1
|
Rostom MM, El-Zohairy MA, Marzouk MA, Berger MR, Schols D, Assal RA, Mandour YM, Adwan H, Zlotos DP. N-[4-(Benzyloxy)-3-methoxybenzyl)]adamantane-1-amine (DZH2), a dual CCR5 and CXCR4 inhibitor as a potential agent against triple negative breast cancer. Arch Pharm (Weinheim) 2024:e2400146. [PMID: 39468982 DOI: 10.1002/ardp.202400146] [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: 02/24/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024]
Abstract
DZH2, a dual inhibitor of the chemokine receptors CCR5 and CXCR4, was discovered from virtual screening for CCR5 antagonists. In specific Ca2+ chemokine signaling assays, DZH2 displayed low micromolar IC50 values at both chemokine receptors. Its binding to intracellular allosteric binding sites of CCR5 and CXCR4 was confirmed by MD simulations and binding free-energy calculations. DZH2 is superior to the CCR5 antagonist maraviroc in terms of its inhibitory activity on the growth of two breast cancer cell lines. In MCF7 and MDA-MB-231 cells, DZH2 was a >100-fold more potent inhibitor of cell viability compared to maraviroc. DZH2 (6.7 µM) reduced migration of MDA-MB-231 cells to 4% compared to 50% inhibition of migration caused by maraviroc (780 µM). Also, DZH2 was a significantly more potent inhibitor of colony formation in MDA-MB-231 cells than maraviroc. In MCF10 cells, DZH2 caused no alteration in the gene expression with respect to cellular pathways mediating cell death, indicating its selectivity to breast cancer cells.
Collapse
Affiliation(s)
- Monica M Rostom
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Mariam A El-Zohairy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Mohamed A Marzouk
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Reem A Assal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Yasmine M Mandour
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
| | - Hassan Adwan
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Darius P Zlotos
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| |
Collapse
|
2
|
CHO EUNHAN, STAMPLEY JAMES, WALL RACHEL, MATTHEWS RACHEL, ZUNICA ELIZABETH, BROWN JUSTINC, JOHANNSEN NEILM, IRVING BRIANA, SPIELMANN GUILLAUME. Acute Exercise Increases NK Cell Mitochondrial Respiration and Cytotoxicity against Triple-Negative Breast Cancer Cells under Hypoxic Conditions. Med Sci Sports Exerc 2023; 55:2132-2142. [PMID: 37436930 PMCID: PMC10662621 DOI: 10.1249/mss.0000000000003250] [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] [Indexed: 07/14/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is an aggressive, highly metastatic malignancy with high recurrence rates. Hypoxia is a hallmark of the TNBC tumor microenvironment, which promotes tumor growth while impairing natural killer (NK) cell cytotoxic functions. Although acute exercise improves NK cell function under normoxic conditions, the effect of exercise on NK cell cytotoxic functions under hypoxic conditions mimicking O 2 tensions observed in solid tumors is unknown. METHODS The cytotoxic functions of resting and postexercise NK cells isolated from thirteen young inactive healthy women were assessed against breast cancer cells expressing different levels of hormone receptors (MCF-7 and MDA-MB-231) under normoxic and hypoxic conditions. Mitochondrial respiration and H 2 O 2 efflux rates of the TNBC-activated NK cells were assessed via high-resolution respirometry. RESULTS Under hypoxia, postexercise NK cells exhibited greater killing of TNBC than resting NK cells. Further, postexercise NK cells were more likely to kill TNBC under hypoxia than normoxic conditions. In addition, mitochondrial respiration associated with oxidative (OXPHOS) capacity of TNBC-activated NK cells was greater in postexercise cells than resting cells under normoxia, but not under hypoxia. Finally, acute exercise was associated with reduced mitochondrial H 2 O 2 efflux by NK cells in both conditions. CONCLUSIONS Together, we present crucial interrelationships between hypoxia and exercise-induced changes in NK cell functions against TNBC cells. By modulating their mitochondrial bioenergetic functions, we postulate that acute exercise improves NK cell function under hypoxic conditions. Specifically, NK cell O 2 and H 2 O 2 flow (pmol·s -1 ·million NK cells -1 ) changes in response to 30-min cycling suggest that exercise primes NK cell tumor killing by reducing mitochondrial oxidative stress and, thus, rescuing their function when exposed to harsh hypoxic environments as observed in the microenvironment of breast solid tumors.
Collapse
Affiliation(s)
- EUNHAN CHO
- School of Kinesiology, Louisiana State University, Baton Rouge, LA
| | - JAMES STAMPLEY
- School of Kinesiology, Louisiana State University, Baton Rouge, LA
| | - RACHEL WALL
- School of Kinesiology, Louisiana State University, Baton Rouge, LA
| | - RACHEL MATTHEWS
- School of Kinesiology, Louisiana State University, Baton Rouge, LA
| | | | | | - NEIL M. JOHANNSEN
- School of Kinesiology, Louisiana State University, Baton Rouge, LA
- Pennington Biomedical Research Center, Baton Rouge, LA
| | - BRIAN A. IRVING
- School of Kinesiology, Louisiana State University, Baton Rouge, LA
- Pennington Biomedical Research Center, Baton Rouge, LA
| | - GUILLAUME SPIELMANN
- School of Kinesiology, Louisiana State University, Baton Rouge, LA
- Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
3
|
Organotropism of breast cancer metastasis: A comprehensive approach to the shared gene network. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
4
|
Effects of the Ethanol and Ethyl Acetate Extracts of Terminalia chebula Retz. on Proliferation, Migration, and HIF-1α and CXCR-4 Expression in MCF-7 Cells: an In Vitro Study. Appl Biochem Biotechnol 2022; 195:3327-3344. [PMID: 36585552 DOI: 10.1007/s12010-022-04301-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 01/01/2023]
Abstract
Over recent years, much attention has been devoted to the field of screening natural products and/or their novel structures because of reversing cancer progression. The current research work was intended to explore the cytotoxic activity of ethanol and ethyl acetate extracts of dried fruit of Terminalia chebula Retz. (T. chebula) in MCF-7 cell line. High-performance thin-layer chromatographic (HPTLC) method and Folin-Ciocalteu colorimetric techniques were performed. Anti-proliferative activities of T. chebula fruit extracts on the MCF-7 cell line were evaluated using MTT assay. Effects of both extracts on the migration of MCF-7 cells and the size of MCF-7-derived spheroids were also evaluated. Moreover, antioxidant properties were measured by DPPH and FRAP methods. Western blotting was used to measure the HIF-1α and CXCR-4 protein levels. Chebulagic acid, gallic acid, chebulinic acid, and ellagic acid were found as major compounds in both extracts. The total phenolic contents based on gallic acid equivalent (GAE) in the ethanol and ethyl acetate extracts of T. chebula were found to be 453.68 ± 0.31 and 495.12 ± 0.43 mg GAE/g dry weight of the extract, respectively. Both extracts exerted a significant dose- and time-dependent cytotoxicity effect on MCF-7 cells. They also had a marked negative effect on the average size of MCF-7-derived spheroids and their migration rate. None of the extracts exhibited stronger antioxidant activities than vitamin C. Furthermore, both extracts at a concentration of 125 µg/ml could meaningfully decrease the expression levels of HIF-1α and CXCR-4 in MCF-7 cells. These data represent that T. chebula may be a valuable medicinal resource in the regulation of breast cancer proliferation, growth, and metastasis.
Collapse
|
5
|
Anticancer activity of herbal formula Jisilhaebaekgyeji-Tang against human breast cancer cells and its mechanism. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00271-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
6
|
Hu X, Wang X, Liu Q, Wu J, Zhang H, Khalique A, Sun Z, Chen R, Wei J, Li H, Kong D, Zhuang J, Yan X, Huang X. Nanozyme-Powered Giant Unilamellar Vesicles for Mimicry and Modulation of Intracellular Oxidative Stress. ACS APPLIED MATERIALS & INTERFACES 2021; 13:21087-21096. [PMID: 33908764 DOI: 10.1021/acsami.1c05512] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The bottom-up construction of enzyme-based artificial cells is generating increasing interest, but achieving artificial cells for "all artificial modules" remains challenging in synthetic biology. Here, we introduce a fully synthetic cell system by integration of biomimetic nanozymes into giant unilamellar vesicles (GUVs). To mimic native peroxidase for free radical generation by taking advantage of Fenton catalysis reactions, we designed and prepared a de novo artificial nanozyme composed of ferritin heavy-chain scaffold protein and catalytic Fe3O4 nanoparticles as the active center. As two examples in bioapplications, we showed this nanozyme-powered GUV system not only mimics intracellular oxidative stress pathways but also induces tumor cell death by sensing and responding to external chemical signals. Specifically, we recreated intracellular biochemical events, including DNA damage and lipid peroxidation, in the compartmentalized GUVs by taking advantage of nanozyme induction of defined catalytic reactions. Additionally, the GUV system also actively induced DNA double-strand breakage and lipid damage of tumor cells, in response to the high expression of H2O2 within the tumor microenvironment. This concept-of-proof study offers a promising option for defining catalysis in biological systems and gives new insights into the de novo creation of artificial cells in a fully synthetic manner.
Collapse
Affiliation(s)
- Xueyan Hu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xinyue Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Qiqi Liu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Jin Wu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- College of Medicine, Nankai University, Tianjin 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Haoqi Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- College of Medicine, Nankai University, Tianjin 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Anila Khalique
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Zhiyuan Sun
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Rui Chen
- School of Materials Science and Engineering, Nankai University, Tianjin 300350, China
| | - Jing Wei
- Beijing Key Laboratory of Construction-Tailorable Advanced Functional Materials and Green Applications, Experimental Center of Advanced Materials School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Hongbo Li
- Beijing Key Laboratory of Construction-Tailorable Advanced Functional Materials and Green Applications, Experimental Center of Advanced Materials School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Jie Zhuang
- College of Medicine, Nankai University, Tianjin 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiyun Yan
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
- CAS Engineering Laboratory for Nanozymes, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
7
|
Asiry S, Kim G, Filippou PS, Sanchez LR, Entenberg D, Marks DK, Oktay MH, Karagiannis GS. The Cancer Cell Dissemination Machinery as an Immunosuppressive Niche: A New Obstacle Towards the Era of Cancer Immunotherapy. Front Immunol 2021; 12:654877. [PMID: 33927723 PMCID: PMC8076861 DOI: 10.3389/fimmu.2021.654877] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Although cancer immunotherapy has resulted in unpreceded survival benefits to subsets of oncology patients, accumulating evidence from preclinical animal models suggests that the immunosuppressive tumor microenvironment remains a detrimental factor limiting benefit for many patient subgroups. Recent efforts on lymphocyte-mediated immunotherapies are primarily focused on eliminating cancer foci at primary and metastatic sites, but few studies have investigated the impact of these therapies on the highly complex process of cancer cell dissemination. The metastatic cascade involves the directional streaming of invasive/migratory tumor cells toward specialized blood vessel intravasation gateways, called TMEM doorways, to the peripheral circulation. Importantly, this process occurs under the auspices of a specialized tumor microenvironment, herewith referred to as "Dissemination Trajectory", which is supported by an ample array of tumor-associated macrophages (TAMs), skewed towards an M2-like polarization spectrum, and which is also vital for providing microenvironmental cues for cancer cell invasion, migration and stemness. Based on pre-existing evidence from preclinical animal models, this article outlines the hypothesis that dissemination trajectories do not only support the metastatic cascade, but also embody immunosuppressive niches, capable of providing transient and localized immunosubversion cues to the migratory/invasive cancer cell subpopulation while in the act of departing from a primary tumor. So long as these dissemination trajectories function as "immune deserts", the migratory tumor cell subpopulation remains efficient in evading immunological destruction and seeding metastatic sites, despite administration of cancer immunotherapy and/or other cytotoxic treatments. A deeper understanding of the molecular and cellular composition, as well as the signaling circuitries governing the function of these dissemination trajectories will further our overall understanding on TAM-mediated immunosuppression and will be paramount for the development of new therapeutic strategies for the advancement of optimal cancer chemotherapies, immunotherapies, and targeted therapies.
Collapse
Affiliation(s)
- Saeed Asiry
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Gina Kim
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Panagiota S. Filippou
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
- National Horizons Centre, Teesside University, Darlington, United Kingdom
| | - Luis Rivera Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - Douglas K. Marks
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY, United States
| | - Maja H. Oktay
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - George S. Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
8
|
Wei C, Wu J, Liu W, Lu J, