1
|
Li Z, Kang D, Xu S, Xi G, Li L, Zheng L, Guo W, Fu F, Wang C, Ma J, Han X, Xu S, Chen J, Chen J. Collagen signature adds prognostically significant information to staging for breast cancer. ESMO Open 2024; 9:103990. [PMID: 39577107 PMCID: PMC11616558 DOI: 10.1016/j.esmoop.2024.103990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Tumor-associated collagen signature (TACS) is an independent prognostic factor for breast cancer. However, it is unclear whether the complete collagen signature, including TACS, the TACS-based collagen microscopic features (TCMF1), and the TACS-based nuclear features (TCMF2), can provide additional prognostic information for the current tumor-node-metastasis (TNM) staging system. PATIENTS AND METHODS We included 941 patients with breast cancer from three cohorts: the training (n = 355), internal (n = 334), and external validation cohorts (n = 252). TACS and TCMF1 were obtained by multiphoton microscopy (MPM). TCMF2 was extracted on the hematoxylin and eosin images colocated with MPM images. They were linearly combined to establish a complete collagen signature score for reclassifying current TNM staging into stage Ⅰ (II and Ⅲ)/low risk and stage Ⅰ (II and Ⅲ)/high risk. RESULTS The low-risk collagen signatures 'downstaged' patients in stage II or Ⅲ, while the high-risk collagen signatures 'upstaged' patients with stage Ⅰ tumors. After incorporating the complete collagen signature into the current TNM staging system, the modified staging system had a higher ability to stratify patients [referent, Ⅰ-new; Ⅱ-new, hazard ratio (HR) 8.655, 6.136, and 4.699 in the training, internal validation, and external validation cohorts, respectively; Ⅲ-new, HR 14.855, 11.201, and 13.245 in the corresponding three cohorts, respectively] than the current TNM staging system (referent, Ⅰ; Ⅱ, HR 1.642, 1.853, and 1.371 in the corresponding three cohorts, respectively; Ⅲ, HR 4.131, 4.283, and 3.711 in the corresponding three cohorts, respectively). Furthermore, the modified staging system showed a higher area under the curve than the current TNM staging system (training cohort: 0.843 versus 0.683; internal validation cohort: 0.792 versus 0.661; and external validation cohort: 0.793 versus 0.646). CONCLUSIONS The complete collagen signature is an independent predictor of survival outcomes in breast cancer. It adds significant information about the biological behavior of the disease to staging for breast cancer.
Collapse
Affiliation(s)
- Z Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - D Kang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - S Xu
- Bio-totem Pte Ltd, Foshan, China
| | - G Xi
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - L Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - L Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - W Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - F Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - C Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - J Ma
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - X Han
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - S Xu
- School of Electronic and Mechanical Engineering, Fujian Polytechnic Normal University, Fuqing, Fujian, China
| | - J Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China; College of Life Science, Fujian Normal University, Fuzhou, China.
| | - J Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
2
|
Zahn LA, Lundin-Schiller S. Evidence for microtubule nucleation at the Golgi in breast cancer cells. Cytoskeleton (Hoboken) 2024; 81:193-205. [PMID: 37905740 DOI: 10.1002/cm.21803] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023]
Abstract
Golgi-derived microtubule (MT) arrays are essential to directionally persistent cell migration and vesicle transport. In this study, we have examined MT nucleation sites in two breast cancer cell lines, MDA-MB-231 and MCF-7, with the hypothesis that only the migratory invasive MDA-MB-231 cells exhibit MTs originating from the Golgi. MTs were disassembled and allowed to slightly regrow so individual nucleation sites could then be observed via fluorescently tagged antibodies (α-tubulin, cis-Golgi marker GM130, and EB1-a MT plus-end binding protein) and confocal microscopy. To determine if MT nucleation at the Golgi is more apparent during active migration compared to when cells are stationary, cells were treated with the chemoattractant epidermal growth factor (EGF) and examined for colocalizations between the Golgi, α-tubulin, and γ-tubulin. Images were analyzed qualitatively for color overlap, and quantitatively using Manders Colocalization Coefficients. Differences between groups were tested for significance using one-way analysis of variances and Tukey's post hoc test. Significantly higher colocalization values (coloc) in the highly invasive MDA-MB-231 cells (α-tubulin coloc GM130 = 0.39, GM130 coloc α-tubulin = 0.82, GM130 coloc EB1 = 0.24, and EB1 coloc GM130 = 0.38) compared to the weakly invasive MCF-7 cells (0.15, 0.08, 0.02, and 0.16, respectively) were observed. EGF-treated cells exhibited higher colocalization values than control cells for three of the four protein combinations tested, but EGF-treated MDA-MB-231 cells exhibited significantly higher values (α-tubulin coloc GM130 = 0.20, GM130 coloc α-tubulin = 0.89, and γ-tubulin coloc GM130 = 0.47) than both control groups as well as the EGF-treated MCF-7 cells. Results support the hypothesis that MT nucleation at the Golgi occurs more frequently in the invasive MDA-MB-231 cell line compared to the weakly invasive MCF-7 cells. The presence or absence of Golgi-derived MTs may help to explain the difference in migratory potential commonly exhibited by these two cell lines.
Collapse
Affiliation(s)
- Laura A Zahn
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
3
|
Singh P, Ramanathan V, Zhang Y, Georgakoudi I, Jay DG. Extracellular Hsp90 Binds to and Aligns Collagen-1 to Enhance Breast Cancer Cell Invasiveness. Cancers (Basel) 2023; 15:5237. [PMID: 37958410 PMCID: PMC10648158 DOI: 10.3390/cancers15215237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/09/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer cell-secreted eHsp90 binds and activates proteins in the tumor microenvironment crucial in cancer invasion. Therefore, targeting eHsp90 could inhibit invasion, preventing metastasis-the leading cause of cancer-related mortality. Previous eHsp90 studies have solely focused on its role in cancer invasion through the 2D basement membrane (BM), a form of extracellular matrix (ECM) that lines the epithelial compartment. However, its role in cancer invasion through the 3D Interstitial Matrix (IM), an ECM beyond the BM, remains unexplored. Using a Collagen-1 binding assay and second harmonic generation (SHG) imaging, we demonstrate that eHsp90 directly binds and aligns Collagen-1 fibers, the primary component of IM. Furthermore, we show that eHsp90 enhances Collagen-1 invasion of breast cancer cells in the Transwell assay. Using Hsp90 conformation mutants and inhibitors, we established that the Hsp90 dimer binds to Collagen-1 via its N-domain. We also demonstrated that while Collagen-1 binding and alignment are not influenced by Hsp90's ATPase activity attributed to the N-domain, its open conformation is crucial for increasing Collagen-1 alignment and promoting breast cancer cell invasion. These findings unveil a novel role for eHsp90 in invasion through the IM and offer valuable mechanistic insights into potential therapeutic approaches for inhibiting Hsp90 to suppress invasion and metastasis.
Collapse
Affiliation(s)
- Pragya Singh
- Department of Developmental, Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; (P.S.); (I.G.)
| | - Varshini Ramanathan
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA 02155, USA; (V.R.); (Y.Z.)
| | - Yang Zhang
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA 02155, USA; (V.R.); (Y.Z.)
| | - Irene Georgakoudi
- Department of Developmental, Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; (P.S.); (I.G.)
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA 02155, USA; (V.R.); (Y.Z.)
| | - Daniel G. Jay
- Department of Developmental, Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; (P.S.); (I.G.)
| |
Collapse
|
4
|
Banerjee M, Devi Rajeswari V. A novel cross-communication of HIF-1α and HIF-2α with Wnt signaling in TNBC and influence of hypoxic microenvironment in the formation of an organ-on-chip model of breast cancer. Med Oncol 2023; 40:245. [PMID: 37454033 DOI: 10.1007/s12032-023-02112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
The microenvironment role is very important in cancer development. The epithelial-mesenchymal transition of the cancer cells depends upon specific signaling and microenvironmental conditions, such as hypoxic conditions. The crosstalk between hypoxia and Wnt signaling through some molecular mechanism in TNBC is related. Cross-communication between hypoxia and Wnt signaling in cancer cells is known, but the detailed mechanism in TNBC is unknown. This review includes the role of the hypoxia microenvironment in TNBC and the novel crosstalk of the Wnt signaling and hypoxia. When targeted, the new pathway and crosstalk link may be a solution for metastatic TNBC and chemoresistance. The microenvironment influences cancer's metastasis, which changes from person to person. Therefore, organ-on-a-chip is a very novel model to test the drugs clinically before going for human trials, focusing on personalized medications can be done. The effect of the hypoxia microenvironment on breast cancer stem cells is still unknown. Apart from all the published papers, this paper mainly focuses only on the hypoxic microenvironment and its association with the growth of TNBC. The medicines or small proteins, drugs, mimics, and inhibitors targeting wnt and hypoxia genes are consolidated in this review paper.
Collapse
Affiliation(s)
- Manosi Banerjee
- Department of Biomedical Science, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - V Devi Rajeswari
- Department of Biomedical Science, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
5
|
Ebbinghaus T, Lang G, Scheibel T. Biomimetic polymer fibers-function by design. BIOINSPIRATION & BIOMIMETICS 2023; 18:041003. [PMID: 37307815 DOI: 10.1088/1748-3190/acddc1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/12/2023] [Indexed: 06/14/2023]
Abstract
Biomimicry applies the fundamental principles of natural materials, processes, and structures to technological applications. This review presents the two strategies of biomimicry-bottom-up and top-down approaches, using biomimetic polymer fibers and suitable spinning techniques as examples. The bottom-up biomimicry approach helps to acquire fundamental knowledge on biological systems, which can then be leveraged for technological advancements. Within this context, we discuss the spinning of silk and collagen fibers due to their unique natural mechanical properties. To achieve successful biomimicry, it is imperative to carefully adjust the spinning solution and processing parameters. On the other hand, top-down biomimicry aims to solve technological problems by seeking solutions from natural role models. This approach will be illustrated using examples such as spider webs, animal hair, and tissue structures. To contextualize biomimicking approaches in practical applications, this review will give an overview of biomimetic filter technologies, textiles, and tissue engineering.
