|
Pomp W, Schakenraad K, Balcıoğlu HE, van Hoorn H, Danen EHJ, Merks RMH, Schmidt T, Giomi L. Cytoskeletal Anisotropy Controls Geometry and Forces of Adherent Cells. PHYSICAL REVIEW LETTERS 2018; 121:178101. [PMID: 30411958 DOI: 10.1103/physrevlett.121.178101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/06/2018] [Indexed: 06/08/2023]
Abstract
We investigate the geometrical and mechanical properties of adherent cells characterized by a highly anisotropic actin cytoskeleton. Using a combination of theoretical work and experiments on micropillar arrays, we demonstrate that the shape of the cell edge is accurately described by elliptical arcs, whose eccentricity expresses the degree of anisotropy of the internal cell stresses. This results in a spatially varying tension along the cell edge, that significantly affects the traction forces exerted by the cell on the substrate. Our work highlights the strong interplay between cell mechanics and geometry and paves the way towards the reconstruction of cellular forces from geometrical data.
Collapse
Affiliation(s)
- Wim Pomp
- Kamerlingh Onnes-Huygens Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA, Leiden, Netherlands
| | - Koen Schakenraad
- Instituut-Lorentz, Leiden University, P.O. Box 9506, 2300 RA Leiden, Netherlands
- Mathematical Institute, Leiden University, P.O. Box 9512, 2300 RA Leiden, Netherlands
| | - Hayri E Balcıoğlu
- Toxicology, Leiden Academic Center for Drug Research, Leiden University, Netherlands
| | - Hedde van Hoorn
- Kamerlingh Onnes-Huygens Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA, Leiden, Netherlands
| | - Erik H J Danen
- Toxicology, Leiden Academic Center for Drug Research, Leiden University, Netherlands
| | - Roeland M H Merks
- Mathematical Institute, Leiden University, P.O. Box 9512, 2300 RA Leiden, Netherlands
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA Leiden, Netherlands
| | - Thomas Schmidt
- Kamerlingh Onnes-Huygens Laboratory, Leiden University, Niels Bohrweg 2, 2333 CA, Leiden, Netherlands
| | - Luca Giomi
- Instituut-Lorentz, Leiden University, P.O. Box 9506, 2300 RA Leiden, Netherlands
| |
Collapse
|
149
|
Golan M, Pribyl J, Pesl M, Jelinkova S, Acimovic I, Jaros J, Rotrekl V, Falk M, Sefc L, Skladal P, Kratochvilova I. Cryopreserved Cells Regeneration Monitored by Atomic Force Microscopy and Correlated With State of Cytoskeleton and Nuclear Membrane. IEEE Trans Nanobioscience 2018; 17:485-497. [PMID: 30307873 DOI: 10.1109/tnb.2018.2873425] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Atomic force microscopy (AFM) helps to describe and explain the mechanobiological properties of living cells on the nanoscale level under physiological conditions. The stiffness of cells is an important parameter reflecting cell physiology. Here, we have provided the first study of the stiffness of cryopreserved cells during post-thawing regeneration using AFM combined with confocal fluorescence microscopy. We demonstrated that the nonfrozen cell stiffness decreased proportionally to the cryoprotectant concentration in the medium. AFM allowed us to map cell surface reconstitution in real time after a freeze/thaw cycle and to monitor the regeneration processes at different depths of the cell and even different parts of the cell surface (nucleus and edge). Fluorescence microscopy showed that the cytoskeleton in fibroblasts, though damaged by the freeze/thaw cycle, is reconstructed after long-term plating. Confocal microscopy confirmed that structural changes affect the nuclear envelopes in cryopreserved cells. AFM nanoindentation analysis could be used as a noninvasive method to identify cells that have regenerated their surface mechanical properties with the proper dynamics and to a sufficient degree. This identification can be important particularly in the field of in vitro fertilization and in future cell-based regeneration strategies.
Collapse
|
150
|
Endothelial Protrusions in Junctional Integrity and Barrier Function. CURRENT TOPICS IN MEMBRANES 2018; 82:93-140. [PMID: 30360784 DOI: 10.1016/bs.ctm.2018.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endothelial cells of the microcirculation form a semi-permeable diffusion barrier between the blood and tissues. This permeability of the endothelium, particularly in the capillaries and postcapillary venules, is a normal physiological function needed for blood-tissue exchange in the microcirculation. During inflammation, microvascular permeability increases dramatically and can lead to tissue edema, which in turn can lead to dysfunction of tissues and organs. The molecular mechanisms that control the barrier function of endothelial cells have been under investigation for several decades and remain an important topic due to the potential for discovery of novel therapeutic strategies to reduce edema. This review highlights current knowledge of the cellular and molecular mechanisms that lead to endothelial hyperpermeability during inflammatory conditions associated with injury and disease. This includes a discussion of recent findings demonstrating temporal protrusions by endothelial cells that may contribute to intercellular junction integrity between endothelial cells and affect the diffusion distance for solutes via the paracellular pathway.
Collapse
|