1
|
Basmaeil Y, Subayyil AA, Kulayb HB, Kondkar AA, Alrodayyan M, Khatlani T. Partial Inhibition of Epithelial-to-Mesenchymal Transition (EMT) Phenotypes by Placenta-Derived DBMSCs in Human Breast Cancer Cell Lines, In Vitro. Cells 2024; 13:2131. [PMID: 39768220 PMCID: PMC11674051 DOI: 10.3390/cells13242131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Stem cell-based therapies hold significant potential for cancer treatment due to their unique properties, including migration toward tumor niche, secretion of bioactive molecules, and immunosuppression. Mesenchymal stem cells (MSCs) from adult tissues can inhibit tumor progression, angiogenesis, and apoptosis of cancer cells. We have previously reported the isolation and characterization of placenta-derived decidua basalis mesenchymal stem cells (DBMSCs), which demonstrated higher levels of pro-migratory and anti-apoptotic genes, indicating potential anti-cancer effects. In this study, we analyzed the anti-cancer effects of DBMSCs on human breast cancer cell lines MDA231 and MCF7, with MCF 10A used as control. We also investigated how these cancer cells lines affect the functional competence of DBMSCs. By co-culturing DBMSCs with cancer cells, we analyzed changes in functions of both cell types, as well as alterations in their genomic and proteomic profile. Our results showed that treatment with DBMSCs significantly reduced the functionality of MDA231 and MCF7 cells, while MCF 10A cells remained unaffected. DBMSC treatment decreased epithelial-to-mesenchymal transition (EMT)-related protein levels in MDA231 cells and modulated expression of other cancer-related genes in MDA231 and MCF7 cells. Although cancer cells reduced DBMSC proliferation, they increased their expression of anti-apoptotic genes. These findings suggest that DBMSCs can inhibit EMT-related proteins and reduce the invasive characteristics of MDA231 and MCF7 breast cancer cells, highlighting their potential as candidates for cell-based cancer therapies.
Collapse
Affiliation(s)
- Yasser Basmaeil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Abdullah Al Subayyil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Haya Bin Kulayb
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Altaf A. Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia;
| | - Maha Alrodayyan
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| |
Collapse
|
2
|
Eikanger MM, Sane S, Schraufnagel KS, Slunecka JL, Potts RA, Freeling J, Sereda G, Rasulev B, Brockstein RL, Emon MAB, Saif MTA, Rezvani K. Veratridine, a plant-derived alkaloid, suppresses the hyperactive Rictor-mTORC2 pathway: a new targeted therapy for primary and metastatic colorectal cancer. RESEARCH SQUARE 2024:rs.3.rs-5199838. [PMID: 39502780 PMCID: PMC11537347 DOI: 10.21203/rs.3.rs-5199838/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2024]
Abstract
Despite considerable advances to improve colorectal cancer (CRC) survival over the last decade, therapeutic challenges remain due to the rapid metastatic dissemination of primary tumors and screening limitations. Meanwhile, the rise of CRC in younger adults (Early-onset CRC), commonly diagnosed with a metastatic form of the disease, shows the pressing need to develop more effective targeted therapies to decrease the high mortality rates associated with metastatic disease. Hyperactivation of the Rictor-mTORC2-AKT signaling pathway drives key metastatic players in diverse malignant tumors, including early- and late-onset colorectal cancer. Selective mTORC2 inhibitors are becoming a potential treatment strategy for CRC due to the therapeutic limitations of mTORC1 inhibitors. Veratridine (VTD), a lipid-soluble alkaloid extracted from Liliaceae plants, can transcriptionally increase UBXN2A, which induces 26S proteasomal degradation of the Rictor protein, a key member in the mTORC2 complex. Destabilization of Rictor protein by VTD decreases Akt phosphorylation on Ser473, which is responsible for metastatic signaling downstream of the mTORC2 pathway in diverse malignant tumors. VTD decreases the population of metastatic colon cancer stem cells and functions as an angiogenesis inhibitor. VTD effectively reduces the spheroid growth rate and restricts cell migration. Live cell migration and invasion assays alongside biomechanical-force-based experiments revealed that VTD suppresses colon cancer cell invasiveness and the ensuing risk of tumor metastasis. A CRC mouse model that mimics the natural stages of human sporadic CRC revealed that VTD treatment significantly decreases tumor growth in a UBXN2A-dependent manner. This study showed a novel mechanistic connection between a ubiquitin-like protein and mTORC2-dependent migration and invasion in CRC tumors. This study revealed the therapeutic benefit of selective inhibition of Rictor in CRC, particularly in tumors with a hyperactive Rictor-mTORC2 signaling pathway. Finally, this study opened a new platform for repurposing VTD, a supplemental anti-hypertension molecule, into an effective targeted therapy in CRC tumors.
Collapse
Affiliation(s)
| | - Sanam Sane
- University of South Dakota Sanford School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Elbialy A, Kappala D, Desai D, Wang P, Fadiel A, Wang SJ, Makary MS, Lenobel S, Sood A, Gong M, Dason S, Shabsigh A, Clinton S, Parwani AV, Putluri N, Shvets G, Li J, Liu X. Patient-Derived Conditionally Reprogrammed Cells in Prostate Cancer Research. Cells 2024; 13:1005. [PMID: 38920635 PMCID: PMC11201841 DOI: 10.3390/cells13121005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024] Open
Abstract
Prostate cancer (PCa) remains a leading cause of mortality among American men, with metastatic and recurrent disease posing significant therapeutic challenges due to a limited comprehension of the underlying biological processes governing disease initiation, dormancy, and progression. The conventional use of PCa cell lines has proven inadequate in elucidating the intricate molecular mechanisms driving PCa carcinogenesis, hindering the development of effective treatments. To address this gap, patient-derived primary cell cultures have been developed and play a pivotal role in unraveling the pathophysiological intricacies unique to PCa in each individual, offering valuable insights for translational research. This review explores the applications of the conditional reprogramming (CR) cell culture approach, showcasing its capability to rapidly and effectively cultivate patient-derived normal and tumor cells. The CR strategy facilitates the acquisition of stem cell properties by primary cells, precisely recapitulating the human pathophysiology of PCa. This nuanced understanding enables the identification of novel therapeutics. Specifically, our discussion encompasses the utility of CR cells in elucidating PCa initiation and progression, unraveling the molecular pathogenesis of metastatic PCa, addressing health disparities, and advancing personalized medicine. Coupled with the tumor organoid approach and patient-derived xenografts (PDXs), CR cells present a promising avenue for comprehending cancer biology, exploring new treatment modalities, and advancing precision medicine in the context of PCa. These approaches have been used for two NCI initiatives (PDMR: patient-derived model repositories; HCMI: human cancer models initiatives).
Collapse
Affiliation(s)
- Abdalla Elbialy
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL 60637, USA
| | - Deepthi Kappala
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Dhruv Desai
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Peng Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Ahmed Fadiel
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL 60637, USA
| | - Shang-Jui Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mina S. Makary
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Division of Vascular and Interventional Radiology, Department of Radiology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Scott Lenobel
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Division of Musculoskeletal Imaging, Department of Radiology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Akshay Sood
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Michael Gong
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Shawn Dason
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ahmad Shabsigh
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Steven Clinton
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
| | - Anil V. Parwani
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14850, USA
| | - Jenny Li
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xuefeng Liu
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (A.E.)
- Departments of Pathology, Urology, and Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
4
|
Mustafa SK, Khan MF, Sagheer M, Kumar D, Pandey S. Advancements in biosensors for cancer detection: revolutionizing diagnostics. Med Oncol 2024; 41:73. [PMID: 38372827 DOI: 10.1007/s12032-023-02297-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/28/2023] [Indexed: 02/20/2024]
Abstract
Cancer stands as the reigning champion of life-threatening diseases, casting a shadow with the highest global mortality rate. Unleashing the power of early cancer treatment is a vital weapon in the battle for efficient and positive outcomes. Yet, conventional screening procedures wield limitations of exorbitant costs, time-consuming endeavors, and impracticality for repeated testing. Enter bio-marker-based cancer diagnostics, which emerge as a formidable force in the realm of early detection, disease progression assessment, and ultimate cancer therapy. These remarkable devices boast a reputation for their exceptional sensitivity, streamlined setup requirements, and lightning fast response times. In this study, we embark on a captivating exploration of the most recent advancements and enhancements in the field of electrochemical marvels, targeting the detection of numerous cancer biomarkers. With each breakthrough, we inch closer to a future where cancer's grip on humanity weakens, guided by the promise of personalized treatment and improved patient outcomes. Together, we unravel the mysteries that cancer conceals and illuminate a path toward triumph against this daunting adversary. This study celebrates the relentless pursuit of progress, where electrochemical innovations take center stage in the quest for a world free from the clutches of carcinoma.
Collapse
Affiliation(s)
- Syed Khalid Mustafa
- Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Zip 71491, Tabuk, Saudi Arabia.
| | - Mohd Farhan Khan
- Faculty of Science, Gagan College of Management & Technology, Aligarh, 202002, India
| | - Mehak Sagheer
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Sadanand Pandey
- Faculty of Applied Sciences and Biotechnology, School of Bioengineering and Food Technology, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| |
Collapse
|
5
|
Arman S, Tilley RD, Gooding JJ. A review of electrochemical impedance as a tool for examining cell biology and subcellular mechanisms: merits, limits, and future prospects. Analyst 2024; 149:269-289. [PMID: 38015145 DOI: 10.1039/d3an01423a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Herein the development of cellular impedance biosensors, electrochemical impedance spectroscopy, and the general principles and terms associated with the cell-electrode interface is reviewed. This family of techniques provides quantitative and sensitive information into cell responses to stimuli in real-time with high temporal resolution. The applications of cell-based impedance biosensors as a readout in cell biology is illustrated with a diverse range of examples. The current state of the field, its limitations, the possible available solutions, and the potential benefits of developing biosensors are discussed.
Collapse
Affiliation(s)
- Seyedyousef Arman
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Australia Centre for Nanomedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Richard D Tilley
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - J Justin Gooding
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Australia Centre for Nanomedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
6
|
Tan YQ, Chiou YS, Guo H, Zhang S, Huang X, Dukanya D, Kumar AM, Basappa S, Liu S, Zhu T, Basappa B, Pandey V, Lobie PE. Vertical pathway inhibition of receptor tyrosine kinases and BAD with synergistic efficacy in triple negative breast cancer. NPJ Precis Oncol 2024; 8:8. [PMID: 38200104 PMCID: PMC10781691 DOI: 10.1038/s41698-023-00489-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Aberrant activation of the PI3K/AKT signaling axis along with the sustained phosphorylation of downstream BAD is associated with a poor outcome of TNBC. Herein, the phosphorylated to non-phosphorylated ratio of BAD, an effector of PI3K/AKT promoting cell survival, was observed to be correlated with worse clinicopathologic indicators of outcome, including higher grade, higher proliferative index and lymph node metastasis. The structural optimization of a previously reported inhibitor of BAD-Ser99 phosphorylation was therefore achieved to generate a small molecule inhibiting the phosphorylation of BAD at Ser99 with enhanced potency and improved oral bioavailability. The molecule 2-((4-(2,3-dichlorophenyl)piperazin-1-yl)(pyridin-3-yl)methyl) phenol (NCK) displayed no toxicity at supra-therapeutic doses and was therefore assessed for utility in TNBC. NCK promoted apoptosis and G0/G1 cell cycle arrest of TNBC cell lines in vitro, concordant with gene expression analyses, and reduced in vivo xenograft growth and metastatic burden, demonstrating efficacy as a single agent. Additionally, combinatorial oncology compound library screening demonstrated that NCK synergized with tyrosine kinase inhibitors (TKIs), specifically OSI-930 or Crizotinib in reducing cell viability and promoting apoptosis of TNBC cells. The synergistic effects of NCK and TKIs were also observed in vivo with complete regression of a percentage of TNBC cell line derived xenografts and prevention of metastatic spread. In patient-derived TNBC xenograft models, NCK prolonged survival times of host animals, and in combination with TKIs generated superior survival outcomes to single agent treatment. Hence, this study provides proof of concept to further develop rational and mechanistic based therapeutic strategies to ameliorate the outcome of TNBC.
Collapse
Grants
- This research was supported by the National Natural Science Foundation of China (82172618 to P.E.L. and 82102768 to Y.Q.T.), China; the Shenzhen Key Laboratory of Innovative Oncotherapeutics (ZDSYS20200820165400003 to P.E.L.) (Shenzhen Science and Technology Innovation Commission), China; Shenzhen Development and Reform Commission Subject Construction Project ([2017]1434 to P.E.L.), China; Universities Stable Funding Key Projects (WDZC20200821150704001 to P.E.L.), China; Guangdong Basic and Applied Basic Research Foundation (2020A1515111064 to Y.Q.T.), China; The Shenzhen Bay Laboratory, Oncotherapeutics (21310031 to P.E.L.), China; Overseas Research Cooperation Project (HW2020008 to V.P.) (Tsinghua Shenzhen International Graduate School), China; Research Fund, Kaohsiung Medical University (KMU-Q112002 to Y.C.), Taiwan and China Postdoctoral Science Foundation (2022M721894 to X.H.), China.
