1
|
Zhang X, Xu Z, Chen Q, Zhou Z. Notch signaling regulates pulmonary fibrosis. Front Cell Dev Biol 2024; 12:1450038. [PMID: 39450276 PMCID: PMC11499121 DOI: 10.3389/fcell.2024.1450038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Pulmonary fibrosis is a progressive interstitial lung disease associated with aging. The pathogenesis of pulmonary fibrosis remains unclear, however, alveolar epithelial cell injury, myofibroblast activation, and extracellular matrix (ECM) accumulation are recognized as key contributors. Moreover, recent studies have implicated cellular senescence, endothelial-mesenchymal transition (EndMT), and epigenetic modifications in the pathogenesis of fibrotic diseases. Various signaling pathways regulate pulmonary fibrosis, including the TGF-β, Notch, Wnt, Hedgehog, and mTOR pathways. Among these, the TGF-β pathway is extensively studied, while the Notch pathway has emerged as a recent research focus. The Notch pathway influences the fibrotic process by modulating immune cell differentiation (e.g., macrophages, lymphocytes), inhibiting autophagy, and promoting interstitial transformation. Consequently, inhibiting Notch signaling represents a promising approach to mitigating pulmonary fibrosis. In this review, we discuss the role of Notch signaling pathway in pulmonary fibrosis, aiming to offer insights for future therapeutic investigations.
Collapse
Affiliation(s)
| | - Zhihao Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | | | | |
Collapse
|
2
|
Anees M, Gupta P, Kaur H, Kharbanda S, Singh H. Concomitant Delivery of Pirarubicin and Salinomycin Synergistically Enhanced the Efficacy of Cancer Therapy and Reduced the Risk of Cancer Relapse. AAPS PharmSciTech 2024; 25:211. [PMID: 39242397 DOI: 10.1208/s12249-024-02918-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/08/2024] [Indexed: 09/09/2024] Open
Abstract
Pirarubicin attracted considerable attention in clinical studies because of its high therapeutic efficacy and reduced toxicity in comparison with other anthracyclines. Nevertheless, ~ 30% patients undergoing PIRA treatment still experience relapse and metastasis. Clinical advancements unveiled that cancer stem cells (CSCs) residing in the tumor constitutes a major factor for such limitations and subsequently are the reason for treatment failure. Consequently, eradicating CSCs alongside bulk tumor is a crucial undertaking to attain utmost therapeutic efficacy of the treatment. Nevertheless, majority of the CSCs inhibitors currently under examination lack specificity, show unsynchronized bioavailability with other primary treatments and exhibit notable toxicity in their therapeutic applications, which is primarily attributable to their inadequate tumor-targeting capabilities. Therefore, we have developed a biodegradable polylactic acid based blend block copolymeric NPs for concomitant delivery of CSCs inhibitor Salinomycin (SAL) & chemotherapeutic drug Pirarubicin (PIRA) with an aim to improve the efficacy of treatment and prevent cancer relapse. Prepared NPs showed < 100 nm size and excellent loading with sustained release for both the drugs. Also, PIRA:SAL co-loaded NPs exhibits synergistically enhanced cytotoxicity against cancer cell as well as CSCs. Most importantly, NPs mediated co-delivery of the drugs showed complete tumor eradication, without any reoccurrence throughout the surveillance period. Additionally, NPs treatment didn't show any histopathological alteration in vital organs confirming their non-toxic nature. Altogether, present study concludes that the developed PIRA:SAL NPs have excellent efficacy for tumor regression as well as prevention of cancer relapse, hence can be used as a potential combination therapy for cancer treatment.
Collapse
Affiliation(s)
- Mohd Anees
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Priya Gupta
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Harshdeep Kaur
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Surender Kharbanda
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, 110016, India.
| |
Collapse
|
3
|
Yadav S, Shah D, Dalai P, Agrawal-Rajput R. The tale of antibiotics beyond antimicrobials: Expanding horizons. Cytokine 2023; 169:156285. [PMID: 37393846 DOI: 10.1016/j.cyto.2023.156285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/02/2023] [Accepted: 06/24/2023] [Indexed: 07/04/2023]
Abstract
Antibiotics had proved to be a godsend for mankind since their discovery. They were once the magical solution to the vexing problem of infection-related deaths. German scientist Paul Ehrlich had termed salvarsan as the silver bullet to treatsyphilis.As time passed, the magic of newly discovered silver bullets got tarnished with raging antibiotic resistance among bacteria and associated side-effects. Still, antibiotics remain the primary line of treatment for bacterial infections. Our understanding of their chemical and biological activities has increased immensely with advancement in the research field. Non-antibacterial effects of antibiotics are studied extensively to optimise their safer, broad-range use. These non-antibacterial effects could be both useful and harmful to us. Various researchers across the globe including our lab are studying the direct/indirect effects and molecular mechanisms behind these non-antibacterial effects of antibiotics. So, it is interesting for us to sum up the available literature. In this review, we have briefed the possible reason behind the non-antibacterial effects of antibiotics, owing to the endosymbiotic origin of host mitochondria. We further discuss the physiological and immunomodulatory effects of antibiotics. We then extend the review to discuss molecular mechanisms behind the plausible use of antibiotics as anticancer agents.