Collapse
Affiliation(s)
- Thomas Ebbinghaus
- Chair of Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Str. 1, 95447 Bayreuth, Germany
| | - Gregor Lang
- Department of Functional Materials in Medicine and Dentistry, University Hospital of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
| | - Thomas Scheibel
- Chair of Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Str. 1, 95447 Bayreuth, Germany
- Bayreuth Center for Colloids and Interfaces (BZKG), University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
- Bavarian Polymer Institute (BPI), University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
- Bayreuth Center for Molecular Biosciences (BZMB), University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
- Bayreuth Center for Material Science (BayMAT), University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| |
Collapse
|
6
|
Narciso M, Martínez Á, Júnior C, Díaz-Valdivia N, Ulldemolins A, Berardi M, Neal K, Navajas D, Farré R, Alcaraz J, Almendros I, Gavara N. Lung Micrometastases Display ECM Depletion and Softening While Macrometastases Are 30-Fold Stiffer and Enriched in Fibronectin. Cancers (Basel) 2023; 15:cancers15082404. [PMID: 37190331 DOI: 10.3390/cancers15082404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Mechanical changes in tumors have long been linked to increased malignancy and therapy resistance and attributed to mechanical changes in the tumor extracellular matrix (ECM). However, to the best of our knowledge, there have been no mechanical studies on decellularized tumors. Here, we studied the biochemical and mechanical progression of the tumor ECM in two models of lung metastases: lung carcinoma (CAR) and melanoma (MEL). We decellularized the metastatic lung sections, measured the micromechanics of the tumor ECM, and stained the sections for ECM proteins, proliferation, and cell death markers. The same methodology was applied to MEL mice treated with the clinically approved anti-fibrotic drug nintedanib. When compared to healthy ECM (~0.40 kPa), CAR and MEL lung macrometastases produced a highly dense and stiff ECM (1.79 ± 1.32 kPa, CAR and 6.39 ± 3.37 kPa, MEL). Fibronectin was overexpressed from the early stages (~118%) to developed macrometastases (~260%) in both models. Surprisingly, nintedanib caused a 4-fold increase in ECM-occupied tumor area (5.1 ± 1.6% to 18.6 ± 8.9%) and a 2-fold in-crease in ECM stiffness (6.39 ± 3.37 kPa to 12.35 ± 5.74 kPa). This increase in stiffness strongly correlated with an increase in necrosis, which reveals a potential link between tumor hypoxia and ECM deposition and stiffness. Our findings highlight fibronectin and tumor ECM mechanics as attractive targets in cancer therapy and support the need to identify new anti-fibrotic drugs to abrogate aberrant ECM mechanics in metastases.
Collapse
Affiliation(s)
- Maria Narciso
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - África Martínez
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Constança Júnior
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Natalia Díaz-Valdivia
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Anna Ulldemolins
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, De Boelelaan 1081, 1081 HV Amsterdam, The Netherlands
- Optics11, Hettenheuvelweg 37-39, 1101 BM Amsterdam, The Netherlands
| | - Kate Neal
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Daniel Navajas
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 08036 Madrid, Spain
| | - Ramon Farré
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 08036 Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Thoracic Oncology Unit, Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Isaac Almendros
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), 08036 Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Núria Gavara
- Unit of Biophysics and Bioengineering, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| |
Collapse
|
7
|
Rafnsdóttir ÓB, Kiuru A, Tebäck M, Friberg N, Revstedt P, Zhu J, Thomasson S, Czopek A, Malakpour-Permlid A, Weber T, Oredsson S. A new animal product free defined medium for 2D and 3D culturing of normal and cancer cells to study cell proliferation and migration as well as dose response to chemical treatment. Toxicol Rep 2023; 10:509-520. [PMID: 37396848 PMCID: PMC10313884 DOI: 10.1016/j.toxrep.2023.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/23/2023] [Accepted: 04/05/2023] [Indexed: 07/04/2023] Open
Abstract
Cell culturing methods are increasingly used to reduce and replace the use of live animals in biomedical research and chemical toxicity testing. Although live animals are avoided when using cell culturing methods, they often contain animal-derived components of which one of the most commonly used is foetal bovine serum (FBS). FBS is added to cell culture media among other supplements to support cell attachment/spreading and cell proliferation. The safety, batch-to-batch variation, and ethical problems with FBS are acknowledged and therefore world-wide efforts are ongoing to produce FBS free media. Here, we present the composition of a new defined medium with only human proteins either recombinant or derived from human tissues. This defined medium supports long-term culturing/routine culturing of normal cells and of cancer cells, and can be used for freezing and thawing of cells, i.e. for cell banking. Here, we show for our defined medium, growth curves and dose response curves of cells grown in two and three dimensions, and applications such as cell migration. Cell morphology was studied in real time by phase contrast and phase holographic microscopy time-lapse imaging. The cell lines used are human cancer-associated fibroblasts, keratinocytes, breast cancer JIMT-1 and MDA-MB-231 cells, colon cancer CaCo-2 cells, and pancreatic cancer MiaPaCa-2 cells as well as the mouse L929 cell line. In conclusion, we present the composition of a defined medium without animal-derived products which can be used for routine culturing and in experimental settings for normal cells and for cancer cells, i.e. our defined medium provides a leap towards a universal animal product free cell culture medium.
Collapse
Affiliation(s)
- Ólöf Birna Rafnsdóttir
- Department of Biology, Lund University, 22362 Lund, Sweden
- Institute of Life and Environmental Sciences, School of Engineering and Natural Sciences, University of Iceland, 101 Reykjavík, Iceland
| | - Anna Kiuru
- Department of Biology, Lund University, 22362 Lund, Sweden
- Occupational and Environmental Dermatology, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Mattis Tebäck
- Department of Biology, Lund University, 22362 Lund, Sweden
| | | | | | - Johan Zhu
- Department of Biology, Lund University, 22362 Lund, Sweden
- Clinical Microbiology and Infection Prevention and Control, Region Skåne, 221 85 Lund, Sweden
| | - Sofia Thomasson
- Department of Biology, Lund University, 22362 Lund, Sweden
- Atos Medical AB, 242 35 Hörby, Sweden
| | | | - Atena Malakpour-Permlid
- Department of Biology, Lund University, 22362 Lund, Sweden
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics, Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Tilo Weber
- Animal Welfare Academy of the German Animal Welfare Federation, 85579 Neubiberg, Germany
| | - Stina Oredsson
- Department of Biology, Lund University, 22362 Lund, Sweden
| |
Collapse
|
8
|
Hashimoto N, Kitai R, Fujita S, Yamauchi T, Isozaki M, Kikuta KI. Single-Cell Analysis of Unidirectional Migration of Glioblastoma Cells Using a Fiber-Based Scaffold. ACS APPLIED BIO MATERIALS 2023; 6:765-773. [PMID: 36758146 PMCID: PMC9945112 DOI: 10.1021/acsabm.2c00958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Glioblastoma (GBM) is a malignant incurable brain tumor in which immature neoplastic cells infiltrate brain tissue by spreading along nerve fibers. The aim of the study was to compare the migration abilities of glioma cells with those of other cancer cells and elucidate the migratory profiles underlying the differential migration of glioma cells using a fiber-based quantitative migration assay. Here, wound healing and transwell assays were used to assess cell mobility in four cell lines: U87-MG glioblastoma cells, MDA-MB-231 breast cancer cells, HCT116 colorectal cancer cells, and MKN45 gastric cancer cells. We also assessed cell mobility using a fiber model that mimics nerve fibers. Time-lapse video microscopy was used to observe cell migration and morphology. The cytoskeleton arrangement was assessed in the fiber model and compared with that in the conventional cell culture model. The conventional evaluation of cell migration ability revealed that the migration ability of breast cancer and glioblastoma cell lines was higher than that of colon cancer and gastric cancer cell lines. The fiber model confirmed that the glioblastoma cell line had a significantly higher migration ability than other cell lines. Tubulin levels were significantly higher in the glioblastoma cells than in other cell lines. In conclusion, the developed fiber-based culture model revealed the specific migratory profile of GBM cells during invasion.
Collapse
Affiliation(s)
- Norichika Hashimoto
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Department of Neurosurgery, Fukui General Hospital, 58-16-1 Egami-cho, Fukui-shi, Fukui 910-8561, Japan
| | - Ryuhei Kitai
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Department of Neurosurgery, Kaga Medical Center, Kaga, Ri 36, Sakumi-machi, Kaga-shi, Ishikawa 922-8522, Japan
| | - Satoshi Fujita
- Department of Frontier Fiber Technology and Science, Graduate School of Engineering, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| | - Takahiro Yamauchi
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| | - Makoto Isozaki
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Ken-Ichiro Kikuta
- Division of Medicine, Department of Neurosurgery, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan.,Organization for Life Science Advancement Programs, University of Fukui, 3-9-1, Bunkyo, Fukui-shi, Fukui 910-8507, Japan
| |
Collapse
|
9
|
Loesel KE, Hiraki HL, Baker BM, Parent CA. An adaptive and versatile method to quantitate and characterize collective cell migration behaviors on complex surfaces. Front Cell Dev Biol 2023; 11:1106653. [PMID: 36776562 PMCID: PMC9909417 DOI: 10.3389/fcell.2023.1106653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
Collective cell migration is critical for proper embryonic development, wound healing, and cancer cell invasion. However, much of our knowledge of cell migration has been performed using flat surfaces that lack topographical features and do not recapitulate the complex fibrous architecture of the extracellular matrix (ECM). The recent availability of synthetic fibrous networks designed to mimic in vivo ECM has been key to identify the topological features that dictate cell migration patterns as well as to determine the underlying mechanisms that regulate topography-sensing. Recent studies have underscored the prevalence of collective cell migration during cancer invasion, and these observations present a compelling need to understand the mechanisms controlling contact guidance within migratory, multicellular groups. Therefore, we designed an integrated migration analysis platform combining tunable electrospun fibers that recapitulate aspects of the biophysical properties of the ECM, and computational approaches to investigate collective cell migration. To quantitatively assess migration as a function of matrix topography, we developed an automated MATLAB code that quantifies cell migration dynamics, including speed, directionality, and the number of detached cells. This platform enables live cell imaging while providing enough cells for biochemical, proteomic, and genomic analyses, making our system highly adaptable to multiple experimental investigations.
Collapse
Affiliation(s)
- Kristen E. Loesel
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Harrison L. Hiraki
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Carole A. Parent
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States,Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States,*Correspondence: Carole A. Parent,
| |
Collapse
|
10
|
Liu YX, Chaparro FJ, Tian Z, Jia Y, Gosser J, Gaumer J, Ross L, Tafreshi H, Lannutti JJ. Visualization of porosity and pore size gradients in electrospun scaffolds using laser metrology. PLoS One 2023; 18:e0282903. [PMID: 36893193 PMCID: PMC9997878 DOI: 10.1371/journal.pone.0282903] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/24/2023] [Indexed: 03/10/2023] Open
Abstract
We applied a recently developed method, laser metrology, to characterize the influence of collector rotation on porosity gradients of electrospun polycaprolactone (PCL) widely investigated for use in tissue engineering. The prior- and post-sintering dimensions of PCL scaffolds were compared to derive quantitative, spatially-resolved porosity 'maps' from net shrinkage. Deposited on a rotating mandrel (200 RPM), the central region of deposition reaches the highest porosity, ~92%, surrounded by approximately symmetrical decreases to ~89% at the edges. At 1100 RPM, a uniform porosity of ~88-89% is observed. At 2000 RPM, the lowest porosity, ~87%, is found in the middle of the deposition, rebounding to ~89% at the edges. Using a statistical model of random fiber network, we demonstrated that these relatively small changes in porosity values produce disproportionately large variations in pore size. The model predicts an exponential dependence of pore size on porosity when the scaffold is highly porous (e.g., >80%) and, accordingly, the observed porosity variation is associated with dramatic changes in pore size and ability to accommodate cell infiltration. Within the thickest regions most likely to 'bottleneck' cell infiltration, pore size decreases from ~37 to 23 μm (38%) when rotational speeds increased from 200 to 2000 RPM. This trend is corroborated by electron microscopy. While faster rotational speeds ultimately overcome axial alignment induced by cylindrical electric fields associated with the collector geometry, it does so at the cost of eliminating larger pores favoring cell infiltration. This puts the bio-mechanical advantages associated with collector rotation-induced alignment at odds with biological goals. A more significant decrease in pore size from ~54 to ~19 μm (65%), well below the minimum associated with cellular infiltration, is observed from enhanced collector biases. Finally, similar predictions show that sacrificial fiber approaches are inefficient in achieving cell-permissive pore sizes.
Collapse
Affiliation(s)
- Yi-xiao Liu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
- * E-mail:
| | | | - Ziting Tian
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Yizhen Jia
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - John Gosser
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Jeremy Gaumer
- Tosoh SMD, Inc., Grove City, OH, United States of America
| | - Liam Ross
- Columbus Academy, Gahanna, OH, United States of America
| | - Hooman Tafreshi
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC, United States of America
| | - John J. Lannutti
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States of America
- Center for Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
11
|
Erickson A, Chiarelli PA, Huang J, Levengood SL, Zhang M. Electrospun nanofibers for 3-D cancer models, diagnostics, and therapy. NANOSCALE HORIZONS 2022; 7:1279-1298. [PMID: 36106417 DOI: 10.1039/d2nh00328g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
As one of the leading causes of global mortality, cancer has prompted extensive research and development to advance efficacious drug discovery, sustained drug delivery and improved sensitivity in diagnosis. Towards these applications, nanofibers synthesized by electrospinning have exhibited great clinical potential as a biomimetic tumor microenvironment model for drug screening, a controllable platform for localized, prolonged drug release for cancer therapy, and a highly sensitive cancer diagnostic tool for capture and isolation of circulating tumor cells in the bloodstream and for detection of cancer-associated biomarkers. This review provides an overview of applied nanofiber design with focus on versatile electrospinning fabrication techniques. The influence of topographical, physical, and biochemical properties on the function of nanofiber assemblies is discussed, as well as current and foreseeable barriers to the clinical translation of applied nanofibers in the field of oncology.
Collapse
Affiliation(s)
- Ariane Erickson
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Peter A Chiarelli
- The Saban Research Institute, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Jianxi Huang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Sheeny Lan Levengood
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
12
|
Collagen Remodeling along Cancer Progression Providing a Novel Opportunity for Cancer Diagnosis and Treatment. Int J Mol Sci 2022; 23:ijms231810509. [PMID: 36142424 PMCID: PMC9502421 DOI: 10.3390/ijms231810509] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a significant factor in cancer progression. Collagens, as the main component of the ECM, are greatly remodeled alongside cancer development. More and more studies have confirmed that collagens changed from a barrier to providing assistance in cancer development. In this course, collagens cause remodeling alongside cancer progression, which in turn, promotes cancer development. The interaction between collagens and tumor cells is complex with biochemical and mechanical signals intervention through activating diverse signal pathways. As the mechanism gradually clears, it becomes a new target to find opportunities to diagnose and treat cancer. In this review, we investigated the process of collagen remodeling in cancer progression and discussed the interaction between collagens and cancer cells. Several typical effects associated with collagens were highlighted in the review, such as fibrillation in precancerous lesions, enhancing ECM stiffness, promoting angiogenesis, and guiding invasion. Then, the values of cancer diagnosis and prognosis were focused on. It is worth noting that several generated fragments in serum were reported to be able to be biomarkers for cancer diagnosis and prognosis, which is beneficial for clinic detection. At a glance, a variety of reported biomarkers were summarized. Many collagen-associated targets and drugs have been reported for cancer treatment in recent years. The new targets and related drugs were discussed in the review. The mass data were collected and classified by mechanism. Overall, the interaction of collagens and tumor cells is complicated, in which the mechanisms are not completely clear. A lot of collagen-associated biomarkers are excavated for cancer diagnosis. However, new therapeutic targets and related drugs are almost in clinical trials, with merely a few in clinical applications. So, more efforts are needed in collagens-associated studies and drug development for cancer research and treatment.
Collapse
|
13
|
Luparello C, Branni R, Abruscato G, Lazzara V, Drahos L, Arizza V, Mauro M, Di Stefano V, Vazzana M. Cytotoxic capability and the associated proteomic profile of cell-free coelomic fluid extracts from the edible sea cucumber Holothuria tubulosa on HepG2 liver cancer cells. EXCLI JOURNAL 2022; 21:722-743. [PMID: 35721581 PMCID: PMC9203982 DOI: 10.17179/excli2022-4825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/13/2022] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive cancer histotype and one of the most common types of cancer worldwide. The identification of compounds that might intervene to restrain neoplastic cell growth appears imperative due to its elevated overall mortality. The marine environment represents a reservoir rich in bioactive compounds in terms of primary and secondary metabolites produced by aquatic animals, mainly invertebrates. In the present study, we determined whether the water-soluble cell-free extract of the coelomic fluid (CFE) of the edible sea cucumber Holothuria tubulosa could play an anti-HCC role in vitro by analyzing the viability and locomotory behavior, cell cycle distribution, apoptosis and autophagy modulation, mitochondrial function and cell redox state of HepG2 HCC cells. We showed that CFE causes an early block in the cell cycle at the G2/M phase, which is coupled to oxidative stress promotion, autophagosome depletion and mitochondrial dysfunction ultimately leading to apoptotic death. We also performed a proteomic analysis of CFE identifying a number of proteins that are seemingly responsible for anti-cancer effects. In conclusion, H. tubulosa's CFE merits further investigation to develop novel promising anti-HCC prevention and/or treatment agents and also beneficial supplements for formulation of functional foods and food packaging material.
Collapse
Affiliation(s)
- Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - Rossella Branni
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - Giulia Abruscato
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - Valentina Lazzara
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - Laszlo Drahos
- MS Proteomics Research Group, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - Manuela Mauro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - Vita Di Stefano
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| | - Mirella Vazzana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Palermo, Italy
| |
Collapse
|
14
|
Sancho A, Taskin MB, Wistlich L, Stahlhut P, Wittmann K, Rossi A, Groll J. Cell Adhesion Assessment Reveals a Higher Force per Contact Area on Fibrous Structures Compared to Flat Substrates. ACS Biomater Sci Eng 2022; 8:649-658. [DOI: 10.1021/acsbiomaterials.1c01290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ana Sancho
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
- Department of Automatic Control and Systems Engineering, University of the Basque Country UPV/EHU, Plaza de Europa 1, 20018 Donostia, Spain
| | - Mehmet Berat Taskin
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
| | - Laura Wistlich
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
| | - Philipp Stahlhut
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
| | - Katharina Wittmann
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
| | - Angela Rossi
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
| |
Collapse
|
15
|
Williams AH, Hebert AM, Boehm RC, Huddleston ME, Jenkins MR, Velev OD, Nelson MT. Bioscaffold Stiffness Mediates Aerosolized Nanoparticle Uptake in Lung Epithelial Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50643-50656. [PMID: 34668373 DOI: 10.1021/acsami.1c09701] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this study, highly porous, ultrasoft polymeric mats mimicking human tissues were formed from novel polyurethane soft dendritic colloids (PU SDCs). PU SDCs have a unique fibrillar morphology controlled by antisolvent precipitation. When filtered from suspension, PU SDCs form mechanically robust nonwoven mats. The stiffness of the SDC mats can be tuned for physiological relevance. The unique physiochemical characteristics of the PU SDC particles dictate the mechanical properties resulting in tunable elastic moduli ranging from 200 to 800 kPa. The human lung A549 cells cultured on both stiff and soft PU SDC membranes were found to be viable, capable of supporting the air-liquid interface (ALI) cell culture, and maintained barrier integrity. Furthermore, A549 cellular viability and uptake efficiency of aerosolized tannic acid-coated gold nanoparticles (Ta-Au) was found to depend on elastic modulus and culture conditions. Ta-Au nanoparticle uptake was twofold and fourfold greater on soft PU SDCs, when cultured at submerged and ALI conditions, respectively. The significant increase in endocytosed Ta-Au resulted in a 20% decrease in viability, and a 4-fold increase in IL-8 cytokine secretion when cultured on soft PU SDCs at ALI. Common tissue culture materials exhibit super-physiological elastic moduli, a factor found to be critical in analyzing nanomaterial cellular interactions and biological responses.