Collapse
Affiliation(s)
- Yan Qin Tan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yi-Shiou Chiou
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Hui Guo
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Shuwei Zhang
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Xiaoming Huang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Dukanya Dukanya
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India
| | - Arun M Kumar
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India
| | - Shreeja Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Key Laboratory of Breast Cancer in Shanghai, Innovation Center for Cell Signaling Network, Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Tao Zhu
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China
- Department of Oncology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
- Hefei National Laboratory for Physical Sciences, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, 570006, Mysore, India.
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China.
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China.
| | - Peter E Lobie
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, People's Republic of China.
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, Guangdong, People's Republic of China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, Guangdong, People's Republic of China.
| |
Collapse
|
7
|
Li D, Liu W, Sun S, Zhang Y, Zhang P, Feng G, Wei J, Chai L. Chinese herbal formula, modified Xianfang Huoming Yin, alleviates the inflammatory proliferation of rat synoviocytes induced by IL-1β through regulating the migration and differentiation of T lymphocytes. JOURNAL OF ETHNOPHARMACOLOGY 2023; 309:116297. [PMID: 36849102 DOI: 10.1016/j.jep.2023.116297] [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: 11/13/2022] [Revised: 02/09/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xianfang Huoming Yin (XFH) is a traditional Chinese herbal formula, which has the effect of clearing heat and detoxifying toxins, dispersing swellings, activating blood circulation, and relieving pain. It is usually applied to treat various autoimmune diseases, including Rheumatoid arthritis (RA). AIM OF THE STUDY The migration of T lymphocytes plays an indispensable role in the pathogenesis of RA. Our previous studies demonstrated that modified Xianfang Huoming Yin (XFHM) could modulate the differentiation of T, B, and NK cells, and contribute to the restoration of immunologic balance. It also could downregulate the production of pro-inflammatory cytokines by regulating the activation of NF-κ B and JAK/STAT signaling pathways in the collagen-induced arthritis mouse model. In this study, we want to investigate whether XFHM has therapeutic effects on the inflammatory proliferation of rat fibroblast-like synovial cells (FLSs) by interfering with the migration of T lymphocytes in vitro experiments. MATERIALS AND METHODS High performance liquid chromatography-electrospray ionization/mass spectrometer system was used to identify the constituents of the XFHM formula. A co-culture system of rat fibroblast-like synovial cells (RSC-364 cells) and peripheral blood lymphocytes stimulated by interleukin-1 beta (IL-1β) was used as the cell model. IL-1β inhibitor (IL-1βRA) was used as a positive control medicine, and two concentrations (100 μg/mL and 250 μg/mL) of freeze-dried XFHM powder were used as intervention measure. The lymphocyte migration levels were analyzed by the Real-time xCELLigence analysis system after 24 h and 48 h of treatment. The percentage of CD3+CD4+ T cells and CD3+CD8+ T cells, and the apoptosis rate of FLSs were detected by flow cytometry. The morphology of RSC-364 cells was observed by hematoxylin-eosin staining. The protein expression of key factors for T cell differentiation and NF-κ B signaling pathway-related proteins in RSC-364 cells were examined by western-blot analysis. The migration-related cytokines levels of P-selectin, VCAM-1, and ICAM-1 in the supernatant were measured by enzyme-linked immunosorbent assay. RESULTS Twenty-one different components in XFHM were identified. The migration CI index of T cells was significantly decreased in treatment with XFHM. XFHM also could significantly downregulate the levels r of CD3+CD4+T cells and CD3+CD8+T cells that migrated to the FLSs layer. Further study found that XFHM suppresses the production of P-selectin, VCAM-1, and ICAM-1. Meanwhile, it downregulated the protein levels of T-bet, ROR γ t, IKKα/β, TRAF2, and NF-κ B p50, upregulated the expression of GATA-3 and alleviated synovial cells inflammation proliferation, contributing to the FLSs apoptosis. CONCLUSION XFHM could attenuate the inflammation of synovium by inhibiting T lymphocyte cell migration, regulating differentiation of T cells through modulating the activation of the NF-κ B signaling pathway.
Collapse
Affiliation(s)
- Dongyang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Song Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yingkai Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Pingxin Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guiyu Feng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Wei
- Department of Pharmacy, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Limin Chai
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
8
|
Subayyil AA, Basmaeil YS, Kulayb HB, Alrodayyan M, Alhaber LAA, Almanaa TN, Khatlani T. Preconditioned Chorionic Villus Mesenchymal Stem/Stromal Cells (CVMSCs) Minimize the Invasive Phenotypes of Breast Cancer Cell Line MDA231 In Vitro. Int J Mol Sci 2023; 24:ijms24119569. [PMID: 37298519 DOI: 10.3390/ijms24119569] [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: 03/05/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 06/12/2023] Open
Abstract
Among the newer choices of targeted therapies against cancer, stem cell therapy is gaining importance because of their antitumor properties. Stem cells suppress growth, metastasis, and angiogenesis, and induce apoptosis in cancer cells. In this study, we have examined the impact of the cellular component and the secretome of preconditioned and naïve placenta-derived Chorionic Villus Mesenchymal Stem Cells (CVMSCs) on the functional characteristics of the Human Breast Cancer cell line MDA231. MDA231 cells were treated with preconditioned CVMSCs and their conditioned media (CM), followed by an evaluation of their functional activities and modulation in gene and protein expression. Human Mammary Epithelial Cells (HMECs) were used as a control. CM obtained from the preconditioned CVMSCs significantly altered the proliferation of MDA231 cells, yet no change in other phenotypes, such as adhesion, migration, and invasion, were observed at various concentrations and time points tested. However, the cellular component of preconditioned CVMSCs significantly inhibited several phenotypes of MDA231 cells, including proliferation, migration, and invasion. CVMSCs-treated MDA231 cells exhibited modulation in the expression of various genes involved in apoptosis, oncogenesis, and Epithelial to Mesenchymal Transition (EMT), explaining the changes in the invasive behavior of MDA231 cells. These studies reveal that preconditioned CVMSCs may make useful candidate in a stem cell-based therapy against cancer.
Collapse
Affiliation(s)
- Abdullah Al Subayyil
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Yasser S Basmaeil
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Hayaa Bin Kulayb
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Maha Alrodayyan
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| | - Lama Abdulaziz A Alhaber
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Tanvir Khatlani
- Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia
| |
Collapse
|
9
|
Hu W, Ding R, Wang M, Huang P, Wei X, Hu X, Hu T. Side population cells derived from hUCMSCs and hPMSCs could inhibit the malignant behaviors of Tn + colorectal cancer cells from modifying their O-glycosylation status. Stem Cell Res Ther 2023; 14:145. [PMID: 37237420 DOI: 10.1186/s13287-023-03334-3] [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: 07/29/2022] [Accepted: 04/07/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Cosmc (C1GalT1C1) mutation could cause aberrant O-glycosylation and result in expression of Tn antigen on the surface of tumor cells (Tn+ cells), which is associated with the metastasis and prognosis of cancer progression. Mesenchymal stem cells (MSCs) could participate in immunoregulation, tissue damage repair, and tumor inhibition and be seen as an ideal candidate for tumor therapy due to their inherent capacity to migrate to tumor sites. However, their therapeutic effectiveness in different tumors is inconsistent and still controversial. Of note, emerging data reveal that side population (SP) cells have a stronger multilineage developmental potential than main population cells and can function as stem/progenitor cells. The effect of SP cells derived from MSCs on the biological behaviors and the O-glycosylation status of tumor cells remains unclear. METHODS SP cells were isolated from human umbilical cord MSCs (hUCMSCs) and human placenta MSCs (hPMSCs). Tn+ cells (LS174T-Tn+ and HT-29-Tn+ cells) and matching Tn- cells (LS174T-Tn- and HT-29-Tn- cells) were isolated from human colorectal cancer cell (CRC) lines LS174T and HT-29 by immune magnetic beads. The proliferation, migration, apoptosis, Tn antigen expression, and O-glycome in Tn+ and Tn- CRC cells before and after co-cultured with SP-MSCs were detected using real-time cell Analysis (RTCA), flow cytometry (FCM), and cellular O-glycome reporter/amplification (CORA), respectively. Cosmc protein and O-glycosyltransferase (T-synthase and C3GnT) activity in CRC cells were, respectively, assessed using western blotting and fluorescence method. RESULTS Both SP cells derived from hUCMSCs and hPMSCs could inhibit proliferation and migration, promote apoptosis of CRC cells, significantly reduce Tn antigen expression on Tn+ CRC cells, generate new core 1-, 2-, and 3-derived O-glycans, increase T-synthase and C3GnT activity, and elevate the levels of Cosmc and T-synthase protein. CONCLUSION SP-hUCMSCs and SP-hPMSCs could inhibit proliferation and migration and promote apoptosis of Tn+ CRC cells via increasing O-glycosyltransferase activity to modify O-glycosylation status, which further adds a new dimension to the treatment of CRC.
Collapse
Affiliation(s)
- Wen Hu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Ruisong Ding
- Department of Immunology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Mengyang Wang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Panpan Huang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Xia Wei
- Department of Immunology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Xingyou Hu
- Qingdao University, Qingdao, 266071, People's Republic of China.
| | - Tao Hu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| |
Collapse
|
10
|
Sivakumar S, Lieber S, Librizzi D, Keber C, Sommerfeld L, Finkernagel F, Roth K, Reinartz S, Bartsch JW, Graumann J, Müller‐Brüsselbach S, Müller R. Basal cell adhesion molecule promotes metastasis-associated processes in ovarian cancer. Clin Transl Med 2023; 13:e1176. [PMID: 36647260 PMCID: PMC9842900 DOI: 10.1002/ctm2.1176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Basal cell adhesion molecule (BCAM) is a laminin α5 (LAMA5) binding membrane-bound protein with a putative role in cancer. Besides full-length BCAM1, an isoform lacking most of the cytoplasmic domain (BCAM2), and a soluble form (sBCAM) of unknown function are known. In ovarian carcinoma (OC), all BCAM forms are abundant and associated with poor survival, yet BCAM's contribution to peritoneal metastatic spread remains enigmatic. METHODS Biochemical, omics-based and real-time cell assays were employed to identify the source of sBCAM and metastasis-related functions of different BCAM forms. OC cells, explanted omentum and a mouse model of peritoneal colonisation were used in loss- and gain-of-function experiments. RESULTS We identified ADAM10 as a major BCAM sheddase produced by OC cells and identified proteolytic cleavage sites proximal to the transmembrane domain. Recombinant soluble BCAM inhibited single-cell adhesion and migration identically to membrane-bound isoforms, confirming its biological activity in OC. Intriguingly, this seemingly anti-tumorigenic potential of BCAM contrasts with a novel pro-metastatic function discovered in the present study. Thus, all queried BCAM forms decreased the compactness of tumour cell spheroids by inhibiting LAMA5 - integrin β1 interactions, promoted spheroid dispersion in a three-dimensional collagen matrix, induced clearance of mesothelial cells at spheroid attachment sites in vitro and enhanced invasion of spheroids into omental tissue both ex vivo and in vivo. CONCLUSIONS Membrane-bound BCAM as well as sBCAM shed by ADAM10 act as decoys rather than signalling receptors to modulate metastasis-related functions. While BCAM appears to have tumour-suppressive effects on single cells, it promotes the dispersion of OC cell spheroids by regulating LAMA5-integrin-β1-dependent compaction and thereby facilitating invasion of metastatic target sites. As peritoneal dissemination is majorly mediated by spheroids, these findings offer an explanation for the association of BCAM with a poor clinical outcome of OC, suggesting novel therapeutic options.
Collapse
Affiliation(s)
- Suresh Sivakumar
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Sonja Lieber
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Damiano Librizzi
- Small Animal Imaging Core FacilityCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Corinna Keber
- Institute for PathologyPhilipps UniversityMarburgGermany
| | - Leah Sommerfeld
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Florian Finkernagel
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
- Bioinformatics Core FacilityCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Katrin Roth
- Cell Imaging Core FacilityCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Silke Reinartz
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | | | - Johannes Graumann
- Biomolecular Mass SpectrometryMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Institute for Translational ProteomicsPhilipps UniversityMarburgGermany
| | - Sabine Müller‐Brüsselbach
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Rolf Müller
- Department of Translational OncologyCenter for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| |
Collapse
|
11
|
Cheng Y, Ren J, Fan S, Wu P, Cong W, Lin Y, Lan S, Song S, Shao B, Dai W, Wang X, Zhang H, Xu B, Li W, Yuan X, He B, Zhang Q. Nanoparticulates reduce tumor cell migration through affinity interactions with extracellular migrasomes and retraction fibers. NANOSCALE HORIZONS 2022; 7:779-789. [PMID: 35703339 DOI: 10.1039/d2nh00067a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Nano-tumor interactions are fundamental for cancer nanotherapy, and the cross-talk of nanomedicines with the extracellular matrix (ECM) is increasingly considered essential. Here, we specifically investigate the nano-ECM interactivity using drug-free nanoparticulates (NPs) and highly metastatic cancer cells as models. We discover with surprise that NPs closely bind to specific types of ECM components, namely, retraction fibers (RFs) and migrasomes, which are located at the rear of tumor cells during their migration. This interaction is observed to alter cell morphology, limit cell motion range and change cell adhesion. Importantly, NPs are demonstrated to inhibit tumor cell removal in vitro, and their anti-metastasis potential is preliminarily confirmed in vivo. Mechanically, the NPs are found to coat and form a rigid shell on the surface of migrasomes and retraction fibers via interaction with lipid raft/caveolae substructures. In this way, NPs block the recognition, endocytosis and elimination of migrasomes by their surrounding tumor cells. Thereby, NPs interfere with the cell-ECM interaction and reduce the promotion effect of migrasomes on cell movement. Additionally, NPs trigger alteration of the expression of proteins related to cell-cell adhesion and cytoskeleton organization, which also restricts cell migration. In summary, all the findings here provide a potential target for anti-tumor metastasis nanomedicines.
Collapse
Affiliation(s)
- Yuxi Cheng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junji Ren
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shumin Fan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Peiyao Wu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wenshu Cong
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuxing Lin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shaojie Lan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Siyang Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bin Shao
- Department of Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing 100142, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
12
|
Docshin PM, Karpov AA, Mametov MV, Ivkin DY, Kostareva AA, Malashicheva AB. Mechanisms of Regenerative Potential Activation in Cardiac Mesenchymal Cells. Biomedicines 2022; 10:1283. [PMID: 35740305 PMCID: PMC9220771 DOI: 10.3390/biomedicines10061283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 11/16/2022] Open
Abstract
Recovery of the contractile function of the heart and the regeneration of the myocardium after ischemic injury are contemporary issues in regenerative medicine and cell biology. This study aimed to analyze early transcriptional events in cardiac tissue after infarction and to explore the cell population that can be isolated from myocardial tissue. We induced myocardial infarction in Wistar rats by permanent ligation of the left coronary artery and showed a change in the expression pattern of Notch-associated genes and Bmp2/Runx2 in post-MI tissues using RNA sequencing and RT-PCR. We obtained primary cardiac mesenchymal cell (CMC) cultures from postinfarction myocardium by enzymatic dissociation of tissues, which retained part of the activation stimulus and had a pronounced proliferative potential, assessed using a "xCELLigence" real-time system. Hypoxia in vitro also causes healthy CMCs to overexpress Notch-associated genes and Bmp2/Runx2. Exogenous activation of the Notch signaling pathway by lentiviral transduction of healthy CMCs resulted in a dose-dependent activation of the Runx2 transcription factor but did not affect the activity of the Bmp2 factor. Thus, the results of this study showed that acute hypoxic stress could cause short-term activation of the embryonic signaling pathways Notch and Bmp in CMCs, and this interaction is closely related to the processes of early myocardial remodeling after a heart attack. The ability to correctly modulate and control the corresponding signals in the heart can help increase the regenerative capacity of the myocardium before the formation of fibrotic conditions.
Collapse
Affiliation(s)
- Pavel M. Docshin
- Almazov National Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 St. Petersburg, Russia; (P.M.D.); (A.A.K.)
| | - Andrei A. Karpov
- Almazov National Medical Research Centre, Institute of Experimental Medicine, 194156 St. Petersburg, Russia;
- Center of Experimental Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
| | - Malik V. Mametov
- Department of Pathophysiology, Pavlov First Saint Petersburg State Medical University, 197022 St. Petersburg, Russia;
| | - Dmitry Y. Ivkin
- Center of Experimental Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
| | - Anna A. Kostareva
- Almazov National Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 St. Petersburg, Russia; (P.M.D.); (A.A.K.)
| | - Anna B. Malashicheva
- Almazov National Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 St. Petersburg, Russia; (P.M.D.); (A.A.K.)
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Science, 194064 St. Petersburg, Russia
| |
Collapse
|
13
|
Abstract
Experiments with cell co-culture systems facilitate investigation of the effects of one cell population on another, when the cells are grown in close proximity. Here we describe co-culture of Simpson-Golabi-Behmel syndrome (SGBS) adipocyte cells with the MCF-7 breast cancer cell line using the Corning® Transwell® 12-mm, 0.4-μm pore polyester membrane insert cell culture system. The SGBS adipocyte cell line, which was developed from cells taken from an infant with Simpson-Golabi-Behmel syndrome is comparable, both functionally and biochemically, to primary preadipocytes. The MCF-7 breast cancer cell line is an ER+/PR+ and HER2- line used very commonly in studies of breast malignancy. Consisting of insert supports with a permeable membrane 'floor,' which sit suspended in wells, the Corning® Transwell® co-culture system allows communication between physically separate cells cultured on the membrane and in the well beneath. This co-culture procedure described here can be applied to analyze the effects of cancer cells on the process of adipogenesis and the changes in cancer cells due to adipocyte-secreted factors.
Collapse
Affiliation(s)
| | - Martin Wabitsch
- Department of Paediatric Endocrinology and Diabetes, ULM University Hospital, Ulm, Germany
| | - Mark Christian
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
14
|
Decidua Parietalis Mesenchymal Stem/Stromal Cells and Their Secretome Diminish the Oncogenic Properties of MDA231 Cells In Vitro. Cells 2021; 10:cells10123493. [PMID: 34944000 PMCID: PMC8700435 DOI: 10.3390/cells10123493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been shown to suppress tumor growth, inhibit angiogenesis, regulate cellular signaling, and induce apoptosis in cancer cells. We have earlier reported that placenta-derived decidua parietalis mesenchymal stem/stromal cells (DPMSCs) not only retained their functional characteristics in the cancer microenvironment but also exhibited increased expression of anti-apoptotic genes, demonstrating their anti-tumor properties in the tumor setting. In this study, we have further evaluated the effects of DPMSCs on the functional outcome of human breast cancer cell line MDA231. MDA231 cells were exposed to DPMSCs, and their biological functions, including adhesion, proliferation, migration, and invasion, were evaluated. In addition, genomic and proteomic modifications of the MDA231 cell line, in response to the DPMSCs, were also evaluated. MDA231 cells exhibited a significant reduction in proliferation, migration, and invasion potential after their treatment with DPMSCs. Furthermore, DPMSC treatment diminished the angiogenic potential of MDA231 cells. DPMSC treatment modulated the expression of various pro-apoptotic as well as oncogenes in MDA231 cells. The properties of DPMSCs to inhibit the invasive characteristics of MDA231 cells demonstrate that they may be a useful candidate in a stem-cell-based therapy against cancer.
Collapse
|
15
|
Andreidesz K, Szabo A, Kovacs D, Koszegi B, Bagone Vantus V, Vamos E, Isbera M, Kalai T, Bognar Z, Kovacs K, Gallyas F. Cytostatic Effect of a Novel Mitochondria-Targeted Pyrroline Nitroxide in Human Breast Cancer Lines. Int J Mol Sci 2021; 22:ijms22169016. [PMID: 34445722 PMCID: PMC8396499 DOI: 10.3390/ijms22169016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondria have emerged as a prospective target to overcome drug resistance that limits triple-negative breast cancer therapy. A novel mitochondria-targeted compound, HO-5114, demonstrated higher cytotoxicity against human breast cancer lines than its component-derivative, Mito-CP. In this study, we examined HO-5114′s anti-neoplastic properties and its effects on mitochondrial functions in MCF7 and MDA-MB-231 human breast cancer cell lines. At a 10 µM concentration and within 24 h, the drug markedly reduced viability and elevated apoptosis in both cell lines. After seven days of exposure, even at a 75 nM concentration, HO-5114 significantly reduced invasive growth and colony formation. A 4 h treatment with 2.5 µM HO-5114 caused a massive loss of mitochondrial membrane potential, a decrease in basal and maximal respiration, and mitochondrial and glycolytic ATP production. However, reactive oxygen species production was only moderately elevated by HO-5114, indicating that oxidative stress did not significantly contribute to the drug’s anti-neoplastic effect. These data indicate that HO-5114 may have potential for use in the therapy of triple-negative breast cancer; however, the in vivo toxicity and anti-neoplastic effectiveness of the drug must be determined to confirm its potential.
Collapse
Affiliation(s)
- Kitti Andreidesz
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Aliz Szabo
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Dominika Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Balazs Koszegi
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Viola Bagone Vantus
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Eszter Vamos
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Mostafa Isbera
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pecs, 7624 Pecs, Hungary; (M.I.); (T.K.)
| | - Tamas Kalai
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pecs, 7624 Pecs, Hungary; (M.I.); (T.K.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary
| | - Zita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Krisztina Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (K.A.); (A.S.); (D.K.); (B.K.); (V.B.V.); (E.V.); (Z.B.); (K.K.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
- Correspondence: ; Tel.: +36-72-536-278
| |
Collapse
|
16
|
Zhang Q, Shi R, Bai Y, Meng L, Hu J, Zhu H, Liu T, De X, Wang S, Wang J, Xu L, Zhou G, Yin R. Meiotic nuclear divisions 1 (MND1) fuels cell cycle progression by activating a KLF6/E2F1 positive feedback loop in lung adenocarcinoma. Cancer Commun (Lond) 2021; 41:492-510. [PMID: 33734616 PMCID: PMC8211349 DOI: 10.1002/cac2.12155] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/19/2020] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
Background Considering the increase in the proportion of lung adenocarcinoma (LUAD) cases among all lung cancers and its considerable contribution to cancer‐related deaths worldwide, we sought to identify novel oncogenes to provide potential targets and facilitate a better understanding of the malignant progression of LUAD. Methods The results from the screening of transcriptome and survival analyses according to the integrated Gene Expression Omnibus (GEO) datasets and The Cancer Genome Atlas (TCGA) data were combined, and a promising risk biomarker called meiotic nuclear divisions 1 (MND1) was selectively acquired. Cell viability assays and subcutaneous xenograft models were used to validate the oncogenic role of MND1 in LUAD cell proliferation and tumor growth. A series of assays, including mass spectrometry, co‐immunoprecipitation (Co‐IP), and chromatin immunoprecipitation (ChIP), were performed to explore the underlying mechanism. Results MND1 up‐regulation was identified to be an independent risk factor for overall survival in LUAD patients evaluated by both tissue microarray staining and third party data analysis. In vivo and in vitro assays showed that MND1 promoted LUAD cell proliferation by regulating cell cycle. The results of the Co‐IP, ChIP and dual‐luciferase reporter assays validated that MND1 competitively bound to tumor suppressor Kruppel‐like factor 6 (KLF6), and thereby protecting E2F transcription factor 1 (E2F1) from KLF6‐induced transcriptional repression. Luciferase reporter and ChIP assays found that E2F1 activated MND1 transcription by binding to its promoter in a feedback manner. Conclusions MND1, KLF6, and E2F1 form a positive feedback loop to regulate cell cycle and confer DDP resistance in LUAD. MND1 is crucial for malignant progression and may be a potential therapeutic target in LUAD patients.
Collapse
Affiliation(s)
- Quanli Zhang
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China.,Department of Scientific Research, Jiangsu Cancer Hospital & the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, 210009, P. R. China
| | - Run Shi
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) München, München, Bayern, D-80539, Germany
| | - Yongkang Bai
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China
| | - Lijuan Meng
- Department of Geriatric Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210009, P. R. China
| | - Jingwen Hu
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China.,Department of Scientific Research, Jiangsu Cancer Hospital & the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, 210009, P. R. China
| | - Hongyu Zhu
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China
| | - Tongyan Liu
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China.,Department of Scientific Research, Jiangsu Cancer Hospital & the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, 210009, P. R. China
| | - Xiaomeng De
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China.,Department of Scientific Research, Jiangsu Cancer Hospital & the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, 210009, P. R. China
| | - Siwei Wang
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China.,The Fourth Clinical College of Nanjing Medical University, Nanjing, Jiangsu, 210009, P. R. China
| | - Jie Wang
- Department of Scientific Research, Jiangsu Cancer Hospital & the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing, Jiangsu, 210009, P. R. China.,Jiangsu Biobank of Clinical Resources, Nanjing, Jiangsu, 210009, P. R. China
| | - Lin Xu
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China
| | - Guoren Zhou
- Department of Oncology, Jiangsu Cancer Hospital & the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, 210009, P. R. China
| | - Rong Yin
- Department of Thoracic Surgery, the Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, 210009, P. R. China.,Jiangsu Biobank of Clinical Resources, Nanjing, Jiangsu, 210009, P. R. China
| |
Collapse
|
17
|
Lei KF, Ho YC, Huang CH, Huang CH, Pai PC. Characterization of stem cell-like property in cancer cells based on single-cell impedance measurement in a microfluidic platform. Talanta 2021; 229:122259. [PMID: 33838770 DOI: 10.1016/j.talanta.2021.122259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/01/2021] [Accepted: 02/24/2021] [Indexed: 11/20/2022]
Abstract
Investigation of stem cell-like property in cancer cells is important for the development of new therapeutic drugs targeting at malignant tumors. Currently, the standard approach for identifying cancer stem cell-like cells relies on the recognition of stem cell surface markers. However, the reliability remains controversial among biologists. In the current work, a dielectrophoretic and impedimetric hybrid microfluidic platform was developed for capturing single cells and characterizing their stem cell-like property. Single cells were captured in 20 μm trapping wells by dielectrophoretic force and their impedance spectra were measured by an impedance analyzer. The result showed that different cancer cell lines could be differentiated by impedance magnitude ranging between 2 and 20 kHz. Moreover, cancer cells and cancer stem cell-like cells could be categorized by a 2-dimensional graph of the impedance magnitudes at 2 and 20 kHz. The stem cell-like property in cancer cells was verified by stem cell surface markers and single-cell derived colony assay. Comparing with bio-chemical approach, i.e., surface markers, bio-physical approach, i.e., cell impedance, is a label-free technique to identify cancer stem cell-like cells.