Collapse
Affiliation(s)
- Shivani Yadav
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Dhruvi Shah
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Parmeswar Dalai
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India.
| |
Collapse
|
4
|
Safa AR. Drug and apoptosis resistance in cancer stem cells: a puzzle with many pieces. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:850-872. [PMID: 36627897 PMCID: PMC9771762 DOI: 10.20517/cdr.2022.20] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023]
Abstract
Resistance to anticancer agents and apoptosis results in cancer relapse and is associated with cancer mortality. Substantial data have provided convincing evidence establishing that human cancers emerge from cancer stem cells (CSCs), which display self-renewal and are resistant to anticancer drugs, radiation, and apoptosis, and express enhanced epithelial to mesenchymal progression. CSCs represent a heterogeneous tumor cell population and lack specific cellular targets, which makes it a great challenge to target and eradicate them. Similarly, their close relationship with the tumor microenvironment creates greater complexity in developing novel treatment strategies targeting CSCs. Several mechanisms participate in the drug and apoptosis resistance phenotype in CSCs in various cancers. These include enhanced expression of ATP-binding cassette membrane transporters, activation of various cytoprotective and survival signaling pathways, dysregulation of stemness signaling pathways, aberrant DNA repair mechanisms, increased quiescence, autophagy, increased immune evasion, deficiency of mitochondrial-mediated apoptosis, upregulation of anti-apoptotic proteins including c-FLIP [cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein], Bcl-2 family members, inhibitors of apoptosis proteins, and PI3K/AKT signaling. Studying such mechanisms not only provides mechanistic insights into these cells that are unresponsive to drugs, but may lead to the development of targeted and effective therapeutics to eradicate CSCs. Several studies have identified promising strategies to target CSCs. These emerging strategies may help target CSC-associated drug resistance and metastasis in clinical settings. This article will review the CSCs drug and apoptosis resistance mechanisms and how to target CSCs.
Collapse
Affiliation(s)
- Ahmad R. Safa
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
5
|
Wang H, Zhang H, Zhu Y, Wu Z, Cui C, Cai F. Anticancer Mechanisms of Salinomycin in Breast Cancer and Its Clinical Applications. Front Oncol 2021; 11:654428. [PMID: 34381705 PMCID: PMC8350729 DOI: 10.3389/fonc.2021.654428] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 06/28/2021] [Indexed: 01/11/2023] Open
Abstract
Breast cancer (BC) is the most frequent cancer among women worldwide and is the leading cause of cancer-related deaths in women. Cancer cells with stem cell-like features and tumor-initiating potential contribute to drug resistance, tumor recurrence, and metastasis. To achieve better clinical outcomes, it is crucial to eradicate both bulk BC cells and breast cancer stem cells (BCSCs). Salinomycin, a monocarboxylic polyether antibiotic isolated from Streptomyces albus, can precisely kill cancer stem cells (CSCs), particularly BCSCs, by various mechanisms, including apoptosis, autophagy, and necrosis. There is increasing evidence that salinomycin can inhibit cell proliferation, invasion, and migration in BC and reverse the immune-inhibitory microenvironment to prevent tumor growth and metastasis. Therefore, salinomycin is a promising therapeutic drug for BC. In this review, we summarize established mechanisms by which salinomycin protects against BC and discuss its future clinical applications.
Collapse
Affiliation(s)
- Hui Wang
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hongyi Zhang
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yihao Zhu
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhonghang Wu
- Department of Scientific Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chunhong Cui
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China.,Department of Scientific Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
6
|
Heffer AM, Wang V, Libby RT, Feldon SE, Woeller CF, Kuriyan AE. Salinomycin inhibits proliferative vitreoretinopathy formation in a mouse model. PLoS One 2020; 15:e0243626. [PMID: 33347461 PMCID: PMC7751870 DOI: 10.1371/journal.pone.0243626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/24/2020] [Indexed: 01/22/2023] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a progressive disease that develops in a subset of patients who undergo surgery for retinal detachment repair, and results in significant vision loss. PVR is characterized by the migration of retinal pigment epithelial (RPE) cells into the vitreous cavity, where they undergo epithelial-to-mesenchymal transition and form contractile membranes within the vitreous and along the retina, resulting in recurrent retinal detachments. Currently, surgical intervention is the only treatment for PVR and there are no pharmacological agents that effectively inhibit or prevent PVR formation. Here, we show that a single intravitreal injection of the polyether ionophore salinomycin (SNC) effectively inhibits the formation of PVR in a mouse model with no evidence of retinal toxicity. After 4 weeks, fundus photography and optical coherence tomography (OCT) demonstrated development of mean PVR grade of 3.5 (SD: 1.3) in mouse eyes injected with RPE cells/DMSO (vehicle), compared to mean PVR grade of 1.6 (SD: 1.3) in eyes injected with RPE cells/SNC (p = 0.001). Additionally, immunohistochemistry analysis showed RPE cells/SNC treatment reduced both fibrotic (αSMA, FN1, Vim) and inflammatory (GFAP, CD3, CD20) markers compared to control RPE cells/DMSO treatment. Finally, qPCR analysis confirmed that Tgfβ, Tnfα, Mcp1 (inflammatory/cytokine markers), and Fn1, Col1a1 and Acta2 (fibrotic markers) were significantly attenuated in the RPE cells/SNC group compared to RPE/DMSO control. These results suggest that SNC is a potential pharmacologic agent for the prevention of PVR in humans and warrants further investigation.
Collapse
Affiliation(s)
- Alison M. Heffer
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Victor Wang
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Richard T. Libby
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Steven E. Feldon
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Collynn F. Woeller
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Ajay E. Kuriyan
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
- Retina Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, United States of America
| |
Collapse
|
7
|
Shen H, Sun CC, Kang L, Tan X, Shi P, Wang L, Liu E, Gong J. Low-dose salinomycin inhibits breast cancer metastasis by repolarizing tumor hijacked macrophages toward the M1 phenotype. Eur J Pharm Sci 2020; 157:105629. [PMID: 33176191 DOI: 10.1016/j.ejps.2020.105629] [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: 06/14/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 11/26/2022]
Abstract
Macrophages are sentinels of the immune system, which are often hijacked by tumor cells to assist tumor growth and metastasis. Herein our results showed that low dose salinomycin (SAL) in the range of 10-50 nM could efficiently induce M1 macrophage polarization in a dose- and time- dependent manner in vitro, with 30 nM SAL being optimal to generate M1-type macrophages from RAW246.7 cells. In animal study, intratumorally injected SAL (50 µg/kg) increased proportion of CD86 cells (by 28.9%), and decreased CD206 cells (by 14.2%) in transplant 4T1 tumors, in comparison with PBS group. Thus it resulted in significant regression in tumor growth (20% tumor inhibition) and pulmonary metastasis (reduced the number of metastatic nodes by 58%) in SAL group, whereas lipopolysaccharide (LPS) and paclitaxel (PTX) groups showed comparable number of metastatic lesions and volume of tumor. LPS treatment could as well lead to inflammatory reactions in tumor with SAL group, but resulted in systemic inflammation (elevated levels of IL-1α, IL-1β and TNF-α in serum), and PTX (10 μg/kg) treatment increased both types of macrophages. For the first time, we employed salinomycin below the dose of direct antitumor activity could effectively prime M1 type macrophage stimulation and regress tumor growth and metastasis.
Collapse
Affiliation(s)
- Huan Shen
- School of Chemical Engineering and Technology, State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, China
| | - Changquan Calvin Sun
- Pharmaceutical Materials Science and Engineering Laboratory, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lichun Kang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xiaoyue Tan
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Peng Shi
- School of Chemical Engineering and Technology, State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, China
| | - Lingyu Wang
- School of Chemical Engineering and Technology, State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, China
| | - Ergang Liu
- Aikedao Biotechnology Co. Ltd., Changde, Hunan, 415000, China.
| | - Junbo Gong
- School of Chemical Engineering and Technology, State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
8
|
López de Andrés J, Griñán-Lisón C, Jiménez G, Marchal JA. Cancer stem cell secretome in the tumor microenvironment: a key point for an effective personalized cancer treatment. J Hematol Oncol 2020; 13:136. [PMID: 33059744 PMCID: PMC7559894 DOI: 10.1186/s13045-020-00966-3] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) represent a tumor subpopulation responsible for tumor metastasis and resistance to chemo- and radiotherapy, ultimately leading to tumor relapse. As a consequence, the detection and eradication of this cell subpopulation represent a current challenge in oncology medicine. CSC phenotype is dependent on the tumor microenvironment (TME), which involves stem and differentiated tumor cells, as well as different cell types, such as mesenchymal stem cells, endothelial cells, fibroblasts and cells of the immune system, in addition to the extracellular matrix (ECM), different in composition to the ECM in healthy tissues. CSCs regulate multiple cancer hallmarks through the interaction with cells and ECM in their environment by secreting extracellular vesicles including exosomes, and soluble factors such as interleukins, cytokines, growth factors and other metabolites to the TME. Through these factors, CSCs generate and activate their own tumor niche by recruiting stromal cells and modulate angiogenesis, metastasis, resistance to antitumor treatments and their own maintenance by the secretion of different factors such as IL-6, VEGF and TGF-ß. Due to the strong influence of the CSC secretome on disease development, the new antitumor therapies focus on targeting these communication networks to eradicate the tumor and prevent metastasis, tumor relapse and drug resistance. This review summarizes for the first time the main components of the CSC secretome and how they mediate different tumor processes. Lastly, the relevance of the CSC secretome in the development of more precise and personalized antitumor therapies is discussed.
Collapse
Affiliation(s)
- Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain.,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain.,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain. .,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain. .,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain. .,Department of Health Sciences, University of Jaén, 23071, Jaén, Spain.