Collapse
Affiliation(s)
- Austin H Williams
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Adrien M Hebert
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - Robert C Boehm
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - Mary E Huddleston
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
- UES, Inc., Dayton, Ohio 45432, United States
| | - Meghan R Jenkins
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
- UES, Inc., Dayton, Ohio 45432, United States
| | - Orlin D Velev
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - M Tyler Nelson
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| |
Collapse
|
16
|
Hsu MY, Hsieh CH, Huang YT, Chu SY, Chen CM, Lee WJ, Liu SJ. Enhanced Paclitaxel Efficacy to Suppress Triple-Negative Breast Cancer Progression Using Metronomic Chemotherapy with a Controlled Release System of Electrospun Poly-d-l-Lactide-Co-Glycolide (PLGA) Nanofibers. Cancers (Basel) 2021; 13:cancers13133350. [PMID: 34283075 PMCID: PMC8268060 DOI: 10.3390/cancers13133350] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Treatment of metastatic triple-negative breast cancer (TNBC) relies on chemotherapy. To improve the efficacy of chemotherapy and avoid systemic toxicity, metronomic chemotherapy using continuous administration of low-dose chemotherapy could be a solution. The paclitaxel-loaded PLGA nanofibers allow for continuous and prolonged drug release, which is compatible with the concept of metronomic chemotherapy. The animal study revealed that the strategy successfully inhibited the growth of the primary tumor and distant metastasis without sarcopenia. These data offer new insights into the role of drug-loaded nanofibers in the treatment of metastatic TNBC. Abstract Triple-negative breast cancer (TNBC) is highly aggressive and responds poorly to conventional chemotherapy. The challenge of TNBC therapy is to maximize the efficacies of conventional chemotherapeutic agents and reduce their toxicities. Metronomic chemotherapy using continuous low-dose chemotherapy has been proposed as a new treatment option, but this approach is limited by the selection of drugs. To improve antitumor therapeutic effects, we developed electrospun paclitaxel-loaded poly-d-l-lactide-co-glycolide (PLGA) nanofibers as a topical implantable delivery device for controlled drug release and site-specific treatment. The subcutaneously implanted paclitaxel-loaded nanofibrous membrane in mice was compatible with the concept of metronomic chemotherapy; it significantly enhanced antitumor activity, inhibited local tumor growth, constrained distant metastasis, and prolonged survival compared with intraperitoneal paclitaxel injection. Furthermore, under paclitaxel-loaded nanofiber treatment, systemic toxicity was low with a persistent increase in lean body weight in mice; in contrast, body weight decreased in other groups. The paclitaxel-loaded nanofibrous membranes provided sustained drug release and site-specific treatment by directly targeting and changing the tumor microenvironment, resulting in low systemic toxicity and a significant improvement in the therapeutic effect and safety compared with conventional chemotherapy. Thus, metronomic chemotherapy with paclitaxel-loaded nanofibrous membranes offers a promising strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Ming-Yi Hsu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Cheng-Hsien Hsieh
- Department of Emergency Medicine, En-Chu-Kong Hospital, New Taipei City 23741, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Ting Huang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Sung-Yu Chu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
| | - Chien-Ming Chen
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
| | - Wei-Jiunn Lee
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11695, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (W.-J.L.); (S.-J.L.); Tel.: +886-2-2930-7930 (ext. 2551/2547) (W.-J.L.); +886-3-2118166 (S.-J.L.)
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- Correspondence: (W.-J.L.); (S.-J.L.); Tel.: +886-2-2930-7930 (ext. 2551/2547) (W.-J.L.); +886-3-2118166 (S.-J.L.)
| |
Collapse
|
17
|
Sharma VP, Williams J, Leung E, Sanders J, Eddy R, Castracane J, Oktay MH, Entenberg D, Condeelis JS. SUN-MKL1 Crosstalk Regulates Nuclear Deformation and Fast Motility of Breast Carcinoma Cells in Fibrillar ECM Microenvironment. Cells 2021; 10:1549. [PMID: 34205257 PMCID: PMC8234170 DOI: 10.3390/cells10061549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
Aligned collagen fibers provide topography for the rapid migration of single tumor cells (streaming migration) to invade the surrounding stroma, move within tumor nests towards blood vessels to intravasate and form distant metastases. Mechanisms of tumor cell motility have been studied extensively in the 2D context, but the mechanistic understanding of rapid single tumor cell motility in the in vivo context is still lacking. Here, we show that streaming tumor cells in vivo use collagen fibers with diameters below 3 µm. Employing 1D migration assays with matching in vivo fiber dimensions, we found a dependence of tumor cell motility on 1D substrate width, with cells moving the fastest and the most persistently on the narrowest 1D fibers (700 nm-2.5 µm). Interestingly, we also observed nuclear deformation in the absence of restricting extracellular matrix pores during high speed carcinoma cell migration in 1D, similar to the nuclear deformation observed in tumor cells in vivo. Further, we found that actomyosin machinery is aligned along the 1D axis and actomyosin contractility synchronously regulates cell motility and nuclear deformation. To further investigate the link between cell speed and nuclear deformation, we focused on the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex proteins and SRF-MKL1 signaling, key regulators of mechanotransduction, actomyosin contractility and actin-based cell motility. Analysis of The Cancer Genome Atlas dataset showed a dramatic decrease in the LINC complex proteins SUN1 and SUN2 in primary tumor compared to the normal tissue. Disruption of LINC complex by SUN1 + 2 KD led to multi-lobular elongated nuclei, increased tumor cell motility and concomitant increase in F-actin, without affecting Lamin proteins. Mechanistically, we found that MKL1, an effector of changes in cellular G-actin to F-actin ratio, is required for increased 1D motility seen in SUN1 + 2 KD cells. Thus, we demonstrate a previously unrecognized crosstalk between SUN proteins and MKL1 transcription factor in modulating nuclear shape and carcinoma cell motility in an in vivo relevant 1D microenvironment.
Collapse
Affiliation(s)
- Ved P. Sharma
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - James Williams
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Edison Leung
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
| | - Joe Sanders
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Robert Eddy
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
| | - James Castracane
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Maja H. Oktay
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Entenberg
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John S. Condeelis
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
18
|
Su CY, Burchett A, Dunworth M, Choi JS, Ewald AJ, Ahn EH, Kim DH. Engineering a 3D collective cancer invasion model with control over collagen fiber alignment. Biomaterials 2021; 275:120922. [PMID: 34126408 DOI: 10.1016/j.biomaterials.2021.120922] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022]
Abstract
Prior to cancer cell invasion, the structure of the extracellular matrix (ECM) surrounding the tumor is remodeled, such that circumferentially oriented matrix fibers become radially aligned. This predisposed radially aligned matrix structure serves as a critical regulator of cancer invasion. However, a biomimetic 3D model recapitulating a tumor's behavioral response to these ECM structures is not yet available. In this study, we have developed a phase-specific, force-guided method to establish a 3D dual topographical tumor model in which each tumor spheroid/organoid is surrounded by radially aligned collagen I fibers on one side and circumferentially oriented fibers on the opposite side. A coaxial rotating cylinder system was employed to construct the dual fiber topography and to pre-seed tumor spheroids/organoids within a single device. This system enables the application of different force mechanisms in the nucleation and elongation phases of collagen fiber polymerization to guide fiber alignment. In the nucleation phase, fiber alignment is enhanced by a horizontal laminar Couette flow driven by the inner cylinder rotation. In the elongation phase, fiber growth is guided by a vertical gravitational force to form a large aligned collagen matrix gel (35 × 25 × 0.5 mm) embedded with >1000 tumor spheroids. The fibers above each tumor spheroid are radially aligned along the direction of gravitational force in contrast to the circumferentially oriented fibers beneath each tumor spheroid/organoid, where the presence of the tumor interferes with the gravity-induced fiber alignment. After tumor invasion, there are more disseminated multicellular clusters on the radially aligned side, compared to the side of the tumor spheroid/organoid facing circumferentially oriented fibers. These results indicate that our 3D dual topographical model recapitulates the preference of tumors to invade and disseminate along radially aligned fibers. We anticipate that this 3D dual topographical model will have broad utility to those studying collective tumor invasion and that it has the potential to identify cancer invasion-targeted therapeutic agents.
Collapse
Affiliation(s)
- Chia-Yi Su
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alice Burchett
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Matthew Dunworth
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jong Seob Choi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andrew J Ewald
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
19
|
Hisey CL, Hearn JI, Hansford DJ, Blenkiron C, Chamley LW. Micropatterned growth surface topography affects extracellular vesicle production. Colloids Surf B Biointerfaces 2021; 203:111772. [PMID: 33894649 DOI: 10.1016/j.colsurfb.2021.111772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are micro and nanoscale packages that circulate in all bodily fluids and play an important role in intercellular communication by shuttling biomolecules to nearby and distant cells. However, producing sufficient amounts of EVs for many types of in vitro studies using standard culture methods can be challenging, and despite the success of some bioreactors in increasing EV-production, it is still largely unknown how individual culture conditions can alter the production and content of EVs. In this study, we demonstrate a simple and inexpensive micropatterning technique that can be used to produce polystyrene microtracks over a 100 mm diameter growth surface area. We then demonstrate that these microtracks can play a significant role in increasing EV production using a triple-negative breast cancer cell line (MDA-MB-231) and that these changes in EV production correlate with increases in cellular aspect ratio, alignment of the cells' long axes to the microtracks, and single-cell migration rates. These findings have implications in both biomanufacturing of EVs and potentially in enhancing the biomimicry of EVs produced in vitro.
Collapse
Affiliation(s)
- Colin L Hisey
- Obstetrics and Gynaecology, University of Auckland, FMHS Building 502-201, 85 Park Rd, Grafton, Auckland, 1023, New Zealand; Hub for Extracellular Vesicle Investigations, University of Auckland, FMHS Building 502-201, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.
| | - James I Hearn
- Molecular Medicine and Pathology, University of Auckland, FMHS Building 502-301, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.
| | - Derek J Hansford
- Biomedical Engineering, The Ohio State University, 293 Bevis Hall, 1080 Carmack Road, Columbus, OH, 43210, USA.
| | - Cherie Blenkiron
- Hub for Extracellular Vesicle Investigations, University of Auckland, FMHS Building 502-201, 85 Park Rd, Grafton, Auckland, 1023, New Zealand; Molecular Medicine and Pathology, University of Auckland, FMHS Building 502-301, 85 Park Rd, Grafton, Auckland, 1023, New Zealand; Auckland Cancer Society Research Centre, University of Auckland, FMHS, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| | - Lawrence W Chamley
- Obstetrics and Gynaecology, University of Auckland, FMHS Building 502-201, 85 Park Rd, Grafton, Auckland, 1023, New Zealand; Hub for Extracellular Vesicle Investigations, University of Auckland, FMHS Building 502-201, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.
| |
Collapse
|
20
|
Banani MA, Rahmatullah M, Farhan N, Hancox Z, Yousaf S, Arabpour Z, Moghaddam ZS, Mozafari M, Sefat F. Adipose tissue-derived mesenchymal stem cells for breast tissue regeneration. Regen Med 2021; 16:47-70. [PMID: 33533667 DOI: 10.2217/rme-2020-0045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
With an escalating incidence of breast cancer cases all over the world and the deleterious psychological impact that mastectomy has on patients along with several limitations of the currently applied modalities, it's plausible to seek unconventional approaches to encounter such a burgeoning issue. Breast tissue engineering may allow that chance via providing more personalized solutions which are able to regenerate, mimicking natural tissues also facing the witnessed limitations. This review is dedicated to explore the utilization of adipose tissue-derived mesenchymal stem cells for breast tissue regeneration among postmastectomy cases focusing on biomaterials and cellular aspects in terms of harvesting, isolation, differentiation and new tissue formation as well as scaffolds types, properties, material-host interaction and an in vitro breast tissue modeling.
Collapse
Affiliation(s)
- Mohammed A Banani
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Mohammed Rahmatullah
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Nawras Farhan
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Zoe Hancox
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Safiyya Yousaf
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Zohreh Arabpour
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Zoha Salehi Moghaddam
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK.,Interdisciplinary Research Centre in Polymer Science & Technology (IRC Polymer), University of Bradford, Bradford, BD7 1DP, UK
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, M5G 1X5, Canada
| | - Farshid Sefat
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK.,Interdisciplinary Research Centre in Polymer Science & Technology (IRC Polymer), University of Bradford, Bradford, BD7 1DP, UK
| |
Collapse
|
21
|
Al-Musawi S, Albukhaty S, Al-Karagoly H, Sulaiman GM, Alwahibi MS, Dewir YH, Soliman DA, Rizwana H. Antibacterial Activity of Honey/Chitosan Nanofibers Loaded with Capsaicin and Gold Nanoparticles for Wound Dressing. Molecules 2020; 25:E4770. [PMID: 33080798 PMCID: PMC7587596 DOI: 10.3390/molecules25204770] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/16/2020] [Accepted: 10/14/2020] [Indexed: 12/27/2022] Open
Abstract
This paper describes the preparation, characterization, and evaluation of honey/tripolyphosphate (TPP)/chitosan (HTCs) nanofibers loaded with capsaicin derived from the natural extract of hot pepper (Capsicum annuumL.) and loaded with gold nanoparticles (AuNPs) as biocompatible antimicrobial nanofibrous wound bandages in topical skin treatments. The capsaicin and AuNPs were packed within HTCs in HTCs-capsaicin, HTCs-AuNP, and HTCs-AuNPs/capsaicin nanofibrous mats. In vitro antibacterial testing against Pasteurella multocida, Klebsiella rhinoscleromatis,Staphylococcus pyogenes, and Vibrio vulnificus was conducted in comparison with difloxacin and chloramphenicol antibiotics. Cell viability and proliferation of the developed nanofibers were evaluated using an MTT assay. Finally, in vivo study of the wound-closure process was performed on New Zealand white rabbits. The results indicate that HTCs-capsaicin and HTCs-AuNPs are suitable in inhibiting bacterial growth compared with HTCs and HTCs-capsaicin/AuNP nanofibers and antibiotics (P < 0.01). The MTT assay demonstrates that the nanofibrous mats increased cell proliferation compared with the untreated control (P < 0.01). In vivo results show that the developed mats enhanced the wound-closure rate more effectively than the control samples. The novel nanofibrous wound dressings provide a relatively rapid and efficacious wound-healing ability, making the obtained nanofibers promising candidates for the development of improved bandage materials.
Collapse
Affiliation(s)
| | - Salim Albukhaty
- Department of Basic Sciences, College of Nursing, University of Misan, Maysan 62001, Iraq;
| | - Hassan Al-Karagoly
- Department of Internal and Preventive Medicine, Veterinary Medicine College, University of Al-Qadisiyah, Al-Diwaniyah 58002, Iraq;
| | - Ghassan M. Sulaiman
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq
| | - Mona S. Alwahibi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.A.); (D.A.S.); (H.R.)
| | - Yaser Hassan Dewir
- College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
- Faculty of Agriculture, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Dina A. Soliman
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.A.); (D.A.S.); (H.R.)
| | - Humaira Rizwana
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.A.); (D.A.S.); (H.R.)
| |
Collapse
|
22
|
Xue J, Wu T, Qiu J, Rutledge S, Tanes ML, Xia Y. Promoting Cell Migration and Neurite Extension along Uniaxially Aligned Nanofibers with Biomacromolecular Particles in a Density Gradient. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002031. [PMID: 33343274 PMCID: PMC7743995 DOI: 10.1002/adfm.202002031] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Indexed: 05/05/2023]
Abstract
A simple method based upon masked electrospray is reported for directly generating both unidirectional and bidirectional density gradients of biomacromolecular particles on uniaxially aligned nanofibers. The method has been successfully applied to different types of biomacromolecules, including collagen and a mixture of collagen and fibronectin or laminin, to suit different types of applications. Collagen particles in a unidirectional or bidirectional gradient are able to promote the linear migration of bone marrow stem cells or NIH-3T3 fibroblasts along the direction of increasing particle density. In the case of particles made of a mixture of collagen and fibronectin, their deposition in a bidirectional gradient promotes the migration of Schwann cells from two opposite sides toward the center, matching the scenario in peripheral nerve repair. As for a mixture of collagen and laminin, the particles in a unidirectional gradient promote the extension of neurites from embryonic chick dorsal root ganglion in the direction of increasing particle density. Taken together, the scaffolds featuring a combination of uniaxially aligned nanofibers and biomacromolecular particles in density gradient can be applied to a range of biological studies and biomedical applications.
Collapse
Affiliation(s)
- Jiajia Xue
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Tong Wu
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Jichuan Qiu
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Sarah Rutledge
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Michael L Tanes
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| |
Collapse
|
23
|
Huang Y, Hakamivala A, Li S, Nair A, Saxena R, Hsieh JT, Tang L. Chemokine releasing particle implants for trapping circulating prostate cancer cells. Sci Rep 2020; 10:4433. [PMID: 32157115 PMCID: PMC7064596 DOI: 10.1038/s41598-020-60696-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 02/04/2020] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is the most prevalent cancer in U.S. men and many other countries. Although primary PCa can be controlled with surgery or radiation, treatment options of preventing metastatic PCa are still limited. To develop a new treatment of eradicating metastatic PCa, we have created an injectable cancer trap that can actively recruit cancer cells in bloodstream. The cancer trap is composed of hyaluronic acid microparticles that have good cell and tissue compatibility and can extend the release of chemokines to 4 days in vitro. We find that erythropoietin (EPO) and stromal derived factor-1α can attract PCa in vitro. Animal results show that EPO-releasing cancer trap attracted large number of circulating PCa and significantly reduced cancer spreading to other organs compared with controls. These results support that cancer trap may serve as a unique device to sequester circulating PCa cells and subsequently reduce distant metastasis.
Collapse
Affiliation(s)
- YiHui Huang
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Amirhossein Hakamivala
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Shuxin Li
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Ashwin Nair
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Ramesh Saxena
- Division of Nephrology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Liping Tang
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA.
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
24
|
Effect of Electrospun Fiber Mat Thickness and Support Method on Cell Morphology. NANOMATERIALS 2019; 9:nano9040644. [PMID: 31010029 PMCID: PMC6523829 DOI: 10.3390/nano9040644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022]
Abstract
Electrospun fiber mats (EFMs) are highly versatile biomaterials used in a myriad of biomedical applications. Whereas some facets of EFMs are well studied and can be highly tuned (e.g., pore size, fiber diameter, etc.), other features are under characterized. For example, although substrate mechanics have been explored by several groups, most studies rely on Young's modulus alone as a characterization variable. The influence of fiber mat thickness and the effect of supports are variables that are often not considered when evaluating cell-mechanical response. To assay the role of these features in EFM scaffold design and to improve understanding of scaffold mechanical properties, we designed EFM scaffolds with varying thickness (50-200 µm) and supporting methodologies. EFM scaffolds were comprised of polycaprolactone and were either electrospun directly onto a support, suspended across an annulus (3 or 10 mm inner diameter), or "tension-released" and then suspended across an annulus. Then, single cell spreading (i.e., Feret diameter) was measured in the presence of these different features. Cells were sensitive to EFM thickness and suspended gap diameter. Overall, cell spreading was greatest for 50 µm thick EFMs suspended over a 3 mm gap, which was the smallest thickness and gap investigated. These results are counterintuitive to conventional understanding in mechanobiology, which suggests that stiffer materials, such as thicker, supported EFMs, should elicit greater cell polarization. Additional experiments with 50 µm thick EFMs on polystyrene and polydimethylsiloxane (PDMS) supports demonstrated that cells can "feel" the support underlying the EFM if it is rigid, similar to previous results in hydrogels. These results also suggest that EFM curvature may play a role in cell response, separate from Young's modulus, possibly because of internal tension generated. These parameters are not often considered in EFM design and could improve scaffold performance and ultimately patient outcomes.
Collapse
|
25
|
Biological Performance of Electrospun Polymer Fibres. MATERIALS 2019; 12:ma12030363. [PMID: 30682805 PMCID: PMC6384992 DOI: 10.3390/ma12030363] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
Abstract
The evaluation of biological responses to polymeric scaffolds are important, given that the ideal scaffold should be biocompatible, biodegradable, promote cell adhesion and aid cell proliferation. The primary goal of this research was to measure the biological responses of cells against various polymeric and collagen electrospun scaffolds (polycaprolactone (PCL) and polylactic acid (PLA) polymers: PCL⁻drug, PCL⁻collagen⁻drug, PLA⁻drug and PLA⁻collagen⁻drug); cell proliferation was measured with a cell adhesion assay and cell viability using 5-bromo-2'-deoxyuridine (BrdU) and resazurin assays. The results demonstrated that there is a distinct lack of growth of cells against any irgasan (IRG) loaded scaffolds and far greater adhesion of cells against levofloxacin (LEVO) loaded scaffolds. Fourteen-day studies revealed a significant increase in cell growth after a 7-day period. The addition of collagen in the formulations did not promote greater cell adhesion. Cell viability studies revealed the levels of IRG used in scaffolds were toxic to cells, with the concentration used 475 times higher than the EC50 value for IRG. It was concluded that the negatively charged carboxylic acid group found in LEVO is attracting positively charged fibronectin, which in turn is attracting the cell to adhere to the adsorbed proteins on the surface of the scaffold. Overall, the biological studies examined in this paper are valuable as preliminary data for potential further studies into more complex aspects of cell behaviour with polymeric scaffolds.