Collapse
Affiliation(s)
- Kin Fong Lei
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan; Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Yu-Chen Ho
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Hao Huang
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Hao Huang
- PhD Program in Biomedical Engineering, College of Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Ping Ching Pai
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
18
|
Jiang Y, Jiang H, Wang K, Liu C, Man X, Fu Q. Hypoxia enhances the production and antitumor effect of exosomes derived from natural killer cells. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:473. [PMID: 33850870 PMCID: PMC8039676 DOI: 10.21037/atm-21-347] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Exosomes are a subgroup of extracellular vesicles that are naturally released by almost all types of cells. However, the factors that promote the capacity of natural killer (NK) cells to release exosomes are unclear. In this study, we investigated whether hypoxia can enhance the yield of NK cell-derived exosomes and improve the immunotherapeutic effects of these cells. Methods Exosomes from NK92 or NK92-hIL-15 cells were isolated from culture medium under normoxic (NK92-Exo and NK92-hIL-15-Exo) or hypoxic (hypoxic NK92-Exo and hypoxic NK92-hIL-15-Exo) conditions. NK92-Exo and hypoxic NK92-Exo were characterized by transmission electron microscopy (TEM), nanoparticle-tracking analysis (NTA), and western blot. Real-time cell assay, wound healing assay, flow cytometry, and western blot were then performed to assess cytotoxicity, cell proliferation, cell migration, apoptosis, and the expression levels of cytotoxicity-associated proteins. Results After 48 hours of hypoxic treatment, NK92-Exo exhibited significantly increased cytotoxicity, enhanced inhibition of cell proliferation, and elevated levels of molecules associated with NK cell cytotoxicity. The hypoxia-treated NK92-Exo and NK92-hIL-15-Exo showed increased expression of three functional proteins of NK cells-specifically FasL, perforin, and granzyme B-as compared with their NK92-Exo counterparts exposed to normoxia. Conclusions As an approach that supports overproduction of exosomes, hypoxic treatment of NK cells may serve as a promising therapeutic option for cancer immunotherapy.
Collapse
Affiliation(s)
- Yanan Jiang
- School of Pharmacy, Binzhou Medical University, Yantai, China.,Department of Immunology, Binzhou Medical University, Yantai, China
| | - Haiming Jiang
- Intensive Care Unit, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Kun Wang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Chunling Liu
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Xuejing Man
- Department of Ophthalmology, Yantai Yuhuangding Hospital, Yantai, China
| | - Qiang Fu
- School of Pharmacy, Binzhou Medical University, Yantai, China.,School of Medicine, University of North Carolina at Chapel Hill, North Carolina, USA.,Shandong Cellogene Pharamaceutics Co. LTD, Yantai, China
| |
Collapse
|
19
|
Thanaphongdecha P, Karinshak SE, Ittiprasert W, Mann VH, Chamgramol Y, Pairojkul C, Fox JG, Suttiprapa S, Sripa B, Brindley PJ. Infection with Helicobacter pylori Induces Epithelial to Mesenchymal Transition in Human Cholangiocytes. Pathogens 2020; 9:E971. [PMID: 33233485 PMCID: PMC7700263 DOI: 10.3390/pathogens9110971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 02/08/2023] Open
Abstract
Recent reports suggest that the East Asian liver fluke infection, caused by Opisthorchis viverrini, which is implicated in opisthorchiasis-associated cholangiocarcinoma, serves as a reservoir of Helicobacter pylori. The opisthorchiasis-affected cholangiocytes that line the intrahepatic biliary tract are considered to be the cell of origin of this malignancy. Here, we investigated interactions in vitro among human cholangiocytes, Helicobacter pylori strain NCTC 11637, and the congeneric bacillus, Helicobacter bilis. Exposure to increasing numbers of H. pylori at 0, 1, 10, 100 bacilli per cholangiocyte of the H69 cell line induced phenotypic changes including the profusion of thread-like filopodia and a loss of cell-cell contact, in a dose-dependent fashion. In parallel, following exposure to H. pylori, changes were evident in levels of mRNA expression of epithelial to mesenchymal transition (EMT)-encoding factors including snail, slug, vimentin, matrix metalloprotease, zinc finger E-box-binding homeobox, and the cancer stem cell marker CD44. Analysis to quantify cellular proliferation, migration, and invasion in real-time by both H69 cholangiocytes and CC-LP-1 line of cholangiocarcinoma cells using the xCELLigence approach and Matrigel matrix revealed that exposure to 10 H. pylori bacilli per cell stimulated migration and invasion by the cholangiocytes. In addition, 10 bacilli of H. pylori stimulated contact-independent colony establishment in soft agar. These findings support the hypothesis that infection by H. pylori contributes to the malignant transformation of the biliary epithelium.
Collapse
Affiliation(s)
- Prissadee Thanaphongdecha
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
- Tropical Disease Research Laboratory, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Shannon E. Karinshak
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| | - Wannaporn Ittiprasert
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| | - Victoria H. Mann
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| | - Yaovalux Chamgramol
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.P.)
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.P.)
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Sutas Suttiprapa
- Tropical Disease Research Laboratory, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Banchob Sripa
- Tropical Disease Research Laboratory, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (Y.C.); (C.P.)
| | - Paul J. Brindley
- Research Center for Neglected Tropical Diseases of Poverty, Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.T.); (S.E.K.); (W.I.); (V.H.M.)
| |
Collapse
|
20
|
SNAIL Transctiption factor in prostate cancer cells promotes neurite outgrowth. Biochimie 2020; 180:1-9. [PMID: 33132158 DOI: 10.1016/j.biochi.2020.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/22/2023]
Abstract
Neurite outgrowth involves reciprocal signaling interactions between tumor cells and nerves where invading tumor cells have acquired the ability to respond to pro-invasive signals within the nerve environment. Neurite outgrowth could serve as a mechanism leading to invasion of cancer cells into the nerve sheath and subsequent metastasis. Snail transcription factor can promote migration and invasion of prostate cancer cells. We hypothesized that prostate cancer cell interaction with nerve cells will be mediated by Snail expression within prostate cancer cells. For this study we utilized various prostate cancer cell lines: C4-2 non-silencing (NS, control); C4-2 Snail shRNA, (stable Snail knockdown); LNCaP Neo (empty vector control) and LNCaP Snail (stably over-expressing Snail). Cancer cell adhesion and migration towards nerve cells (snF96.2 or NS20Y) was examined by co-culture assays. Conditioned media (CM) collected from C4-2 cells was cultured with nerve cells (PC-12 or NS20Y) for 48 h followed by qualitative or quantitative neurite outgrowth assay. Our results showed that cancer cells expressing high levels of Snail (LNCaP Snail/C4-2 NS) displayed significantly higher migration adherence to nerve cells, compared to cells with lower levels of Snail (LNCaP Neo/C4-2 Snail shRNA). Additionally, LNCaP Snail or C4-2 NS (Snail-high) CM led to a higher neurite outgrowth compared to the LNCaP Neo or C4-2 Snail shRNA (Snail-low). In conclusion, Snail promotes migration and adhesion to nerve cells, as well as neurite outgrowth via secretion of soluble factors. Therefore, targeting cancer cell interaction with nerves may contribute to halting prostate cancer progression/metastasis.
Collapse
|
21
|
Collagen and fibronectin promote an aggressive cancer phenotype in breast cancer cells but drive autonomous gene expression patterns. Gene 2020; 761:145024. [PMID: 32755659 DOI: 10.1016/j.gene.2020.145024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/08/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023]
Abstract
Understanding how various pathologies of breast cancer respond to their environment may be imperative in the creation of novel therapeutic targets. Central to the organisation and behaviour of cells within the tumour microenvironment is the extracellular matrix (ECM), a meshwork of fibrous proteins and glycoproteins that directly influences cell behaviour and the bioavailability of signalling molecules. Our appreciation on how the composition of the ECM can influence cancer behaviour has evolved significantly and although we are highly cognisant of the dramatic impact the ECM can have on cancer cell behaviour, we continue to neglect this during diagnosis and treatment. In the following study, we aimed to identify how three breast cancer cell lines respond functionally and genetically to common components of the ECM. Using real time and end point assays we have identified similar patterns of behaviour among the three breast cancer cell lines in response to commonly found ECM components of the breast. Using a selected gene panel, we have been able to identify cell line specific changes in gene differentiation when breast cancer cells are in contact with these elements. Although the response of our cells to these elements differ at the genetic level, their functional responses are consistent. This work adds to the growing arguments that highlight a need for histologically assessing ECM composition of breast tumours. In particular monitoring of fibrous protein deposition at the site of malignancy could provide critical information during clinical assessment influencing disease prognosis and treatment decisions for breast cancer patients.
Collapse
|
22
|
Zhang A, Xiong Y, Xu F, Wang Z, Ma J, Zhao N, Hu T, Yi J, Zhou Y, Luan X. IL-1β enhances human placenta-derived mesenchymal stromal cells ability to mediate Th1/Th2 and Th1/CD4 +IL-10 + T cell balance and regulates its adhesion, proliferation and migration via PD-L1. Cell Immunol 2020; 352:104113. [PMID: 32331794 DOI: 10.1016/j.cellimm.2020.104113] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/11/2020] [Accepted: 04/11/2020] [Indexed: 12/17/2022]
Abstract
Human placenta-derived mesenchymal stromal cells (hPMSCs) are promising candidates for the treatment of graft-versus-host disease (GVHD), which is associated with high IL-1β levels. In this study, the effects of IL-1β and hPMSCs on each other were investigated by analyzing the proportion of Th1, Th2 and CD4+IL-10+ T cells and PD-L1 expression, as well as the adhesion, migration, and proliferation of hPMSCs. The results showed that hPMSCs decreased IL-1β levels and downregulated Th1/Th2 and Th1/CD4+IL-10+ T cells ratios in the GVHD model. The in vitro results revealed that IL-1β strengthened the hPMSCs capacity to reduce the Th1/Th2 and Th1/CD4+IL-10+ T cell ratios, inhibited the adhesion and proliferation of hPMSCs and increased PD-L1 expression on hPMSCs via the JAK and NF-κB pathways. Overall, these findings suggested that hPMSCs alleviate GVHD by decreasing IL-1β level and maintaining the balance among different T cell subsets. IL-1β enhanced the ability of hPMSCs to balance different T cell subsets and inhibited hPMSCs adhesion and proliferation by regulating PD-L1 expression via the JAK and NF-κB pathways.
Collapse
Affiliation(s)
- Aiping Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China
| | - Yanlian Xiong
- Department of Anatomy, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China
| | - Fenghuang Xu
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province 570102, China
| | - Zhuoya Wang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China
| | - Junjie Ma
- Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong Province 264000, China
| | - Nannan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China
| | - Tao Hu
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China
| | - Junzhu Yi
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China
| | - Yuming Zhou
- Laboratory Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong Province 264100, China.
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, People's Republic of China.
| |
Collapse
|
23
|
Wang M, Su Z, Amoah Barnie P. Crosstalk among colon cancer-derived exosomes, fibroblast-derived exosomes, and macrophage phenotypes in colon cancer metastasis. Int Immunopharmacol 2020; 81:106298. [PMID: 32058925 DOI: 10.1016/j.intimp.2020.106298] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 12/13/2022]
Abstract
Cellular crosstalk is an important mechanism in the pathogenesis of inflammatory disorders and cancers. One significant means by which cells communicate with each other is through the release of exosomes. Exosomes are extracellular vesicles formed by the outward budding of plasma membranes, which are then released from cells into the extracellular space. Many studies have suggested that microvesicles released by colon cancer cells initiate crosstalk and modulate the fibroblast activities and macrophage phenotypes. Interestingly, crosstalk among colon cancer cells, macrophages and cancer-associated fibroblasts maximizes the mechanical composition of the stromal extracellular matrix (ECM). Exosomes contribute to cancer cell migration and invasion, which are critical for colon cancer progression to metastasis. The majority of the studies on colorectal cancers (CRCs) have focused on developing exosomal biomarkers for the early detection and prediction of CRC prognosis. This study highlights the crosstalk among colon cancer-derived exosomes, macrophage phenotypes and fibroblasts during colon cancer metastasis.
Collapse
Affiliation(s)
- Meiyun Wang
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu Province, PR China.
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, Jiangsu Province, PR China.
| | - Prince Amoah Barnie
- International Genome Center, Jiangsu University, Zhenjiang 212013, Jiangsu Province, PR China; Department of Biomedical Sciences, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana.
| |
Collapse
|
24
|
Sharma JR, Lebeko M, Kidzeru EB, Khumalo NP, Bayat A. In Vitro and Ex Vivo Models for Functional Testing of Therapeutic Anti-scarring Drug Targets in Keloids. Adv Wound Care (New Rochelle) 2019; 8:655-670. [PMID: 31827980 PMCID: PMC6904937 DOI: 10.1089/wound.2019.1040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
Significance: Keloids are benign fibro-proliferative raised dermal lesions that spread beyond the original borders of the wound, continue to grow, rarely regress, and are the most common in pigmented individuals after an abnormal wound healing response. The current treatment failure and respective challenges involved highlighting the underlying issue that the etiopathogenesis of keloids is still not well understood. Disease models are required to better understand the disease pathogenesis. It is not possible to establish keloids in animals because of the uniqueness of this disease to human skin. To address this challenge, along these lines, non-animal reproducible models are vital in investigating molecular mechanisms of keloid pathogenesis and therapeutics development. Recent Advances: Various non-animal models have been developed to better understand the molecular mechanisms involved in keloid scarring and aid in identifying and evaluating the therapeutic potential of novel drug candidates. In this scenario, the current review aims at describing in vitro monocultures, co-cultures, organotypic cultures, and ex vivo whole skin keloid tissue organ culture models. Critical Issues and Future Directions: Current treatment options for keloids are far from securing a cure or preventing disease recurrence. Identifying universally accepted effective therapy for keloids has been hampered by the absence of appropriate disease model systems. Animal models do not accurately mimic the disease, thus non-animal model systems are pivotal in keloid research. The use of these models is essential not only for a better understanding of disease biology but also for identifying and evaluating novel drug targets.