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain. .,Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100, Granada, Spain. .,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain. .,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain.
| |
Collapse
|
9
|
Zhang JY, Luo Q, Xu JR, Bai J, Mu LM, Yan Y, Duan JL, Cui YN, Su ZB, Xie Y, Lu WL. Regulating Stem Cell-Related Genes Induces the Plastic Differentiation of Cancer Stem Cells to Treat Breast Cancer. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:396-408. [PMID: 32913889 PMCID: PMC7452009 DOI: 10.1016/j.omto.2020.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/27/2020] [Indexed: 11/02/2022]
Abstract
Relapse of cancer is associated with multidirectional differentiation and unrestricted proliferative replication potential of cancer stem cells. Herein, we propose the plastic differentiation strategy for irreversible differentiation of cancer stem cells; further, salinomycin and its newly constructed functional liposomes are used to implement this strategy. Whole gene, cancer stem cell-related RNA, and protein expression analyses reveal that salinomycin induces the cancer stem cells into normal cells, dormant cells, and mature cancer cells. Besides, the results indicate that the gatekeeper is related to the inhibition of the protein kinase C (PKC) α signaling pathway. The differentiated normal or dormant cells are incorporated into normal tissue, whereas the rest are killed by chemotherapy. The findings would offer the evidence for plastic differentiation of cancer stem cells and propose a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Jing-Ying Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qian Luo
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jia-Rui Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jing Bai
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Li-Min Mu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yan Yan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jia-Lun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yi-Nuo Cui
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhan-Bo Su
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wan-Liang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
10
|
Salinomycin Treatment Specifically Inhibits Cell Proliferation of Cancer Stem Cells Revealed by Longitudinal Single Cell Tracking in Combination with Fluorescence Microscopy. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10144732] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A cell line derived from a tumor is a heterogeneous mixture of phenotypically different cells. Such cancer cell lines are used extensively in the search for new anticancer drugs and for investigating their mechanisms of action. Most studies today are population-based, implying that small subpopulations of cells, reacting differently to the potential drug go undetected. This is a problem specifically related to the most aggressive single cancer cells in a tumor as they appear to be insensitive to the drugs used today. These cells are not detected in population-based studies when developing new anticancer drugs. Thus, to get a deeper understanding of how all individual cancer cells react to chemotherapeutic drugs, longitudinal tracking of individual cells is needed. Here we have used digital holography for long time imaging and longitudinal tracking of individual JIMT-1 breast cancer cells. To gain further knowledge about the tracked cells, we combined digital holography with fluorescence microscopy. We grouped the JIMT-1 cells into different subpopulations based on expression of CD24 and E-cadherin and analyzed cell proliferation and cell migration for 72 h. We investigated how the cancer stem cell (CSC) targeting drug salinomycin affected the different subpopulations. By uniquely combining digital holography with fluorescence microscopy we show that salinomycin specifically targeted the CD24− subpopulation, i.e., the CSCs, by inhibiting cell proliferation, which was evident already after 24 h of drug treatment. We further found that after salinomycin treatment, the surviving cells were more epithelial-like due to the selection of the CD24+ cells.
Collapse
|
11
|
Heffer AM, Proaño J, Roztocil E, Phipps RP, Feldon SE, Huxlin KR, Sime PJ, Libby RT, Woeller CF, Kuriyan AE. The polyether ionophore salinomycin targets multiple cellular pathways to block proliferative vitreoretinopathy pathology. PLoS One 2019; 14:e0222596. [PMID: 31527897 PMCID: PMC6748436 DOI: 10.1371/journal.pone.0222596] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 09/02/2019] [Indexed: 11/21/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is characterized by membranes that form in the vitreous cavity and on both surfaces of the retina, which results in the formation of tractional membranes that can cause retinal detachment and intrinsic fibrosis of the retina, leading to retina foreshortening. Currently, there are no pharmacologic therapies that are effective in inhibiting or preventing PVR formation. One of the key aspects of PVR pathogenesis is retinal pigment epithelial (RPE) cell epithelial mesenchymal transition (EMT). Here we show that the polyether ionophore compound salinomycin (SNC) effectively inhibits TGFβ-induced EMT of RPE cells. SNC blocks the activation of TGFβ-induced downstream targets alpha smooth muscle actin (αSMA) and collagen 1 (Col1A1). Additionally, SNC inhibits TGFβ-induced RPE cell migration and contraction. We show that SNC functions to inhibit RPE EMT by targeting both the pTAK1/p38 and Smad2 signaling pathways upon TGFβ stimulation. Additionally, SNC is able to inhibit αSMA and Col1A1 expression in RPE cells that have already undergone TGFβ-induced EMT. Together, these results suggest that SNC could be an effective therapeutic compound in both the prevention and treatment of PVR.
Collapse
Affiliation(s)
- Alison M. Heffer
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- * E-mail: (AH); (AK); (CFW)
| | - Jacob Proaño
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Elisa Roztocil
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
| | - Richard P. Phipps
- Department of Environmental Medicine, University of Rochester, Rochester, NY, United States of America
| | - Steven E. Feldon
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Krystel R. Huxlin
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Patricia J. Sime
- Department of Medicine, University of Rochester, Rochester, NY, United States of America
| | - Richard T. Libby
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
| | - Collynn F. Woeller
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- * E-mail: (AH); (AK); (CFW)
| | - Ajay E. Kuriyan
- Flaum Eye Institute, University of Rochester, Rochester, NY, United States of America
- Center for Visual Sciences, University of Rochester, Rochester, NY, United States of America
- * E-mail: (AH); (AK); (CFW)
| |
Collapse
|
12
|
Scientific reports concerning the impact of interleukin 4, interleukin 10 and transforming growth factor β on cancer cells. Cent Eur J Immunol 2019; 44:190-200. [PMID: 31530989 PMCID: PMC6745546 DOI: 10.5114/ceji.2018.76273] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/12/2018] [Indexed: 02/07/2023] Open
Abstract
Cytokines are signalling proteins generated in most part by immune cells that have critical functions in cellular lifespan. Here we present recent data on three selected anti-inflammatory cytokines: interleukin (IL)-10, IL-4 and transforming growth factor β (TGF-β). IL-10 inhibits the synthesis of major pro-inflammatory cytokines, chemokines, and mediates anti-inflammatory reactions. IL-4 is a multifunctional cytokine which plays a crucial role in the regulation of immune responses and is involved in processes associated with development and differentiation of lymphocytes and regulation of T cell survival. Transforming TGF-β, which in normal cells or pre-cancerous cells, promotes proliferation arrest which represses tumour growth. In this review, we focus on the influence of IL-10, IL-4 and TGF-β on various types of cancer as well as potential of these selected cytokines to serve as new biomarkers which can support effective therapies for cancer patients. This article is presented based on a review of the newest research results.
Collapse
|
13
|
Zhang Y, Li F, Liu L, Jiang H, Hu H, Du X, Ge X, Cao J, Wang Y. Salinomycin triggers endoplasmic reticulum stress through ATP2A3 upregulation in PC-3 cells. BMC Cancer 2019; 19:381. [PMID: 31023247 PMCID: PMC6482559 DOI: 10.1186/s12885-019-5590-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Background Salinomycin is a monocarboxylic polyether antibiotic and is a potential chemotherapy drug. Our previous studies showed that salinomycin inhibited cell growth and targeted CSCs in prostate cancer. However, the precise target of salinomycin action is unclear. Methods In this work, we analyzed and identified differentially expressed genes (DEGs) after treatment with or without salinomycin using a gene expression microarray in vitro (PC-3 cells) and in vivo (NOD/SCID mice xenograft model generated from implanted PC-3 cells). Western blotting and immunohistochemical staining were used to analyze the expression of ATP2A3 and endoplasmic reticulum (ER) stress biomarkers. Flow cytometry was used to analyze the cell cycle, apoptosis and intracellular Ca2+ concentration. Results A significantly upregulated gene, ATPase sarcoplasmatic/endoplasmatic reticulum Ca2+ transporting 3 (ATP2A3), was successfully identified. In subsequent studies, we found that ATP2A3 overexpression could trigger ER stress and exert anti-cancer effects in PC-3 and DU145 cells. ATP2A3 was slightly expressed, but the ER stress biomarkers showed strong staining in prostate cancer tissues. We also found that salinomycin could trigger ER stress, which might be related to ATP2A3-mediated Ca2+ release in PC-3 cells. Furthermore, we found that salinomycin-triggered ER stress could promote apoptosis and thus exert anti-cancer effects in prostate cancer cells. Conclusion This study demonstrates that ATP2A3 might be one of the potential targets for salinomycin, which can inhibit Ca2+ release and trigger ER stress to exert anti-cancer effects.
Collapse
Affiliation(s)
- Yunsheng Zhang
- Clinical Research Institute, The Second Affiliated Hospital, University of South China; Clinical Research Center For Breast & Thyroid Disease Prevention In Hunan Province, Hengyang, 421001, People's Republic of China
| | - Fang Li
- College of Nursing, Hunan Polytechnic of Environment and Biology, Hengyang, 421005, People's Republic of China
| | - Luogen Liu
- Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Hongtao Jiang
- Department of Urology, The Second Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Hua Hu
- Cancer Research Institute, The Second Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Xiaobo Du
- Department of Urology, The First People's Hospital Yueyang, Yueyang, 414000, People's Republic of China
| | - Xin Ge
- Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Jingsong Cao
- Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, People's Republic of China
| | - Yi Wang
- Department of Urology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102; Clinical Research Institute, The Second Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China.
| |
Collapse
|
14
|
Dong J, Yang W, Han J, Cheng R, Guo X, Li L. Effect of dihydroartemisinin on epithelial-to-mesenchymal transition in canine mammary tumour cells. Res Vet Sci 2019; 124:240-247. [PMID: 30947110 DOI: 10.1016/j.rvsc.2019.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 03/23/2019] [Accepted: 03/24/2019] [Indexed: 01/18/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process by which epithelial cells acquire mesenchymal properties, generating metastases. Dihydroartemisinin (DHA) exhibits potent anti-cancer activities. CMTs is a potential suitable model for studies in human breast cancer research. In the present study, we separated DHA-untreated cells from CMTs cells to be a control group, the rest of the CMTs cells treated by DHA which were composed of three concentrations 5,10 as well as 20 μM. After that we cultivate the cells severally under the condition of 24, 48 and 72 h at 37 °C. CMTs cells were evaluated by Cell viability assay, Wound healing assay, Invasion assay and RT-PCR analysis for the expression of Slug, ZEB1, ZEB2 and Twist. Our results showed that DHA increased the inhibitory rate of CMTs cell and restrain TGF-β1-induced migration and invasion of cells in a time and concentration reliant manner efficaciously. Our result also show DHA inhibits the expression of Slug, ZEB1, ZEB2 and Twist at the mRNA in vitro, the media concentration DHA (10 μM) decreased the expression level of the Slug, ZEB1, ZEB2 mRNA significantly. In conclusion, DHA inhibits canine mammary tumours cells migration and invasiveness by regulating the expression of EMT-related genes.
Collapse
Affiliation(s)
- Jing Dong
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Wenhui Yang
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Jiaqi Han
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Rongjie Cheng
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Xin Guo
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China
| | - Lin Li
- The Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science &Veterinary Medicine, Shenyang Agricultural University, Shenyang 110161,China.
| |
Collapse
|
15
|
Antoszczak M. A medicinal chemistry perspective on salinomycin as a potent anticancer and anti-CSCs agent. Eur J Med Chem 2019; 164:366-377. [DOI: 10.1016/j.ejmech.2018.12.057] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 01/30/2023]
|
16
|
Versini A, Saier L, Sindikubwabo F, Müller S, Cañeque T, Rodriguez R. Chemical biology of salinomycin. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Lv X, Geng Z, Fan Z, Wang S, Pei W, Chen H. A PDMS Device Coupled with Culture Dish for In Vitro Cell Migration Assay. Appl Biochem Biotechnol 2018; 186:633-643. [PMID: 29707732 DOI: 10.1007/s12010-018-2737-z] [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: 01/19/2018] [Accepted: 03/12/2018] [Indexed: 02/02/2023]
Abstract
Cell migration and invasion are important factors during tumor progression and metastasis. Wound-healing assay and the Boyden chamber assay are efficient tools to investigate tumor development because both of them could be applied to measure cell migration rate. Therefore, a simple and integrated polydimethylsiloxane (PDMS) device was developed for cell migration assay, which could perform quantitative evaluation of cell migration behaviors, especially for the wound-healing assay. The integrated device was composed of three units, which included cell culture dish, PDMS chamber, and wound generation mold. The PDMS chamber was integrated with cell culture chamber and could perform six experiments under different conditions of stimuli simultaneously. To verify the function of this device, it was utilized to explore the tumor cell migration behaviors under different concentrations of fetal bovine serum (FBS) and transforming growth factor (TGF-β) at different time points. This device has the unique capability to create the "wound" area in parallel during cell migration assay and provides a simple and efficient platform for investigating cell migration assay in biomedical application.
Collapse
Affiliation(s)
- Xiaoqing Lv
- State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, 100083, China.,College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaoxin Geng
- State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, 100083, China. .,School of Information Engineering, Minzu University of China, Beijing, 100081, China.
| | - Zhiyuan Fan
- State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, 100083, China.,College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shicai Wang
- State Key Laboratory of crystal materials, Shandong University, Jinan, 250022, China
| | - WeiHua Pei
- State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, 100083, China
| | - Hongda Chen
- State Key Laboratory of Integrated Optoelectronics, Institute of Semiconductors, Chinese Academy of Sciences, Beijing, 100083, China
| |
Collapse
|
18
|
Li T, Liu X, Shen Q, Yang W, Huo Z, Liu Q, Jiao H, Chen J. Salinomycin exerts anti-angiogenic and anti-tumorigenic activities by inhibiting vascular endothelial growth factor receptor 2-mediated angiogenesis. Oncotarget 2018; 7:26580-92. [PMID: 27058891 PMCID: PMC5042000 DOI: 10.18632/oncotarget.8555] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 03/12/2016] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenesis targeting VEGFR2 has been an attractive strategy for cancer therapy for its role in promoting cancer growth and metastasis. However, the currently available drugs have unexpected side effects. Therefore, development of novel VEGFR2 inhibitors with less toxicity would be of great value. In this study, we describe a novel and safely VEGFR2 inhibitor, Salinomycin (Sal), which was screened from the drug libraries of Food and Drug Administration (FDA) and prohibited the binding of the ATP at its binding pocket of VEGFR2 using molecular docking model. Sal could interfere a series of VEGF-induced angiogenesis processes including proliferation, migration, and tube formation in HUVECS in vitro. Matrigel plug model demonstrated Sal strongly inhibited angiogenesis in vivo. We found that Sal significantly decreased VEGF-induced phosphorylation of VEGFR2 and its downstream STAT3 in dose- and time-dependent manner in HUVECs. Besides, Sal could directly reduce the cell viability and induce apoptosis in SGC-7901 cancer cells in vitro. Sal inhibited constitutive STAT3 activation by blocking its DNA binding and reduced various gene products including Bcl-2, Bcl-xL and VEGF both at mRNA and protein levels. Intra-peritoneal injection of Sal at doses of 3 and 5 mg/kg/day markedly suppressed human gastric cancer xenografts angiogenesis and growth without causing obvious toxicities. Taken together, Sal inhibits tumor angiogenesis and growth of gastric cancer; our results reveal unique characteristics of Sal as a promising anticancer drug candidate.
Collapse
Affiliation(s)
- Tao Li
- Department of Oncology, General Hospital of the Ningxia Medical University, Yinchuan 750004, China
| | - Xiaoxia Liu
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Qin Shen
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Wenjun Yang
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Zhenghao Huo
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Qilun Liu
- Department of Oncology, General Hospital of the Ningxia Medical University, Yinchuan 750004, China
| | - Haiyan Jiao
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| | - Jing Chen
- Department of Medical Genetic and Cell Biology, Ningxia Medical University, Yinchuan 750004, China.,Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan 750004, China
| |
Collapse
|
19
|
Kamlund S, Strand D, Janicke B, Alm K, Oredsson S. Influence of salinomycin treatment on division and movement of individual cancer cells cultured in normoxia or hypoxia evaluated with time-lapse digital holographic microscopy. Cell Cycle 2017; 16:2128-2138. [PMID: 28933990 PMCID: PMC5731424 DOI: 10.1080/15384101.2017.1380131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Most studies on new cancer drugs are based on population-derived data, where the absence of response of a small population may pass unnoticed. Thus, individual longitudinal tracking of cells is important for the future development of efficient cancer treatments. We have used digital holographic microscopy to track individual JIMT-1 human breast cancer cells and L929 mouse fibroblast cultivated in normoxia or hypoxia. In addition, JIMT-1 cells were treated with salinomycin, a cancer stem cell targeting compound. Three-day time-lapse movies were captured and individual cells were analysed with respect to cell division (cell cycle length) and cell movement. Comparing population-doubling time derived from population-based growth curves and individual cell cycle time data from time-lapse movies show that the former hide a sub-population of dividing cells. Salinomycin treatment increased the motility of cells, however, this motility did not result in an increased distant migration i.e. the cells increased their local movement. MCF-7 breast cancer cells showed similar motility behaviour as salinomycin-treated JIMT-1 cells. We suggest that combining features, such as motility and migration, can be used to distinguish cancer cells with mesenchymal (JIMT-1) and epithelial (MCF-7) features. The data clearly emphasize the importance of longitudinal cell tracking to understand the biology of individual cells under different conditions.
Collapse
Affiliation(s)
- Sofia Kamlund
- a Phase Holographic Imaging AB , Lund , Sweden.,b Department of Biology , Lund University , Lund , Sweden
| | - Daniel Strand
- c Department of Chemistry , Centre for Analysis and Synthesis, Lund University , Lund , Sweden
| | | | - Kersti Alm
- a Phase Holographic Imaging AB , Lund , Sweden
| | - Stina Oredsson
- b Department of Biology , Lund University , Lund , Sweden
| |
Collapse
|
20
|
Tveitarås MK, Reigstad I, Leiss L, Reed RK, Stuhr L. Single factors alone can induce mesenchymal-like morphology, but not promote full EMT in breast cancer cell lines with different hormone statuses. Exp Cell Res 2017; 359:257-265. [DOI: 10.1016/j.yexcr.2017.07.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/28/2017] [Accepted: 07/17/2017] [Indexed: 12/12/2022]
|
21
|
Hedrick E, Safe S. Transforming Growth Factor β/NR4A1-Inducible Breast Cancer Cell Migration and Epithelial-to-Mesenchymal Transition Is p38α (Mitogen-Activated Protein Kinase 14) Dependent. Mol Cell Biol 2017; 37:e00306-17. [PMID: 28674186 PMCID: PMC5574050 DOI: 10.1128/mcb.00306-17] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor β (TGF-β)-induced migration of triple-negative breast cancer (TNBC) cells is dependent on nuclear export of the orphan receptor NR4A1, which plays a role in proteasome-dependent degradation of SMAD7. In this study, we show that TGF-β induces p38α (mitogen-activated protein kinase 14 [MAPK14]), which in turn phosphorylates NR4A1, resulting in nuclear export of the receptor. TGF-β/p38α and NR4A1 also play essential roles in the induction of epithelial-to-mesenchymal transition (EMT) and induction of β-catenin in TNBC cells, and these TGF-β-induced responses and nuclear export of NR4A1 are blocked by NR4A1 antagonists, the p38 inhibitor SB202190, and kinase-dead [p38(KD)] and dominant-negative [p38(DN)] forms of p38α. Inhibition of NR4A1 nuclear export results in nuclear export of TGF-β-induced β-catenin, which then undergoes proteasome-dependent degradation. TGF-β-induced β-catenin also regulates NR4A1 expression through formation of the β-catenin-TCF-3/TCF-4/LEF-1 complex on the NR4A1 promoter. Thus, TGF-β-induced nuclear export of NR4A1 in TNBC cells plays an essential role in cell migration, SMAD7 degradation, EMT, and induction of β-catenin, and all of these pathways are inhibited by bis-indole-derived NR4A1 antagonists that inhibit nuclear export of the receptor and thereby block TGF-β-induced migration and EMT.
Collapse
Affiliation(s)
- Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
22
|
Dewangan J, Srivastava S, Rath SK. Salinomycin: A new paradigm in cancer therapy. Tumour Biol 2017; 39:1010428317695035. [PMID: 28349817 DOI: 10.1177/1010428317695035] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The primary hurdle in the treatment of cancer is acquisition of resistance by the tumor cells toward multiple drugs and selectively targeting the cancer stem cells. This problem was overcome by the chemotherapeutic property of recently discovered drug salinomycin. Exact mechanism of action of salinomycin is not yet known, but there are multiple pathways by which salinomycin inhibits tumor growth. Salinomycin decreases the expression of adenosine triphosphate-binding cassette transporter in multidrug resistance cells and interferes with Akt signaling pathway, Wnt/β-catenin, Hedgehog, and Notch pathways of cancer progression. Salinomycin selectively targets cancer stem cells. The potential of salinomycin to eliminate both cancer stem cells and therapy-resistant cancer cells may characterize the compound as a novel and an efficient chemotherapeutic drug.
Collapse
Affiliation(s)
- Jayant Dewangan
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sonal Srivastava
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Srikanta Kumar Rath
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
23
|
David JM, Dominguez C, Palena C. Pharmacological and immunological targeting of tumor mesenchymalization. Pharmacol Ther 2016; 170:212-225. [PMID: 27916651 DOI: 10.1016/j.pharmthera.2016.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Controlling the spread of carcinoma cells to distant organs is the foremost challenge in cancer treatment, as metastatic disease is generally resistant to therapy and is ultimately incurable for the majority of patients. The plasticity of tumor cell phenotype, in which the behaviors and functions of individual tumor cells differ markedly depending upon intrinsic and extrinsic factors, is now known to be a central mechanism in cancer progression. Our expanding knowledge of epithelial and mesenchymal phenotypic states in tumor cells, and the dynamic nature of the transitions between these phenotypes has created new opportunities to intervene to better control the behavior of tumor cells. There are now a variety of innovative pharmacological approaches to preferentially target tumor cells that have acquired mesenchymal features, including cytotoxic agents that directly kill these cells, and inhibitors that block or revert the process of mesenchymalization. Furthermore, novel immunological strategies have been developed to engage the immune system in seeking out and destroying mesenchymalized tumor cells. This review highlights the relevance of phenotypic plasticity in tumor biology, and discusses recently developed pharmacological and immunological means of targeting this phenomenon.
Collapse
Affiliation(s)
- Justin M David
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Charli Dominguez
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
24
|
Lu X, Guo H, Chen X, Xiao J, Zou Y, Wang W, Chen Q. Effect of RhoC on the epithelial-mesenchymal transition process induced by TGF-β1 in lung adenocarcinoma cells. Oncol Rep 2016; 36:3105-3112. [PMID: 27748883 PMCID: PMC5112615 DOI: 10.3892/or.2016.5146] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/31/2016] [Indexed: 12/30/2022] Open
Abstract
According to recent research, Ras homolog gene family member C (RhoC) is confirmed to have a powerful regulatory effect on cell motility mediated by the cytoskeleton, and this process is closely associated with tumor invasion and metastasis. In addition, the epithelial-mesenchymal transition (EMT) process which causes cytoskeleton rearrangement, also plays a pivotal role in tumor invasion and metastasis.Consequently, in the present study, we aimed to ascertain whether RhoC has an effect on the EMT process induced by TGF-β1 in lung adenocarcinoma cells and whether RhoC promotes tumor invasion by mediating the occurrence of EMT. Based on the findings, we demonstrated that RhoC was an essential mediator of the EMT process in lung adenocarcinoma cell line A549 which was evaluated by observing the morphological characteristics of the cells and by assessing the expression levels of two EMT marker proteins: E-cadherin and vimentin. During the process of EMT in the A549 cells induced by TGF-β1 (5 ng/ml), upregulated RhoC protein and RhoC activity were detected, which was associated with the enhanced invasive capability of the cells in vitro. Conversely, downregulation of the expression of RhoC by shRNA markedly impeded EMT progression as well as the invasion of A549 cells. Our results may provide a novel target towards the prevention of metastasis in advanced lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaoxiao Lu
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Honglan Guo
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Xi Chen
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Jian Xiao
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Yong Zou
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Wei Wang
- Department of Nephrology Medicine, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Qiong Chen
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|