Collapse
|
26
|
In Kim J, Kim CS. Harnessing nanotopography of PCL/collagen nanocomposite membrane and changes in cell morphology coordinated with wound healing activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 91:824-837. [DOI: 10.1016/j.msec.2018.06.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 03/15/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
|
27
|
Chen S, Boda SK, Batra SK, Li X, Xie J. Emerging Roles of Electrospun Nanofibers in Cancer Research. Adv Healthc Mater 2018; 7:e1701024. [PMID: 29210522 PMCID: PMC5867260 DOI: 10.1002/adhm.201701024] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/01/2017] [Indexed: 02/01/2023]
Abstract
This article reviews the recent progress of electrospun nanofibers in cancer research. It begins with a brief introduction to the emerging potential of electrospun nanofibers in cancer research. Next, a number of recent advances on the important features of electrospun nanofibers critical for cancer research are discussed including the incorporation of drugs, control of release kinetics, orientation and alignment of nanofibers, and the fabrication of 3D nanofiber scaffolds. This article further highlights the applications of electrospun nanofibers in several areas of cancer research including local chemotherapy, combinatorial therapy, cancer detection, cancer cell capture, regulation of cancer cell behavior, construction of in vitro 3D cancer model, and engineering of bone microenvironment for cancer metastasis. This progress report concludes with remarks on the challenges and future directions for design, fabrication, and application of electrospun nanofibers in cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Shixuan Chen
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sunil Kumar Boda
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xiaoran Li
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
28
|
Blum KM, Drews JD, Breuer CK. Tissue-Engineered Heart Valves: A Call for Mechanistic Studies. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:240-253. [PMID: 29327671 DOI: 10.1089/ten.teb.2017.0425] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Heart valve disease carries a substantial risk of morbidity and mortality. Outcomes are significantly improved by valve replacement, but currently available mechanical and biological replacement valves are associated with complications of their own. Mechanical valves have a high rate of thromboembolism and require lifelong anticoagulation. Biological prosthetic valves have a much shorter lifespan, and they are prone to tearing and degradation. Both types of valves lack the capacity for growth, making them particularly problematic in pediatric patients. Tissue engineering has the potential to overcome these challenges by creating a neovalve composed of native tissue that is capable of growth and remodeling. The first tissue-engineered heart valve (TEHV) was created more than 20 years ago in an ovine model, and the technology has been advanced to clinical trials in the intervening decades. Some TEHVs have had clinical success, whereas others have failed, with structural degeneration resulting in patient deaths. The etiologies of these complications are poorly understood because much of the research in this field has been performed in large animals and humans, and, therefore, there are few studies of the mechanisms of neotissue formation. This review examines the need for a TEHV to treat pediatric patients with valve disease, the history of TEHVs, and a future that would benefit from extension of the reverse translational trend in this field to include small animal studies.
Collapse
Affiliation(s)
- Kevin M Blum
- 1 Center for Regenerative Medicine, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,2 The Ohio State University College of Medicine , Columbus, Ohio
| | - Joseph D Drews
- 1 Center for Regenerative Medicine, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,3 Department of Surgery, The Ohio State University Wexner Medical Center , Columbus, Ohio
| | - Christopher K Breuer
- 1 Center for Regenerative Medicine, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,3 Department of Surgery, The Ohio State University Wexner Medical Center , Columbus, Ohio
| |
Collapse
|
29
|
Design of Fiber Networks for Studying Metastatic Invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1092:289-318. [DOI: 10.1007/978-3-319-95294-9_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
Huang YL, Segall JE, Wu M. Microfluidic modeling of the biophysical microenvironment in tumor cell invasion. LAB ON A CHIP 2017; 17:3221-3233. [PMID: 28805874 PMCID: PMC6007858 DOI: 10.1039/c7lc00623c] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Tumor cell invasion, whether penetrating through the extracellular matrix (ECM) or crossing a vascular endothelium, is a critical step in the cancer metastatic cascade. Along the way from a primary tumor to a distant metastatic site, tumor cells interact actively with the microenvironment either via biomechanical (e. g. ECM stiffness) or biochemical (e.g. secreted cytokines) signals. Increasingly, it is recognized that the tumor microenvironment (TME) is a critical player in tumor cell invasion. A main challenge for the mechanistic understanding of tumor cell-TME interactions comes from the complexity of the TME, which consists of extracellular matrices, fluid flows, cytokine gradients and other cell types. It is difficult to control TME parameters in conventional in vitro experimental designs such as Boyden chambers or in vivo such as in mouse models. Microfluidics has emerged as an enabling tool for exploring the TME parameter space because of its ease of use in recreating a complex and physiologically realistic three dimensional TME with well-defined spatial and temporal control. In this perspective, we will discuss designing principles for modeling the biophysical microenvironment (biological flows and ECM) for tumor cells using microfluidic devices and the potential microfluidic technology holds in recreating a physiologically realistic tumor microenvironment. The focus will be on applications of microfluidic models in tumor cell invasion.
Collapse
Affiliation(s)
- Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, 306 Riley-Robb Hall, Ithaca, NY 14853, USA.
| | | | | |
Collapse
|
31
|
Rianna C, Radmacher M. Influence of microenvironment topography and stiffness on the mechanics and motility of normal and cancer renal cells. NANOSCALE 2017; 9:11222-11230. [PMID: 28752168 DOI: 10.1039/c7nr02940c] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The tumor microenvironment highly influences cancer cell modes and dynamics, above all during invasive and metastatic processes. When aiming at studying cancer cell behavior in vitro, the use of conventional cell culture systems, such as Petri dishes, fails in recapitulating the mechanical and topographical properties of the natural extracellular matrix (ECM). Here the versatility of stiffness-tunable hydrogels and the efficacy of the replica molding technique with silicone polymers are exploited, aiming at studying cancer and normal cell behavior with platforms able to capture the heterogeneities of the natural in vivo context. We compared the mechanical properties of normal and cancer renal cells on different stiffness value gels (with Young's moduli of 3, 17 and 31 kPa) by using atomic force microscopy (AFM) and investigated cell indentation phenomena on compliant gels with confocal microscopy. Moreover, we studied cell mechanics, morphology and migration on isotropic linear structures, spaced at 1.5 μm, aiming at mimicking the aligned fiber bundles typically observed at tumor borders. By using this approach, we could highlight differences in the way healthy and cancer renal cells react to changes in their microenvironment. Our results may potentially pave the way to unravel the complex processes involved in cancer progression, especially in tissue invasion and migration during metastasis formation.
Collapse
Affiliation(s)
- C Rianna
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany.
| | | |
Collapse
|
32
|
Rianna C, Kumar P, Radmacher M. The role of the microenvironment in the biophysics of cancer. Semin Cell Dev Biol 2017; 73:107-114. [PMID: 28746843 DOI: 10.1016/j.semcdb.2017.07.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 01/23/2023]
Abstract
During the last decades, cell mechanics has been recognized as a quantitative measure to discriminate between many physiological and pathological states of single cells. In the field of biophysics of cancer, a large body of research has been focused on the comparison between normal and cancer mechanics and slowly the hypothesis that cancer cells are softer than their normal counterparts has been accepted, even though in situ tumor tissue is usually stiffer than the surrounding normal tissue. This corroborates the idea that the extra-cellular matrix (ECM) has a critical role in regulating tumor cell properties and behavior. Rearrangements in ECM can lead to changes in cancer cell mechanics and in specific conditions the general assumption about cancer cell softening could be confuted. Here, we highlight the contribution of ECM in cancer cell mechanics and argue that the statement that cancer cells are softer than normal cells should be firmly related to the properties of cell environment and the specific stage of cancer cell progression. In particular, we will discuss that when employing cell mechanics in cancer diagnosis and discrimination, the chemical, the topographical and - last but not least - the mechanical properties of the microenvironment are very important.
Collapse
Affiliation(s)
- Carmela Rianna
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany
| | - Prem Kumar
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany
| | - Manfred Radmacher
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany.
| |
Collapse
|
33
|
Balhouse BN, Patterson L, Schmelz EM, Slade DJ, Verbridge SS. N-(3-oxododecanoyl)-L-homoserine lactone interactions in the breast tumor microenvironment: Implications for breast cancer viability and proliferation in vitro. PLoS One 2017; 12:e0180372. [PMID: 28692660 PMCID: PMC5503244 DOI: 10.1371/journal.pone.0180372] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 06/14/2017] [Indexed: 01/19/2023] Open
Abstract
It is well documented that the tumor microenvironment profoundly impacts the etiology and progression of breast cancer, yet the contribution of the resident microbiome within breast tissue remains poorly understood. Tumor microenvironmental conditions, such as hypoxia and dense tumor stroma, predispose progressive phenotypes and therapy resistance, however the role of bacteria in this interplay remains uncharacterized. We hypothesized that the effect of individual bacterial secreted molecules on breast cancer viability and proliferation would be modulated by these tumor-relevant stressors differentially for cells at varying stages of progression. To test this, we incubated human breast adenocarcinoma cells (MDA-MB-231, MCF-DCIS.com) and non-malignant breast epithelial cells (MCF-10A) with N-(3-oxododecanoyl)-L-homoserine lactone (OdDHL), a quorum-sensing molecule from Pseudomonas aeruginosa that regulates bacterial stress responses. This molecule was selected because Pseudomonas was recently characterized as a significant fraction of the breast tissue microbiome and OdDHL is documented to impact mammalian cell viability. After OdDHL treatment, we demonstrated the greatest decrease in viability with the more malignant MDA-MB-231 cells and an intermediate MCF-DCIS.com (ductal carcinoma in situ) response. The responses were also culture condition (i.e. microenvironment) dependent. These results contrast the MCF-10A response, which demonstrated no change in viability in any culture condition. We further determined that the observed trends in breast cancer viability were due to modulation of proliferation for both cell types, as well as the induction of necrosis for MDA-MB-231 cells in all conditions. Our results provide evidence that bacterial quorum-sensing molecules interact with the host tissue environment to modulate breast cancer viability and proliferation, and that the effect of OdDHL is dependent on both cell type as well as microenvironment. Understanding the interactions between bacterial signaling molecules and the host tissue environment will allow for future studies that determine the contribution of bacteria to the onset, progression, and therapy response of breast cancer.
Collapse
Affiliation(s)
- Brittany N. Balhouse
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, United States of America
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States of America
| | - Logan Patterson
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, United States of America
- Department of Pathology, University of Virginia, Charlottesville, VA, United States of America
| | - Eva M. Schmelz
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, United States of America
| | - Daniel J. Slade
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, United States of America
| | - Scott S. Verbridge
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA, United States of America
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States of America
- * E-mail:
| |
Collapse
|
34
|
Liu XQ, Fourel L, Dalonneau F, Sadir R, Leal S, Lortat-Jacob H, Weidenhaupt M, Albiges-Rizo C, Picart C. Biomaterial-enabled delivery of SDF-1α at the ventral side of breast cancer cells reveals a crosstalk between cell receptors to promote the invasive phenotype. Biomaterials 2017; 127:61-74. [PMID: 28279922 PMCID: PMC5777630 DOI: 10.1016/j.biomaterials.2017.02.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 02/17/2017] [Accepted: 02/26/2017] [Indexed: 12/31/2022]
Abstract
The SDF-1α chemokine (CXCL12) is a potent bioactive chemoattractant known to be involved in hematopoietic stem cell homing and cancer progression. The associated SDF-1α/CXCR4 receptor signaling is a hallmark of aggressive tumors, which can metastasize to distant sites such as lymph nodes, lung and bone. Here, we engineered a biomimetic tumoral niche made of a thin and soft polyelectrolyte film that can retain SDF-1α to present it, in a spatially-controlled manner, at the ventral side of the breast cancer cells. Matrix-bound SDF-1α but not soluble SDF-1α induced a striking increase in cell spreading and migration in a serum-containing medium, which was associated with the formation of lamellipodia and filopodia in MDA-MB231 cells and specifically mediated by CXCR4. Other Knockdown and inhibition experiments revealed that CD44, the major hyaluronan receptor, acted in concert, via a spatial coincidence, to drive a specific matrix-bound SDFα-induced cell response associated with ERK signaling. In contrast, the β1 integrin adhesion receptor played only a minor role on cell polarity. The CXCR4/CD44 mediated cellular response to matrix-bound SDF-1α involved the Rac1 RhoGTPase and was sustained solely in the presence of matrix-bound SDFα, in contrast with the transient signaling observed in response to soluble SDF-1α. Our results highlight that a biomimetic tumoral niche enables to reveal potent cellular effects and so far hidden molecular mechanisms underlying the breast cancer response to chemokines. These results open new insights for the design of future innovative therapies in metastatic cancers, by inhibiting CXCR4-mediated signaling in the tumoral niche via dual targeting of receptors (CXCR4 and CD44) or of associated signaling molecules (CXCR4 and Rac1).
Collapse
Affiliation(s)
- Xi Qiu Liu
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France; FONDATION ARC, 9 rue Guy Môquet, 94803, Villejuif, France; Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Laure Fourel
- Inserm U1209, Université Grenoble Alpes, Institut Albert Bonniot, Site Santé, 38042, Grenoble cedex 9, France; CNRS UMR5309, Institute for Advanced Biosciences, Institut Albert Bonniot, 38700, La Tronche, France
| | - Fabien Dalonneau
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Rabia Sadir
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CNRS, CEA, F-38027, Grenoble, France
| | - Salome Leal
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Hugues Lortat-Jacob
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CNRS, CEA, F-38027, Grenoble, France
| | - Marianne Weidenhaupt
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Corinne Albiges-Rizo
- Inserm U1209, Université Grenoble Alpes, Institut Albert Bonniot, Site Santé, 38042, Grenoble cedex 9, France; CNRS UMR5309, Institute for Advanced Biosciences, Institut Albert Bonniot, 38700, La Tronche, France
| | - Catherine Picart
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France.
| |
Collapse
|
35
|
Domura R, Sasaki R, Okamoto M, Hirano M, Kohda K, Napiwocki B, Turng LS. Comprehensive study on cellular morphologies, proliferation, motility, and epithelial-mesenchymal transition of breast cancer cells incubated on electrospun polymeric fiber substrates. J Mater Chem B 2017; 5:2588-2600. [PMID: 32264037 DOI: 10.1016/j.mtchem.2018.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The progress of microenvironment-mediated tumor progression in an artificial extracellular matrix explores the design criteria to understand the cancer progression mechanism and metastatic potential. This study was aimed at examining the combination of both surface topographies (fiber alignments) and different stiffness of polymeric substrates (PLLA and PCL) to evaluate the effects on the cellular morphologies, proliferation, motility, and gene expression regarding epithelial to mesenchymal transition (EMT) of two different types of breast cancer cells (MDA-MB-231 and MCF-7). The cellular morphologies (roundness and nuclear elongation factor), E-cadherin and vimentin expression, and cellular motility in terms of cellular migration speed, persistent time, and diffusivity have been comprehensively discussed. We demonstrated that the microenvironment of cell culture substrates influences cancer progression and metastatic potential.
Collapse
Affiliation(s)
- Ryota Domura
- Advanced Polymeric Nanostructured Materials Engineering, Graduate School of Engineering, Toyota Technological Institute, 2-12-1 Hisakata, Tempaku, Nagoya 468 8511, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
Leight JL, Drain AP, Weaver VM. Extracellular Matrix Remodeling and Stiffening Modulate Tumor Phenotype and Treatment Response. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2017. [DOI: 10.1146/annurev-cancerbio-050216-034431] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jennifer L. Leight
- Department of Biomedical Engineering and The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
| | - Allison P. Drain
- University of California, Berkeley–University of California, San Francisco Graduate Program in Bioengineering, Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, Department of Anatomy, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and Helen Diller Comprehensive Cancer Center, University of California, San Francisco, California 94143
| |
Collapse
|
37
|
Kennedy KM, Bhaw-Luximon A, Jhurry D. Cell-matrix mechanical interaction in electrospun polymeric scaffolds for tissue engineering: Implications for scaffold design and performance. Acta Biomater 2017; 50:41-55. [PMID: 28011142 DOI: 10.1016/j.actbio.2016.12.034] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/10/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022]
Abstract
Engineered scaffolds produced by electrospinning of biodegradable polymers offer a 3D, nanofibrous environment with controllable structural, chemical, and mechanical properties that mimic the extracellular matrix of native tissues and have shown promise for a number of tissue engineering applications. The microscale mechanical interactions between cells and electrospun matrices drive cell behaviors including migration and differentiation that are critical to promote tissue regeneration. Recent developments in understanding these mechanical interactions in electrospun environments are reviewed, with emphasis on how fiber geometry and polymer structure impact on the local mechanical properties of scaffolds, how altering the micromechanics cues cell behaviors, and how, in turn, cellular and extrinsic forces exerted on the matrix mechanically remodel an electrospun scaffold throughout tissue development. Techniques used to measure and visualize these mechanical interactions are described. We provide a critical outlook on technological gaps that must be overcome to advance the ability to design, assess, and manipulate the mechanical environment in electrospun scaffolds toward constructs that may be successfully applied in tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE Tissue engineering requires design of scaffolds that interact with cells to promote tissue development. Electrospinning is a promising technique for fabricating fibrous, biomimetic scaffolds. Effects of electrospun matrix microstructure and biochemical properties on cell behavior have been extensively reviewed previously; here, we consider cell-matrix interaction from a mechanical perspective. Micromechanical properties as a driver of cell behavior has been well established in planar substrates, but more recently, many studies have provided new insights into mechanical interaction in fibrillar, electrospun environments. This review provides readers with an overview of how electrospun scaffold mechanics and cell behavior work in a dynamic feedback loop to drive tissue development, and discusses opportunities for improved design of mechanical environments that are conducive to tissue development.
Collapse
|
38
|
Accelerated Wound Closure - Differently Organized Nanofibers Affect Cell Migration and Hence the Closure of Artificial Wounds in a Cell Based In Vitro Model. PLoS One 2017; 12:e0169419. [PMID: 28060880 PMCID: PMC5218412 DOI: 10.1371/journal.pone.0169419] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/17/2016] [Indexed: 02/07/2023] Open
Abstract
Nanofiber meshes holds great promise in wound healing applications by mimicking the topography of extracellular matrix, hence providing guidance for crucial cells involved in the regenerative processes. Here we explored the influence of nanofiber alignment on fibroblast behavior in a novel in vitro wound model. The model included electrospun poly-ε-caprolactone scaffolds with different nanofiber orientation. Fibroblasts were cultured to confluency for 24h before custom-made inserts were removed, creating cell-free zones serving as artificial wounds. Cell migration into these wounds was evaluated at 0-, 48- and 96h. Cell morphological analysis was performed using nuclei- and cytoskeleton stainings. Cell viability was assessed using a biochemical assay. This study demonstrates a novel in vitro wound assay, for exploring of the impact of nanofibers on wound healing. Additionally we show that it’s possible to affect the process of wound closure in a spatial manner using nanotopographies, resulting in faster closure on aligned fiber substrates.
Collapse
|
39
|
Domura R, Sasaki R, Okamoto M, Hirano M, Kohda K, Napiwocki B, Turng LS. Comprehensive study on cellular morphologies, proliferation, motility, and epithelial–mesenchymal transition of breast cancer cells incubated on electrospun polymeric fiber substrates. J Mater Chem B 2017; 5:2588-2600. [DOI: 10.1039/c7tb00207f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Aligned fibers substrates caused elongation and alignment of the MDA-MB-231 cells along the fiber directionsviareducing the cell roundness and E-cadherin expression.
Collapse
Affiliation(s)
- Ryota Domura
- Advanced Polymeric Nanostructured Materials Engineering
- Graduate School of Engineering
- Toyota Technological Institute
- Tempaku
- Japan
| | - Rie Sasaki
- Advanced Polymeric Nanostructured Materials Engineering
- Graduate School of Engineering
- Toyota Technological Institute
- Tempaku
- Japan
| | - Masami Okamoto
- Advanced Polymeric Nanostructured Materials Engineering
- Graduate School of Engineering
- Toyota Technological Institute
- Tempaku
- Japan
| | | | | | - Brett Napiwocki
- Department of Engineering Physics
- University of Wisconsin-Madison
- USA
| | - Lih-Sheng Turng
- Wisconsin Institute for Discovery and Polymer Engineering Center
- Department of Mechanical Engineering
- University of Wisconsin-Madison
- USA
| |
Collapse
|
40
|
Shologu N, Szegezdi E, Lowery A, Kerin M, Pandit A, Zeugolis DI. Recreating complex pathophysiologies in vitro with extracellular matrix surrogates for anticancer therapeutics screening. Drug Discov Today 2016; 21:1521-1531. [DOI: 10.1016/j.drudis.2016.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/17/2016] [Accepted: 06/01/2016] [Indexed: 12/12/2022]
|
41
|
Czeisler C, Short A, Nelson T, Gygli P, Ortiz C, Catacutan FP, Stocker B, Cronin J, Lannutti J, Winter J, Otero JJ. Surface topography during neural stem cell differentiation regulates cell migration and cell morphology. J Comp Neurol 2016; 524:3485-3502. [PMID: 27418162 DOI: 10.1002/cne.24078] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 07/08/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022]
Abstract
We sought to determine the contribution of scaffold topography to the migration and morphology of neural stem cells by mimicking anatomical features of scaffolds found in vivo. We mimicked two types of central nervous system scaffolds encountered by neural stem cells during development in vitro by constructing different diameter electrospun polycaprolactone (PCL) fiber mats, a substrate that we have shown to be topographically similar to brain scaffolds. We compared the effects of large fibers (made to mimic blood vessel topography) with those of small-diameter fibers (made to mimic radial glial process topography) on the migration and differentiation of neural stem cells. Neural stem cells showed differential migratory and morphological reactions with laminin in different topographical contexts. We demonstrate, for the first time, that neural stem cell biological responses to laminin are dependent on topographical context. Large-fiber topography without laminin prevented cell migration, which was partially reversed by treatment with rock inhibitor. Cell morphology complexity assayed by fractal dimension was inhibited in nocodazole- and cytochalasin-D-treated neural precursor cells in large-fiber topography, but was not changed in small-fiber topography with these inhibitors. These data indicate that cell morphology has different requirements on cytoskeletal proteins dependent on the topographical environment encountered by the cell. We propose that the physical structure of distinct scaffolds induces unique signaling cascades that regulate migration and morphology in embryonic neural precursor cells. J. Comp. Neurol. 524:3485-3502, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Catherine Czeisler
- Department of Pathology, Division of Neuropathology, The Ohio State University College of Medicine, Columbus, Ohio, 43210
| | - Aaron Short
- Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, Ohio, 43210
| | - Tyler Nelson
- Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, Ohio, 43210
| | - Patrick Gygli
- Department of Pathology, Division of Neuropathology, The Ohio State University College of Medicine, Columbus, Ohio, 43210
| | - Cristina Ortiz
- Department of Pathology, Division of Neuropathology, The Ohio State University College of Medicine, Columbus, Ohio, 43210
| | - Fay Patsy Catacutan
- Department of Pathology, Division of Neuropathology, The Ohio State University College of Medicine, Columbus, Ohio, 43210
| | - Ben Stocker
- Department of Pathology, Division of Neuropathology, The Ohio State University College of Medicine, Columbus, Ohio, 43210
| | - James Cronin
- Department of Pathology, Division of Neuropathology, The Ohio State University College of Medicine, Columbus, Ohio, 43210
| | - John Lannutti
- Department of Materials Science and Engineering, the Ohio State University College of Engineering, Columbus, Ohio, 43210
| | - Jessica Winter
- Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, Ohio, 43210. .,William G. Lowrie Department of Chemical Engineering, The Ohio State University College of Engineering, Columbus, Ohio, 43210.
| | - José Javier Otero
- Department of Pathology, Division of Neuropathology, The Ohio State University College of Medicine, Columbus, Ohio, 43210.
| |
Collapse
|
42
|
Aligned Nanotopography Promotes a Migratory State in Glioblastoma Multiforme Tumor Cells. Sci Rep 2016; 6:26143. [PMID: 27189099 PMCID: PMC4870554 DOI: 10.1038/srep26143] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/27/2016] [Indexed: 01/17/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive, Grade IV astrocytoma with a poor survival rate, primarily due to the GBM tumor cells migrating away from the primary tumor site along the nanotopography of white matter tracts and blood vessels. It is unclear whether this nanotopography influences the biomechanical properties (i.e. cytoskeletal stiffness) of GBM tumor cells. Although GBM tumor cells have an innate propensity to migrate, we believe this capability is enhanced due to the influence of nanotopography on the tumor cells’ biomechanical properties. In this study, we used an aligned nanofiber film that mimics the nanotopography in the tumor microenvironment to investigate the mechanical properties of GBM tumor cells in vitro. The data demonstrate that the cytoskeletal stiffness, cell traction stress, and focal adhesion area were significantly lower in the GBM tumor cells compared to healthy astrocytes. Moreover, the cytoskeletal stiffness was significantly reduced when cultured on aligned nanofiber films compared to smooth and randomly aligned nanofiber films. Gene expression analysis showed that tumor cells cultured on the aligned nanotopography upregulated key migratory genes and downregulated key proliferative genes. Therefore, our data suggest that the migratory potential is elevated when GBM tumor cells are migrating along aligned nanotopographical substrates.
Collapse
|
43
|
Tavangar A, Premnath P, Tan B, Venkatakrishnan K. Noble Hybrid Nanostructures as Efficient Anti-Proliferative Platforms for Human Breast Cancer Cell. ACS APPLIED MATERIALS & INTERFACES 2016; 8:10253-10265. [PMID: 27035281 DOI: 10.1021/acsami.6b02720] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Nanomaterials have proven to possess great potential in biomaterials research. Recently, they have suggested considerable promise in cancer diagnosis and therapy. Among others, silicon (Si) nanomaterials have been extensively employed for various biomedical applications; however, the utilization of Si for cancer therapy has been limited to nanoparticles, and its potential as anticancer substrates has not been fully explored. Noble nanoparticles have also received considerable attention owing to unique anticancer properties to improve the efficiency of biomaterials for numerous biological applications. Nevertheless, immobilization and control over delivery of the nanoparticles have been challenge. Here, we develop hybrid nanoplatforms to efficiently hamper breast cancer cell adhesion and proliferation. Platforms are synthesized by femtosecond laser processing of Si into multiphase nanostructures, followed by sputter-coating with gold (Au)/gold-palladium (Au-Pd) nanoparticles. The performance of the developed platforms was then examined by exploring the response of normal fibroblast and metastatic breast cancer cells. Our results from the quantitative and qualitative analyses show a dramatic decrease in the number of breast cancer cells on the hybrid platform compared to untreated substrates. Whereas, fibroblast cells form stable adhesion with stretched and elongated cytoskeleton and actin filaments. The hybrid platforms perform as dual-acting cytophobic/cytostatic stages where Si nanostructures depress breast cancer cell adhesion while immobilized Au/Au-Pd nanoparticles are gradually released to affect any surviving cell on the nanostructures. The nanoparticles are believed to be taken up by breast cancer cells via endocytosis, which subsequently alter the cell nucleus and may cause cell death. The findings suggest that the density of nanostructures and concentration of coated nanoparticles play critical roles on cytophobic/cytostatic properties of the platforms on human breast cancer cells while having no or even cytophilic effects on fibroblast cells. Because of the remarkable contrary responses of normal and cancer cells to the proposed platform, we envision that it will provide novel applications in cancer research.
Collapse
Affiliation(s)
- Amirhossein Tavangar
- Micro/Nanofabrication Laboratory, Department of Mechanical and Industrial Engineering, Ryerson University , 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Priyatha Premnath
- Micro/Nanofabrication Laboratory, Department of Mechanical and Industrial Engineering, Ryerson University , 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Bo Tan
- Nanocharacterization Laboratory, Department of Aerospace Engineering, Ryerson University , 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Krishnan Venkatakrishnan
- Micro/Nanofabrication Laboratory, Department of Mechanical and Industrial Engineering, Ryerson University , 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Affiliate Scientist, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
44
|
Topography induces differential sensitivity on cancer cell proliferation via Rho-ROCK-Myosin contractility. Sci Rep 2016; 6:19672. [PMID: 26795068 PMCID: PMC4726280 DOI: 10.1038/srep19672] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/07/2015] [Indexed: 01/15/2023] Open
Abstract
Although the role of stiffness on proliferative response of cancer cells has been well studied, little is known about the effect of topographic cues in guiding cancer cell proliferation. Here, we examined the effect of topographic cues on cancer cell proliferation using micron scale topographic features and observed that anisotropic features like microgratings at specific dimension could reduce proliferation of non-cancer breast epithelial cells (MCF-10A) but not that for malignant breast cancer cells (MDA-MB-231 and MCF-7). However, isotropic features such as micropillars did not affect proliferation of MCF-10A, indicating that the anisotropic environmental cues are essential for this process. Interestingly, acto-myosin contraction inhibitory drugs, Y-27632 and blebbistatin prevented micrograting-mediated inhibition on proliferation. Here, we propose the concept of Mechanically-Induced Dormancy (MID) where topographic cues could activate Rho-ROCK-Myosin signaling to suppress non-cancerous cells proliferation whereas malignant cells are resistant to this inhibitory barrier and therefore continue uncontrolled proliferation.
Collapse
|
45
|
Liu JP, Liu D, Gu JF, Zhu MM, Cui L. Shikonin inhibits the cell viability, adhesion, invasion and migration of the human gastric cancer cell line MGC-803 via the Toll-like receptor 2/nuclear factor-kappa B pathway. J Pharm Pharmacol 2015; 67:1143-55. [PMID: 25880237 DOI: 10.1111/jphp.12402] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 01/25/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Shikonin is an active naphthoquinone pigment isolated from the root of Lithospermum erythrorhizon. This study was designed to explore the inhibition of Shikonin on cell viability, adhesion, migration and invasion ability of gastric cancer (GC) and its possible mechanism. METHODS 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed for cell viability and adhesion ability of MGC-803 cells. Cell scratch repair experiments were conducted for the determination of migration ability while transwell assay for cell invasion ability. Western blot analysis and real-time polymerase chain reaction assay were used for the detection of protein and mRNA expressions. KEY FINDINGS Fifty per cent inhibitory concentration of Shikonin on MGC-803 cells was 1.854 μm. Shikonin (1 μm) inhibited significantly the adhesion, invasion and migratory ability of MGC-803 cells. Interestingly, Shikonin in the presence or absence of anti-Toll-like receptor 2 (TLR2) antibody (2 μg) and nuclear factor-kappa B (NF-κB) inhibitor MG-132 (10 μm) could decrease these ability of MGC-803 cells markedly, as well as the expression levels of matrix metalloproteinases (MMP)-2, MMP-7, TLR2 and p65 NF-κB. In addition, the co-incubation of Shikonin and anti-TLR2/MG-132 has a significant stronger activity than anti-TLR2 or MG-132 alone. CONCLUSIONS The results indicated that Shikonin could suppress the cell viability, adhesion, invasion and migratory ability of MGC-803 cells through TLR2- or NF-κB-mediated pathway. Our findings provide novel information for the treatment of Shikonin on GC.
Collapse
Affiliation(s)
- Ji Ping Liu
- Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, China.,Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Dan Liu
- Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, China
| | - Jun Fei Gu
- Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, China
| | - Mao Mao Zhu
- Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, China
| | - Li Cui
- Key Laboratory of New Drug Delivery Systems of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, China
| |
Collapse
|