Collapse
Affiliation(s)
- Jyoti R. Sharma
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Maribanyana Lebeko
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Elvis B. Kidzeru
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Centre for Medical Research, Institute of Medical Research and Medical Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Nonhlanhla P. Khumalo
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Plastic and Reconstructive Surgery Research, Division of Musculoskeletal & Dermatological Sciences, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
25
|
A Potent Autophagy Inhibitor (Lys05) Enhances the Impact of Ionizing Radiation on Human Lung Cancer Cells H1299. Int J Mol Sci 2019; 20:ijms20235881. [PMID: 31771188 PMCID: PMC6928878 DOI: 10.3390/ijms20235881] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy inhibition through small-molecule inhibitors is one of the approaches to increase the efficiency of radiotherapy in oncological patients. A new inhibitor-Lys05-with the potential to accumulate within lysosomes and to block autophagy was discovered a few years ago. Several studies have addressed its chemosensitizing effects but nothing is known about its impact in the context of ionizing radiation (IR). To describe its role in radiosensitization, we employed radioresistant human non-small cell lung carcinoma cells (H1299, p53-negative). Combined treatment of H1299 cells by Lys05 together with IR decreased cell survival in the clonogenic assay and real-time monitoring of cell growth more than either Lys05 or IR alone. Immunodetection of LC3 and p62/SQSTM1 indicated that autophagy was inhibited, which correlated with increased SQSTM1 and decreased BNIP3 gene expression determined by qRT-PCR. Fluorescence microscopy and flow cytometry uncovered an accumulation of lysosomes. Similarly, transmission electron microscopy demonstrated the accumulation of autophagosomes confirming the ability of Lys05 to potentiate autophagy inhibition in H1299 cells. We report here for the first time that Lys05 could be utilized in combination with IR as a promising future strategy in the eradication of lung cancer cells.
Collapse
|
26
|
Kappel S, Stokłosa P, Hauert B, Ross‐Kaschitza D, Borgström A, Baur R, Galván JA, Zlobec I, Peinelt C. TRPM4 is highly expressed in human colorectal tumor buds and contributes to proliferation, cell cycle, and invasion of colorectal cancer cells. Mol Oncol 2019; 13:2393-2405. [PMID: 31441200 PMCID: PMC6822246 DOI: 10.1002/1878-0261.12566] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 12/21/2022] Open
Abstract
Transient receptor potential melastatin-4 channel (TRPM4) dysregulation contributes to heart conditions, immune diseases, and cervical and prostate cancer. Up to now, the involvement of TRPM4 in colorectal cancer (CRC) pathophysiology remains unknown. Here, we investigated tumor tissue microarrays from 379 CRC patients and analyzed TRPM4 protein expression, tumor characteristics, and clinical outcome. High TRPM4 protein expression was associated with unfavorable tumor features characteristic for epithelial-mesenchymal transition and infiltrative growth patterns, that is, a high number of tumor buds and a low percentage in tumor border configuration. Compared to CRC cells representing early cancer stages, TRPM4 protein expression was the highest in cells representing late-stage metastatic cancer. Investigation of CRC cell line HCT116 and five CRISPR/cas9 TRPM4 knockout clones demonstrated that TRPM4 exhibited large Na+ current densities (~ 60 pA/pF). In addition, CRISPR/cas9 TRPM4 knockout clones showed a tendency toward decreased migration and invasion, cell viability, and proliferation and exhibited a shift in cell cycle when compared to HCT116. Stable overexpression of TRPM4 (TRPM4 wild-type) in two CRISPR/cas9 TRPM4 knockout clones rescued the decrease in cell viability and cell cycle shift. Stable overexpression of a nonconducting, dominant-negative TRPM4 mutant (TRPM4 D894A) did not rescue the decrease in viability or cell cycle shift. Taken together, these findings pointed to TRPM4 ion channel conductivity as the underlying mechanism for decreased viability and cell cycle shift in the TRPM4 knockout clones. Together with previous findings, our present data suggest that TRPM4 plays a versatile role in cancer cell proliferation, cell cycle, and invasion.
Collapse
Affiliation(s)
- Sven Kappel
- Institute of Biochemistry and Molecular MedicineUniversity of BernSwitzerland
| | - Paulina Stokłosa
- Institute of Biochemistry and Molecular MedicineUniversity of BernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernSwitzerland
| | - Barbara Hauert
- Institute of Biochemistry and Molecular MedicineUniversity of BernSwitzerland
| | | | - Anna Borgström
- Institute of Biochemistry and Molecular MedicineUniversity of BernSwitzerland
| | - Roland Baur
- Institute of Biochemistry and Molecular MedicineUniversity of BernSwitzerland
| | - José A. Galván
- Institute of PathologyTranslational Research UnitUniversity of BernSwitzerland
| | - Inti Zlobec
- Institute of PathologyTranslational Research UnitUniversity of BernSwitzerland
| | - Christine Peinelt
- Institute of Biochemistry and Molecular MedicineUniversity of BernSwitzerland
| |
Collapse
|
27
|
Linalool attenuates oxidative stress and mitochondrial dysfunction mediated by glutamate and NMDA toxicity. Biomed Pharmacother 2019; 118:109295. [DOI: 10.1016/j.biopha.2019.109295] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/17/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
|
28
|
Yu S, Choi HH, Kim IW, Kim TJ. Conditioned medium from asbestos-exposed fibroblasts affects proliferation and invasion of lung cancer cell lines. PLoS One 2019; 14:e0222160. [PMID: 31491033 PMCID: PMC6730856 DOI: 10.1371/journal.pone.0222160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/22/2019] [Indexed: 12/28/2022] Open
Abstract
The importance of the role of fibroblasts in cancer microenvironment is well-recognized. However, the relationship between fibroblasts and asbestos-induced lung cancer remains underexplored. To investigate the effect of the asbestos-related microenvironment on lung cancer progression, lung cancer cells (NCI-H358, Calu-3, and A549) were cultured in media derived from IMR-90 lung fibroblasts exposed to 50 mg/L asbestos (chrysotile, amosite, and crocidolite) for 24 h. The kinetics and migration of lung cancer cells in the presence of asbestos-exposed lung fibroblast media were monitored using a real-time cell analysis system. Proliferation and migration of A549 cells increased in the presence of media derived from asbestos-exposed lung fibroblasts than in the presence of media derived from normal lung fibroblasts. We observed no increase in proliferation and migration in lung cancer cells cultured in asbestos-exposed lung cancer cell medium. In contrast, increased proliferation and migration in lung cancer cells exposed to media from asbestos-exposed lung fibroblasts was observed for all types of asbestos. Media derived from lung fibroblasts exposed to other stressors, such as hydrogen peroxide and UV radiation didn't show as similar effect as asbestos exposure. An enzyme-linked immunosorbent assay (ELISA)-based cytokine array identified interleukin (IL)-6 and IL-8, which show pleiotropic regulatory effects on lung cancer cells, to be specifically produced in higher amounts by the three types of asbestos-exposed lung fibroblasts than normal lung fibroblasts. Thus, the present study demonstrated that interaction of lung fibroblasts with asbestos may support the growth and metastasis of lung cancer cells and that chrysotile exposure can lead to lung cancer similar to that caused by amphibole asbestos (amosite and crocidolite).
Collapse
Affiliation(s)
- Seunghye Yu
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Chemical Engineering, Soongsil University, Seoul, Korea
| | - Hee-Hyun Choi
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Il Won Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Korea
| | - Tae-Jung Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- * E-mail:
| |
Collapse
|
29
|
Uncoupling Traditional Functionalities of Metastasis: The Parting of Ways with Real-Time Assays. J Clin Med 2019; 8:jcm8070941. [PMID: 31261795 PMCID: PMC6678138 DOI: 10.3390/jcm8070941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
The experimental evaluation of metastasis overly focuses on the gain of migratory and invasive properties, while disregarding the contributions of cellular plasticity, extra-cellular matrix heterogeneity, niche interactions, and tissue architecture. Traditional cell-based assays often restrict the inclusion of these processes and warrant the implementation of approaches that provide an enhanced spatiotemporal resolution of the metastatic cascade. Time lapse imaging represents such an underutilized approach in cancer biology, especially in the context of disease progression. The inclusion of time lapse microscopy and microfluidic devices in routine assays has recently discerned several nuances of the metastatic cascade. Our review emphasizes that a complete comprehension of metastasis in view of evolving ideologies necessitates (i) the use of appropriate, context-specific assays and understanding their inherent limitations; (ii) cautious derivation of inferences to avoid erroneous/overestimated clinical extrapolations; (iii) corroboration between multiple assay outputs to gauge metastatic potential; and (iv) the development of protocols with improved in situ implications. We further believe that the adoption of improved quantitative approaches in these assays can generate predictive algorithms that may expedite therapeutic strategies targeting metastasis via the development of disease relevant model systems. Such approaches could potentiate the restructuring of the cancer metastasis paradigm through an emphasis on the development of next-generation real-time assays.
Collapse
|
30
|
Kocyigit-Kaymakcioglu B, Yazici SS, Tok F, Dikmen M, Engür S, Oruc-Emre EE, Iyidogan A. Synthesis and Anticancer Activity of New Hydrazide-hydrazones and Their Pd(II) Complexes. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666180816124102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background:
Hydrazones, one of the important classes of organic molecules, are pharmaceutical
agents comprising –CO-NH-N=CH- group in the structure therefore and exhibiting significant
biological activity.
Methods:
5-Chloro-N’-[(substituted)methylidene] pyrazine-2-carbohydrazide (3a-g) and their Pd(II)
complexes (4a-h) were synthesized and investigated in vitro anticancer activity on A549, Caco2 cancer
and normal 3T3 fibroblast cell lines, using the MTT assay.
Results:
Anticancer activity screening results revealed that some compounds showed remarkable cytotoxic
effect. Among them, 5-chloro-N'-[(4-hydroxyphenyl)methylidene] pyrazine-2-carbohydrazide
(3c) displayed higher cytotoxic activity against A549 cancer cell line than the reference drug cisplatin.
Conclusion:
Compound 3c showed high cytotoxic activity against A549 cancer cell line but it showed
low cytotoxic effect against normal 3T3 fibroblast cell line. Antiproliferative and antimetastatic effects
of 3c were determined by the real-time monitoring of cell proliferative system (RTCA DP). The cell
proliferation, metastatic and invasive activities of A549 cells were decreased due to increased concentration
of 3c.
Collapse
Affiliation(s)
| | - Senem Sinem Yazici
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul-34668, Turkey
| | - Fatih Tok
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul-34668, Turkey
| | - Miriş Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir-26470, Turkey
| | - Selin Engür
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir-26470, Turkey
| | - Emine Elçin Oruc-Emre
- Department of Chemistry, Faculty of Arts and Sciences, Gaziantep University, Gaziantep-27310, Turkey
| | - Aysegul Iyidogan
- Department of Chemistry, Faculty of Arts and Sciences, Gaziantep University, Gaziantep-27310, Turkey
| |
Collapse
|
31
|
Zheng XB, Liu HS, Zhang LJ, Liu XH, Zhong XL, Zhou C, Hu T, Wu XR, Hu JC, Lian L, Deng QL, Chen YF, Ke J, He XW, Wu XJ, He XS, Lan P. Engulfment and Cell Motility Protein 1 Protects Against DSS-induced Colonic Injury in Mice via Rac1 Activation. J Crohns Colitis 2019; 13:100-114. [PMID: 30219846 DOI: 10.1093/ecco-jcc/jjy133] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Mucosal healing is an emerging therapeutic goal that could result in clinical remission of inflammatory bowel disease [IBD]. We sought to determine the role of engulfment and cell motility protein 1 [ELMO1] in wound healing in vitro and in vivo and to investigate the underlying pathways. METHODS RNA transcriptome sequencing was performed to detect the expression profiles of mRNA between inflamed tissues and corresponding non-inflamed tissues of IBD patients, followed by Gene Expression Omnibus [GEO] datasets and western blot analysis. The effects of ELMO1 overexpression or knockdown on cell migration and proliferation were determined. The dependence of these effects on Rac1 was assessed using a Rac1 inhibitor [NSC23766] and a Rac1 pull-down assay. We identified the underlying pathways involved by Gene Ontology [GO] analysis. A dextran sulphate sodium [DSS]-induced colitis model was established to evaluate the role of ELMO1 in colonic mucosal healing. RESULTS ELMO1 was upregulated in inflamed tissues compared with corresponding non-inflamed tissues. ELMO1 overexpression increased cell migration in a Rac1-dependent manner. Depletion of ELMO1, or NSC23766 administration, abolished this effect. GO analysis revealed that ELMO1 overexpression preferentially affected pathways involved in cytoskeletal regulation and wound healing, which was demonstrated by enhanced F-actin staining and increased numbers of extending lamellipodia in cells overexpressing ELMO1. In DSS-induced colitis, systemic delivery of pSin-EF2-ELMO1-Pur attenuated colonic inflammation and promoted recovery from colonic injury. The protective effect of ELMO1 was dependent on Rac1 activation. CONCLUSIONS ELMO1 protects against DSS-induced colonic injury in mice through its effect on epithelial migration via Rac1 activation.
Collapse
Affiliation(s)
- Xiao-Bin Zheng
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hua-Shan Liu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Long-Juan Zhang
- Laboratory of Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xuan-Hui Liu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Li Zhong
- Joint Cardiac Surgery Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Chi Zhou
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tuo Hu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xian-Rui Wu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian-Cong Hu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lei Lian
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qi-Ling Deng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu-Feng Chen
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia Ke
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Wen He
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Jian Wu
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Sheng He
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Lan
- Department of Colorectal Surgery, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
32
|
Stolwijk JA, Wegener J. Impedance-Based Assays Along the Life Span of Adherent Mammalian Cells In Vitro: From Initial Adhesion to Cell Death. BIOANALYTICAL REVIEWS 2019. [DOI: 10.1007/11663_2019_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
33
|
Samal R, Sappa PK, Gesell Salazar M, Wenzel K, Reinke Y, Völker U, Felix SB, Hammer E, Könemann S. Global secretome analysis of resident cardiac progenitor cells from wild-type and transgenic heart failure mice: Why ambience matters. J Cell Physiol 2018; 234:10111-10122. [PMID: 30575044 DOI: 10.1002/jcp.27677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/04/2018] [Indexed: 01/08/2023]
Abstract
Resident cardiac progenitor cells (CPCs) have gained attention in cardiac regenerative medicine primarily due to their paracrine activity. In our current study we determined the role of pathological conditions such as heart failure on the autocrine-paracrine action of stem cell antigen-1 (Sca-1) expressing CPC. This comparative secretome profiling of Sca-1+ cells derived from transgenic heart failure (αMHC-cyclin-T1/Gαq overexpression [Cyc] cells) versus healthy (wild-type [Wt] cells) mice, achieved via mass-spectrometric quantification, enabled the identification of over 700 proteins. Our results demonstrate that the heart failure milieu caused a 2-fold enrichment of extracellular matrix proteins (ECM) like biglycan, versican, collagen XII, and angiogenic factors like heparan sulfate proteoglycan 2, plasminogen activator inhibitor 1 in the secretome. We further elucidated the direct influence of the secretome on the functional behavior of Sca-1 + cells via in vitro tube forming assay. Secreted factors present in the diseased milieu induced tube formation in Cyc cells (1.7-fold; p < 0.01) when compared with Wt cells after 24 hr of exposure. The presence of conditioned media moderately increased the proliferation of Cyc cells but had a more pronounced effect on Wt cells. Overall, these findings revealed global modifications in the secretory activity of adult Sca-1 + cells in the heart failure milieu. The secretion of ECM proteins and angiogenic factors, which are crucial for cardiac remodeling and recovery, was notably enriched in the supernatant of Cyc cells. Thus, during heart failure the microenvironment of Sca-1 + cells might favor angiogenesis and proliferation suggesting their potential to recover the damaged heart.
Collapse
Affiliation(s)
- Rasmita Samal
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Praveen Kumar Sappa
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,Department of Hematology and Oncology, Internal Medicine C, University Greifswald, Greifswald, Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Kristin Wenzel
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Yvonne Reinke
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Stephan Burkhard Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| | - Stephanie Könemann
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research, partner site Greifswald), Greifswald, Germany
| |
Collapse
|
34
|
Yan G, Du Q, Wei X, Miozzi J, Kang C, Wang J, Han X, Pan J, Xie H, Chen J, Zhang W. Application of Real-Time Cell Electronic Analysis System in Modern Pharmaceutical Evaluation and Analysis. Molecules 2018; 23:E3280. [PMID: 30544947 PMCID: PMC6321149 DOI: 10.3390/molecules23123280] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/11/2022] Open
Abstract
Objective: We summarized the progress of the xCELLigence real-time cell analysis (RTCA) technology application in recent years for the sake of enriching and developing the application of RTCA in the field of Chinese medicine. Background: The RTCA system is an established electronic cellular biosensor. This system uses micro-electronic biosensor technology that is confirmed for real-time, label-free, dynamic and non-offensive monitoring of cell viability, migration, growth, spreading, and proliferation. Methods: We summarized the relevant experiments and literature of RTCA technology from the principles, characteristics, applications, especially from the latest application progress. Results and conclusion: RTCA is attracting more and more attention. Now it plays an important role in drug screening, toxicology, Chinese herbal medicine and so on. It has wide application prospects in the area of modern pharmaceutical evaluation and analysis.
Collapse
Affiliation(s)
- Guojun Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Qian Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Xuchao Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jackelyn Miozzi
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - Chen Kang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Jinnv Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xinxin Han
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jinhuo Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hui Xie
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jun Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Weihua Zhang
- Beijing Body Revival Medical Technology Co., Ltd., Beijing 100088, China.
| |
Collapse
|
35
|
Zhao Q, Hong B, Liu T, Ji Y, Tang X, Gong K, Ye L, Yang Y, Zhang N. VEGI174 protein and its functional domain peptides exert antitumour effects on renal cell carcinoma. Int J Oncol 2018; 54:390-398. [PMID: 30431089 DOI: 10.3892/ijo.2018.4632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/24/2018] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI) has been identified as an anti‑angiogenic cytokine. However, the effects of VEGI174 protein, and its functional domain peptides V7 and V8, on renal cell carcinoma (RCC) remain unknown. In the present study, the protein and peptides were biosynthesised as experimental agents. The A498 and 786‑O RCC cell lines, and an established mouse xenograft model, were separately treated with VEGI174, V7 or V8. Cellular functions, including proliferation, migration and invasion, were subsequently detected. Cell migration and invasion were monitored using the xCELLigence system. Furthermore, tumour growth and mouse behaviours, including mobility, appetite and body weight, were assessed. The results demonstrated that VEGI174, V7 and V8 inhibited the proliferation, migration and invasion of A498 and 786‑O cell lines when administered at concentrations of 1 and 100 pM, 10 nM and 1 µM. The inhibitory effects exhibited dose‑ and time‑dependent antitumour activity. Furthermore, VEGI174, V7 and V8 inhibited tumour growth in A498 and 786‑O xenograft mice. In the A498 xenografts, the tumour growth inhibition (TGI) rates in the VEGI174‑, V7‑ and V8‑treated groups were 71, 20 and 31%, respectively. In the 786‑O xenografts, the TGI rates in the VEGI174‑, V7‑ and V8‑treated groups were 34, 26 and 31%, respectively. There was no significant loss in body weight and no cases of mortality were observed for all treated mice. In conclusion, VEGI174, V7 and V8 exhibited potential antitumour effects and were well tolerated in vivo. V7 and V8, as functional domain peptides of the VEGI174 protein, may be studied for the future treatment of RCC.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Baoan Hong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing 100034, P.R. China
| | - Tiezhu Liu
- Department of Urology, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163001, P.R. China
| | - Yongpeng Ji
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Xinxin Tang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing 100034, P.R. China
| | - Lin Ye
- Metastasis and Angiogenesis Research Group, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Yong Yang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Ning Zhang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| |
Collapse
|
36
|
Jelinkova P, Vesely R, Cihalova K, Hegerova D, Ananbeh HAAA, Richtera L, Smerkova K, Brtnicky M, Kynicky J, Moulick A, Adam V. Effect of arsenic (III and V) on oxidative stress parameters in resistant and susceptible Staphylococcus aureus. ENVIRONMENTAL RESEARCH 2018; 166:394-401. [PMID: 29936287 DOI: 10.1016/j.envres.2018.06.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/26/2018] [Accepted: 06/11/2018] [Indexed: 06/08/2023]
Abstract
The presented study deals with the observation of properties of methicillin-susceptible Staphylococcus aureus (MSSA) and methicillin-resistant S. aureus (MRSA) in the toxic arsenic environment and influence of arsenic on antioxidant capacity. Two forms of arsenic (As(III), As(V)) with different concentrations were used for induction of the oxidative stress in tested strains. Microbiological methods showed that the growth inhibition of MSSA was higher than that of MRSA in presence of both arsenic ions. As(III) showed 24% and 33% higher anti-microbial effects than As(V) against MSSA and MRSA respectively. A similar result was found also in the experiment of reduction of biofilm-formation. By using spectrophotometry, it was revealed that As(III) induced higher antioxidant production in both bacterial cultures. Methicillin-susceptible S. aureus produced an app. 50 mg equivalent of gallic acid (GAE/1 mg of protein) and MRSA produced an app. 15 mg of GAE/1 mg of protein. The productions of metallothionein in MSSA and MRSA were decreased up to 62.41% and 55.84% respectively in presence of As ions. Reduction of As(III) and As(V) concentrations leads to a decrease in antioxidant production and increased the formation of metallothionein. All of these changes in the results were found to be significant statistically. Taken together, these experiments proved that in comparison with MSSA, MRSA is less susceptible not only to the antimicrobial effects of antibiotics but also against effects caused by metalloids, as arsenic. Thus, it can be stated that MRSA abounds with complex defensive mechanisms, which may in the future constitute significant problem in the efficiency of antibiotics alternatives as metal ions or nanoparticles.
Collapse
Affiliation(s)
- Pavlina Jelinkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic
| | - Radek Vesely
- Department of Traumatology at the Medical Faculty, Masaryk University and Trauma Hospital of Brno, Ponavka 6, CZ-662 50 Brno, Czech Republic
| | - Kristyna Cihalova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic
| | - Dagmar Hegerova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic
| | - Hanadi Abd Alrahman Ali Ananbeh
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic
| | - Lukas Richtera
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic
| | - Kristyna Smerkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic
| | - Martin Brtnicky
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic; Department of Geology and Pedology, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic
| | - Jindrich Kynicky
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic
| | - Amitava Moulick
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| |
Collapse
|
37
|
Nikshoar MS, Khosravi S, Jahangiri M, Zandi A, Miripour ZS, Bonakdar S, Abdolahad M. Distinguishment of populated metastatic cancer cells from primary ones based on their invasion to endothelial barrier by biosensor arrays fabricated on nanoroughened poly(methyl methacrylate). Biosens Bioelectron 2018; 118:51-57. [DOI: 10.1016/j.bios.2018.07.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/08/2018] [Accepted: 07/16/2018] [Indexed: 01/15/2023]
|
38
|
Day JP, Whiteley E, Freeley M, Long A, Malacrida B, Kiely P, Baillie GS. RAB40C regulates RACK1 stability via the ubiquitin-proteasome system. Future Sci OA 2018; 4:FSO317. [PMID: 30112187 PMCID: PMC6088270 DOI: 10.4155/fsoa-2018-0022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/15/2018] [Indexed: 11/17/2022] Open
Abstract
AIM RACK1 is a multifunctional scaffolding protein that is expressed in many cellular compartments, orchestrating a number of signaling processes. RACK1 acts as a signaling hub to localize active enzymes to discrete locations; therefore tight control of RACK1 is vital to cellular homeostasis. Our aim was to identify the mechanisms responsible for RACK1 turnover and show that degradation is directed by the ubiquitin proteasome system. RESULTS Using siRNA screening, we identified RAB40C as the ubiquitin E3 ligase responsible for ubiquitination of RACK1, and that the action of RAB40C in controlling RACK1 levels is crucial to both cancer cell growth and migration of T cells. CONCLUSION Our data suggest that manipulation of RACK1 levels in this way may provide a novel strategy to explore RACK1 function.
Collapse
Affiliation(s)
- Jon P Day
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ellanor Whiteley
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Michael Freeley
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College, Dublin, D08 W9RT, Ireland
| | - Aideen Long
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College, Dublin, D08 W9RT, Ireland
| | - Beatrice Malacrida
- Materials & Surface Science Institute & Health Research Institute, University of Limerick, Limerick, Ireland
| | - Patrick Kiely
- Materials & Surface Science Institute & Health Research Institute, University of Limerick, Limerick, Ireland
| | - George S Baillie
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
39
|
Zheng XB, He XW, Zhang LJ, Qin HB, Lin XT, Liu XH, Zhou C, Liu HS, Hu T, Cheng HC, He XS, Wu XR, Chen YF, Ke J, Wu XJ, Lan P. Bone marrow-derived CXCR4-overexpressing MSCs display increased homing to intestine and ameliorate colitis-associated tumorigenesis in mice. Gastroenterol Rep (Oxf) 2018; 7:127-138. [PMID: 30976426 PMCID: PMC6454852 DOI: 10.1093/gastro/goy017] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 01/28/2018] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
Background and Objective Increasing interest has developed in the therapeutic potential of bone marrow-derived mesenchymal stem cells (MSCs) for the treatment of inflammatory bowel disease (IBD) and IBD-induced cancer. However, whether MSCs have the ability to suppress or promote tumor development remains controversial. The stromal cell-derived factor 1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4) axis is well known to play a critical role in the homing of MSCs. In this study, we aimed to evaluate the role of CXCR4-overexpressing MSCs on the tumorigenesis of IBD. Methods MSCs were transduced with lentiviral vector carrying either CXCR4 or green fluorescent protein (GFP). Chemotaxis and invasion assays were used to detect CXCR4 expression. A mouse model of colitis-associated tumorigenesis was established using azoxymethane and dextran sulfate sodium (DSS). The mice were divided into three groups and then injected with phosphate buffer saline (PBS), MSC-GFP or MSC-CXCR4. Results Compared with the mice injected with MSC-GFP, the mice injected with MSC-CXCR4 showed relieved weight loss, longer colons, lower tumor numbers and decreased tumor load; expression of pro-inflammatory cytokines decreased, and signal transducer and activator of transcription 3 (STAT3) phosphorylation level in colon tissue was down-regulated. Conclusion CXCR4-overexpressing MSCs exhibited effective anti-tumor function, which may be associated with enhanced homing to inflamed intestinal tissues.
Collapse
Affiliation(s)
- Xiao-Bin Zheng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xiao-Wen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Long-Juan Zhang
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hua-Bo Qin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xu-Tao Lin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xuan-Hui Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Chi Zhou
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hua-Shan Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Tuo Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hai-Chun Cheng
- Department of Gastrointestinal Surgery, Shenzhen Baoan Shajing People's Hospital, Guangzhou Medical University, Shenzhen, Guangdong, P.R. China
| | - Xiao-Sheng He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xian-Rui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Yu-Feng Chen
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Jia Ke
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xiao-Jian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
40
|
Toledo A, Grieger E, Karram K, Morrison H, Baader SL. Neurofibromatosis type 2 tumor suppressor protein is expressed in oligodendrocytes and regulates cell proliferation and process formation. PLoS One 2018; 13:e0196726. [PMID: 29715273 PMCID: PMC5929554 DOI: 10.1371/journal.pone.0196726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 04/18/2018] [Indexed: 12/25/2022] Open
Abstract
The neurofibromatosis type 2 (NF2) tumor suppressor protein Merlin functions as a negative regulator of cell growth and actin dynamics in different cell types amongst which Schwann cells have been extensively studied. In contrast, the presence and the role of Merlin in oligodendrocytes, the myelin forming cells within the CNS, have not been elucidated. In this work, we demonstrate that Merlin immunoreactivity was broadly distributed in the white matter throughout the central nervous system. Following Merlin expression during development in the cerebellum, Merlin could be detected in the cerebellar white matter tract at early postnatal stages as shown by its co-localization with Olig2-positive cells as well as in adult brain sections where it was aligned with myelin basic protein containing fibers. This suggests that Merlin is expressed in immature and mature oligodendrocytes. Expression levels of Merlin were low in oligodendrocytes as compared to astrocytes and neurons throughout development. Expression of Merlin in oligodendroglia was further supported by its identification in either immortalized cell lines of oligodendroglial origin or in primary oligodendrocyte cultures. In these cultures, the two main splice variants of Nf2 could be detected. Merlin was localized in clusters within the nuclei and in the cytoplasm. Overexpressing Merlin in oligodendrocyte cell lines strengthened reduced impedance in XCELLigence measurements and Ki67 stainings in cultures over time. In addition, the initiation and elongation of cellular projections were reduced by Merlin overexpression. Consistently, cell migration was retarded in scratch assays done on Nf2-transfected oligodendrocyte cell lines. These data suggest that Merlin actively modulates process outgrowth and migration in oligodendrocytes.
Collapse
Affiliation(s)
- Andrea Toledo
- Institute of Anatomy, Anatomy and Cell Biology, Bonn, Germany
- Laboratorio de Cultivo de Tejidos, Sección Biología Celular, Facultad de Ciencias, UdelaR, Montevideo, Uruguay
| | - Elena Grieger
- Institute of Anatomy, Anatomy and Cell Biology, Bonn, Germany
| | - Khalad Karram
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Helen Morrison
- Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany
| | - Stephan L. Baader
- Institute of Anatomy, Anatomy and Cell Biology, Bonn, Germany
- * E-mail:
| |
Collapse
|
41
|
Xu F, Yi J, Wang Z, Hu Y, Han C, Xue Q, Zhang X, Luan X. IL-27 regulates the adherence, proliferation, and migration of MSCs and enhances their regulatory effects on Th1 and Th2 subset generations. Immunol Res 2018; 65:903-912. [PMID: 28612255 PMCID: PMC5544780 DOI: 10.1007/s12026-017-8929-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Interleukin 27 (IL-27) regulates T cell function and is involved in inflammation. It has been reported that human placenta-derived mesenchymal stromal cells (hPMSCs) can inhibit T cell responses and attenuate inflammation reactions. However, it is unclear whether IL-27 can regulate hPMSC function. Here, we examined the effects of IL-27 upon adherence, migration, and proliferation as well as the immunomodulatory effects of hPMSCs. The results show that IL-27 receptor α chain (IL-27Rα) is expressed in hPMSCs. IL-27 at 30 ng/ml inhibited hPMSC adherence and proliferation, while the migration of hPMSCs was promoted with IL-27 at doses of 20 or 30 ng/ml, as determined with use of real-time cell analysis (RTCA). Moreover, IL-27 promoted regulatory effects of hPMSCs through enhancing Th2 and suppressing Th1 subset generation from activated T cells in human peripheral blood. IL-27 also enhanced the ability of hPMSCs to secrete IL-10 from CD4+T cells through increased expression levels of the programmed death ligand 1 (PDL1) in hPMSCs via the Janus kinase (JAK)/signal transducer and activator of transcription 1 (STAT1) signaling pathway. In conclusion, IL-27 has significant modulatory effects on adherence, proliferation, and migration of hPMSCs. IL-27 increased PDL1 expression levels in hPMSCs via the JAK/STAT1 pathway, which then enhanced the regulatory effects of hPMSCs upon Th1 and Th2 cell generations and IL-10 secretion from CD4+T cells.
Collapse
Affiliation(s)
- Fenghuang Xu
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Junzhu Yi
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Zhuoya Wang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Yejia Hu
- Department of Pathophysiology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Chunlei Han
- Department of Health Statistics, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Qun Xue
- Medical College of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Xueguang Zhang
- Medical College of Soochow University, Suzhou, Jiangsu, 215006, People's Republic of China
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China. .,Taishan Scholar Immunology Program, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China.
| |
Collapse
|
42
|
Gharaei MA, Xue Y, Mustafa K, Lie SA, Fristad I. Human dental pulp stromal cell conditioned medium alters endothelial cell behavior. Stem Cell Res Ther 2018; 9:69. [PMID: 29562913 PMCID: PMC5861606 DOI: 10.1186/s13287-018-0815-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/01/2018] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background Angiogenesis is of utmost importance for tissue regeneration and repair. Human dental pulp stromal cells (hDPSCs) possess angiogenic potential, as they secrete paracrine factors that may alter the host microenvironment. However, more insight into how hDPSCs guide endothelial cells (ECs) in a paracrine fashion is yet to be obtained. Therefore, the current study aimed to investigate the effect(s) of conditioned medium derived from hDPSCs (hDPSC-CM) on EC behavior in vitro. Methods hDPSCs were harvested from third molars scheduled for surgical removal under informed consent. The angiogenic profile of hDPSC-CM was identified using human angiogenesis antibody array and enzyme-linked immunosorbent assay (ELISA). Using real-time reverse transcription-polymerase chain reaction (RT-PCR) and ELISA, the mRNA and protein expression level of specific angiogenic biomarkers was determined in human umbilical vein endothelial cells (HUVECs) exposed to hDPSC-CM. The effect of hDPSC-CM on HUVEC attachment, proliferation and migration was evaluated by crystal violet staining, MTT, transwell migration along with real-time cell monitoring assays (xCELLigence; ACEA Biosciences, Inc.). A Matrigel assay was included to examine the influence of hDPSC-CM on HUVEC network formation. Endothelial growth medium (EGM-2) and EGM-2 supplemented with hDPSC-CM served as experimental groups, whereas endothelial basal medium (EBM-2) was set as negative control. Results A wide range of proangiogenic and antiangiogenic factors, including vascular endothelial growth factor, tissue inhibitor of metalloproteinase protein 1, plasminogen activator inhibitor (serpin E1), urokinase plasminogen activator and stromal cell-derived factor 1, was abundantly detected in hDPSC-CM by protein profiling array and ELISA. hDPSC-CM significantly accelerated the adhesion phases, from sedimentation to attachment and spreading, the proliferation rate and migration of HUVECs as shown in both endpoint assays and real-time cell analysis recordings. Furthermore, Matrigel assay demonstrated that hDPSC-CM stimulated tubulogenesis, affecting angiogenic parameters such as the number of nodes, meshes and total tube length. Conclusions The sustained proangiogenic and promaturation effects of hDPSC-CM shown in this in vitro study strongly suggest that the trophic factors released by hDPSCs are able to trigger pronounced angiogenic responses, even beyond EGM-2 considered as an optimal culture condition for ECs.
Collapse
Affiliation(s)
- M A Gharaei
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Y Xue
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - K Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - S A Lie
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - I Fristad
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway.
| |
Collapse
|
43
|
You S, Li W, Guan Y. Tunicamycin inhibits colon carcinoma growth and aggressiveness via modulation of the ERK-JNK-mediated AKT/mTOR signaling pathway. Mol Med Rep 2018; 17:4203-4212. [PMID: 29344654 PMCID: PMC5802191 DOI: 10.3892/mmr.2018.8444] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 07/05/2017] [Indexed: 12/21/2022] Open
Abstract
Epidemiology and evidence have demonstrated that colon carcinoma is one of the most common gastrointestinal tumors in the clinic. Reports have suggested that Tunicamycin significantly inhibits aggressiveness of colon carcinoma cells by promotion of apoptosis. In the present study, the inhibitory effect of tunicamycin on colon cancer cells and the potential underlying molecular mechanism was investigated. Western blotting, immunohistochemistry, apoptotic assays and immunofluorescence were used to analyze the therapeutic effects of tunicamycin on apoptosis, growth, aggressiveness and cell cycle of colon tumor cells, by downregulation of fibronectin, vimentin and E‑cadherin expression levels. In vitro experiments demonstrated that tunicamycin significantly inhibited growth, migration and invasion of colon carcinoma cells. In addition, tunicamycin administration promoted apoptosis of colon carcinoma cells via upregulation of apoptotic protease activating factor 1 and cytochrome c expression levels, which are proteins that have a role in mitochondrial apoptosis signaling. Cell cycle assays revealed that tunicamycin suppressed proliferation and arrested S phase entry of colon carcinoma cells. Mechanistic analysis demonstrated that tunicamycin reduced expression and phosphorylation levels of extracellular signal‑regulated kinase (ERK), c‑JUN N‑terminal kinase (JNK) and protein kinase B (AKT), and inhibited mammalian target of rapamycin (mTOR) expression levels in colon carcinoma cells. Endogenous overexpression of ERK inhibited tunicamycin‑mediated downregulation of JNK, AKT and mTOR expression, which further blocked tunicamycin‑mediated inhibition of growth and aggressiveness of colon carcinoma. In vivo assays revealed that tunicamycin treatment significantly inhibited tumor growth and promoted apoptosis, which led to long‑term survival of tumor‑bearing mice compared with the control group. In conclusion, these results suggested that tunicamycin may inhibit growth and aggressiveness of colon cancer via the ERK‑JNK‑mediated AKT/mTOR signaling pathway, and suggested that tunicamycin may be a potential anti‑cancer agent for colon carcinoma therapy.
Collapse
Affiliation(s)
- Shuping You
- Department of Anus and Bowel Surgery, Jingmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Weihong Li
- Department of Anus and Bowel Surgery, Jingmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Yun Guan
- Department of Anus and Bowel Surgery, Jingmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
44
|
Rodríguez-Razón CM, Yañez-Sánchez I, Ramos-Santillan VO, Velásquez-Ordóñez C, Gutiérrez-Rubio SA, García-García MR, López-Roa RI, Sánchez-Hernández PE, Daneri-Navarro A, García-Iglesias T. Adhesion, proliferation, and apoptosis in different molecular portraits of breast cancer treated with silver nanoparticles and its pathway-network analysis. Int J Nanomedicine 2018; 13:1081-1095. [PMID: 29503542 PMCID: PMC5826086 DOI: 10.2147/ijn.s152237] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Silver nanoparticles (AgNPs) have attracted considerable attention due to the variety of their applications in medicine and other sciences. AgNPs have been used in vitro for treatment of various diseases, such as hepatitis B and herpes simplex infections as well as colon, cervical, and lung cancers. In this study, we assessed the effect on proliferation, adhesion, and apoptosis in breast cancer cell lines of different molecular profiles (MCF7, HCC1954, and HCC70) exposed to AgNPs (2–9 nm). Methods Breast cancer cell lines were incubated in vitro; MTT assay was used to assess proliferation. Adhesion was determined by real-time analysis with the xCELLingence system. Propidium iodide and fluorescein isothiocyanate-Annexin V assay were used to measure apoptosis. The transcriptome was assessed by gene expression microarray and Probabilistic Graphical Model (PGM) analyses. Results The results showed a decreased adhesion in breast cancer cell lines and the control exposed to AgNPs was noted in 24 hours (p≤0.05). We observed a significant reduction in the proliferation of MCF7 and HCC70, but not in HCC1954. Apoptotic activity was seen in all cell lines exposed to AgNPs, with an apoptosis percentage of more than 60% in cancer cell lines and less than 60% in the control. PGM analysis confirmed, to some extent, the effects of AgNPs primarily on adhesion by changes in the extracellular matrix. Conclusion Exposure to AgNPs causes an antiproliferative, apoptotic, and anti-adhesive effect in breast cancer cell lines cultured in vitro. More research is needed to evaluate the potential use of AgNPs to treat different molecular profiles of breast cancer in humans.
Collapse
Affiliation(s)
- Christian M Rodríguez-Razón
- Laboratory of Immunology and Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, University of Guadalajara, Jalisco, Mexico
| | - Irinea Yañez-Sánchez
- Center for Research in Nanosciences and Nanotechnology, Department of Natural and Exact Sciences, University Center of the Valleys, University of Guadalajara, Jalisco, Mexico
| | - Vicente O Ramos-Santillan
- Laboratory of Immunology and Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, University of Guadalajara, Jalisco, Mexico
| | - Celso Velásquez-Ordóñez
- Center for Research in Nanosciences and Nanotechnology, Department of Natural and Exact Sciences, University Center of the Valleys, University of Guadalajara, Jalisco, Mexico
| | - Susan A Gutiérrez-Rubio
- Laboratory of Immunology and Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, University of Guadalajara, Jalisco, Mexico
| | - Maritza R García-García
- Department of Health Sciences, University Center of the High University of Guadalajara, Jalisco, Mexico
| | - Roció I López-Roa
- Laboratory of Immunology and Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, University of Guadalajara, Jalisco, Mexico
| | - Pedro E Sánchez-Hernández
- Laboratory of Immunology and Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, University of Guadalajara, Jalisco, Mexico
| | - Adrian Daneri-Navarro
- Laboratory of Immunology and Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, University of Guadalajara, Jalisco, Mexico
| | - Trinidad García-Iglesias
- Laboratory of Immunology and Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, University of Guadalajara, Jalisco, Mexico
| |
Collapse
|
45
|
Acland M, Mittal P, Lokman NA, Klingler-Hoffmann M, Oehler MK, Hoffmann P. Mass Spectrometry Analyses of Multicellular Tumor Spheroids. Proteomics Clin Appl 2018; 12:e1700124. [PMID: 29227035 DOI: 10.1002/prca.201700124] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/13/2017] [Indexed: 12/21/2022]
Abstract
Multicellular tumor spheroids (MCTS) are a powerful biological in vitro model, which closely mimics the 3D structure of primary avascularized tumors. Mass spectrometry (MS) has established itself as a powerful analytical tool, not only to better understand and describe the complex structure of MCTS, but also to monitor their response to cancer therapeutics. The first part of this review focuses on traditional mass spectrometry approaches with an emphasis on elucidating the molecular characteristics of these structures. Then the mass spectrometry imaging (MSI) approaches used to obtain spatially defined information from MCTS is described. Finally the analysis of primary spheroids, such as those present in ovarian cancer, and the great potential that mass spectrometry analysis of these structures has for improved understanding of cancer progression and for personalized in vitro therapeutic testing is discussed.
Collapse
Affiliation(s)
- Mitchell Acland
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Institute of Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, South Australia, Australia
| | - Parul Mittal
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Institute of Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, South Australia, Australia
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Manuela Klingler-Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Peter Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
46
|
Dowling CM, Phelan J, Callender JA, Cathcart MC, Mehigan B, McCormick P, Dalton T, Coffey JC, Newton AC, O'Sullivan J, Kiely PA. Protein kinase C beta II suppresses colorectal cancer by regulating IGF-1 mediated cell survival. Oncotarget 2018; 7:20919-33. [PMID: 26989024 PMCID: PMC4991501 DOI: 10.18632/oncotarget.8062] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/31/2016] [Indexed: 12/11/2022] Open
Abstract
Despite extensive efforts, cancer therapies directed at the Protein Kinase C (PKC) family of serine/threonine kinases have failed in clinical trials. These therapies have been directed at inhibiting PKC and have, in some cases, worsened disease outcome. Here we examine colon cancer patients and show not only that PKC Beta II is a tumour suppressor, but patients with low levels of this isozyme have significantly decreased disease free survival. Specifically, analysis of gene expression levels of all PKC genes in matched normal and cancer tissue samples from colon cancer patients revealed a striking down-regulation of the gene coding PKC Beta in the cancer tissue (n = 21). Tissue microarray analysis revealed a dramatic down-regulation of PKC Beta II protein levels in both the epithelial and stromal diseased tissue (n = 166). Of clinical significance, low levels of the protein in the normal tissue of patients is associated with a low (10%) 10 year survival compared with a much higher (60%) survival in patients with relatively high levels of the protein. Consistent with PKC Beta II levels protecting against colon cancer, overexpression of PKC Beta II in colon cancer cell lines reveals that PKC Beta II reverses transformation in cell based assays. Further to this, activation of PKC Beta II results in a dramatic downregulation of IGF-I-induced AKT, indicating a role for PKCs in regulating IGF-1 mediated cell survival. Thus, PKC Beta II is a tumour suppressor in colon cancer and low levels serve as a predictor for poor survival outcome.
Collapse
Affiliation(s)
- Catríona M Dowling
- Graduate Entry Medical School and Health Research Institute (HRI), University of Limerick, Limerick, Ireland.,Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland.,Stokes Research Institute, University of Limerick, Limerick, Ireland
| | - James Phelan
- Department of Surgery, Trinity College Dublin, Dublin, Ireland
| | - Julia A Callender
- Department of Pharmacology, University of California at San Diego, La Jolla, CA, USA
| | | | | | | | - Tara Dalton
- Stokes Research Institute, University of Limerick, Limerick, Ireland
| | - John C Coffey
- 4i Centre for Interventions in Infection, Inflammation and Immunity, Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - Alexandra C Newton
- Department of Pharmacology, University of California at San Diego, La Jolla, CA, USA
| | | | - Patrick A Kiely
- Graduate Entry Medical School and Health Research Institute (HRI), University of Limerick, Limerick, Ireland.,Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland.,Stokes Research Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
47
|
Parekh A, Das D, Das S, Dhara S, Biswas K, Mandal M, Das S. Bioimpedimetric analysis in conjunction with growth dynamics to differentiate aggressiveness of cancer cells. Sci Rep 2018; 8:783. [PMID: 29335481 PMCID: PMC5768811 DOI: 10.1038/s41598-017-18965-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/04/2017] [Indexed: 12/19/2022] Open
Abstract
Determination of cancer aggressiveness is mainly assessed in tissues by looking at the grade of cancer. There is a lack of specific method to determine aggressiveness of cancer cells in vitro. In our present work, we have proposed a bio-impedance based non-invasive method to differentiate aggressive property of two breast cancer cell lines. Real-time impedance analysis of MCF-7 (less aggressive) and MDA-MB-231 cells (more aggressive) demonstrated unique growth pattern. Detailed slope-analysis of impedance curves at different growth phases showed that MDA-MB-231 had higher proliferation rate and intrinsic resistance to cell death, when allowed to grow in nutrient and space limiting conditions. This intrinsic nature of death resistance of MDA-MB-231 was due to modulation and elongation of filopodia, which was also observed during scanning electron microscopy. Results were also similar when validated by cell cycle analysis. Additionally, wavelet based analysis was used to demonstrate that MCF-7 had lesser micromotion based cellular activity, when compared with MDA-MB-231. Combined together, we hypothesize that analysis of growth rate, death resistance and cellular energy, through bioimpedance based analysis can be used to determine and compare aggressiveness of multiple cancer cell lines. This further opens avenues for extrapolation of present work to human tumor tissue samples.
Collapse
Affiliation(s)
- Aditya Parekh
- School of Medical Science and Technology, IIT Kharagpur, West Bengal, India
| | - Debanjan Das
- Department of Electronics and Communications Engineering, DSPM IIIT, Naya Raipur, India
| | - Subhayan Das
- School of Medical Science and Technology, IIT Kharagpur, West Bengal, India
| | - Santanu Dhara
- School of Medical Science and Technology, IIT Kharagpur, West Bengal, India
| | - Karabi Biswas
- Department of Electrical Engineering, IIT Kharagpur, West Bengal, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, IIT Kharagpur, West Bengal, India.
| | - Soumen Das
- School of Medical Science and Technology, IIT Kharagpur, West Bengal, India.
| |
Collapse
|
48
|
High Sensitivity of Human Adipose Stem Cells to Differentiate into Myofibroblasts in the Presence of C. aspersa Egg Extract. Stem Cells Int 2017; 2017:9142493. [PMID: 29445405 PMCID: PMC5763135 DOI: 10.1155/2017/9142493] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 11/28/2017] [Indexed: 01/07/2023] Open
Abstract
Introduction Regeneration therapy using adipose-derived stem cells (ADSC) has been proposed in the treatment of skin aging. Myofibroblast plays a relevant role in the organization of the extracellular matrix of the damaged skin. A natural extract was derived from the eggs of the mollusk Cryptomphalus aspersa (e-CAF) that seems to play a role on skin repair. We have investigated the potential effects of e-CAF in the differentiation of ADSC. Materials and methods ADSC were cultured in the absence or presence of e-CAF (50 and 200 μg/mL) for 24 hours and 7 days. Real-time cell assay, morphological, immunofluorescence, and RT-PCR techniques were used to evaluate the cell culture and expression of αSMA, collagen I, and tropoelastin. Results e-CAF induced significant reduction in the rate of growth of ADSC from 24 hours to 7 days of culture. e-CAF also induced bigger sizes, higher levels of cytoplasmic refringence and complexity, and a more polyhedral morphological changes in the cultured ADSC. The protein and mRNA expression of αSMA was significantly increased in e-CAF-cultured ADSC. Conclusion e-CAF promotes ADSC differentiation to myofibroblasts and should be considered as a potential agent for the prevention and treatment of skin aging.
Collapse
|
49
|
Kozak J, Wdowiak P, Maciejewski R, Torres A. A guide for endometrial cancer cell lines functional assays using the measurements of electronic impedance. Cytotechnology 2017; 70:339-350. [PMID: 28988392 PMCID: PMC5809663 DOI: 10.1007/s10616-017-0149-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/22/2017] [Indexed: 12/29/2022] Open
Abstract
Endometrial cancer cell lines are critical tools to investigate the molecular mechanism of tumorigenesis using the end point cell-based assay such as proliferation, cytotoxicity, apoptosis, anoikis or migration and invasion. The proper assay optimization and performance is essential for physiologically relevant results interpretation. In this study we use label-free real-time cell analysis platform (xCELLigence) to optimize growing conditions for proliferation and migration experiments of two types of endometrial cancer cell lines HEC-1-B, HEC-1-A, KLE, and Ishikawa. Profiling of cell lines by cell index measurement in proliferation and migration experiments was performed. Our experimental approach allowed us to monitor particular stage of the cell growth, to see the relation between seeding density and dynamic cell growth as well as to choose the optimal serum concentration as chemoattractant in migration experiment. The highest rate of proliferation was shown for Ishikawa cells. The rapid pace of cellular migration was observed in case of KLE and HEC-1-B cells as compared to weak migratory activity of Ishikawa cells. The cell index that reflects the cell status characterized real-time cytological profile of each analyzed cell line. These cell profiles were crucial for better planning the classical end-point assays used in further research.
Collapse
Affiliation(s)
- Joanna Kozak
- Laboratory of Biostructure, Department of Normal Anatomy, Medical University of Lublin, 20-090, Lublin, Poland.
| | - Paulina Wdowiak
- Laboratory of Biostructure, Department of Normal Anatomy, Medical University of Lublin, 20-090, Lublin, Poland
| | - Ryszard Maciejewski
- Laboratory of Biostructure, Department of Normal Anatomy, Medical University of Lublin, 20-090, Lublin, Poland
| | - Anna Torres
- Laboratory of Biostructure, Department of Normal Anatomy, Medical University of Lublin, 20-090, Lublin, Poland
| |
Collapse
|
50
|
Bi L, Yan X, Yang Y, Qian L, Tian Y, Mao JH, Chen W. The component formula of Salvia miltiorrhiza and Panax ginseng induces apoptosis and inhibits cell invasion and migration through targeting PTEN in lung cancer cells. Oncotarget 2017; 8:101599-101613. [PMID: 29254189 PMCID: PMC5731899 DOI: 10.18632/oncotarget.21354] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022] Open
Abstract
Lung cancer still remains the leading cause of cancer-related death worldwide. It is an urgent need for development of novel therapeutic agents to improve current treatment of this disease. Here we investigate whether the effective component formula of traditional Chinese Medicine could serve as new potential therapeutic drugs to treat lung cancer. We optimize the most effective component formula of Salvia miltiorrhiza and Panax Ginseng (FMG), which is composed of Salvianolic acid A, 20(S)-Ginsenoside and Ginseng polysaccharide. We discovered that FMG selectively inhibited lung cancer cell proliferation and induced apoptosis but had no any cytotoxic effects on normal lung epithelial BEAS-2B cells. Moreover, FMG inhibited lung cancer cell migration and invasion. Mechanistically, we found that FMG significantly promoted p-PTEN expression and subsequently inhibited PI3K/AKT signaling pathway. The phosphatase activity of PTEN protein was increased after FMG bound to PTEN protein, indicating that PTEN is one of the FMG targeted proteins. In addition, FMG regulated expression of some marker proteins relevant to cell apoptosis, migration and invasion. Collectively, these results provide mechanistic insight into the anti-NSCLC of FMG by enhancing the phosphatase activity of PTEN, and suggest that FMG could be as a potential option for lung cancer treatment.
Collapse
Affiliation(s)
- Lei Bi
- School of Preclinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaojing Yan
- Changzhou Affiliated Hospital, Nanjing University of Chinese Medicine, Changzhou 213003, China
| | - Ye Yang
- School of Preclinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Qian
- School of Preclinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuan Tian
- School of Preclinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Weiping Chen
- School of Preclinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|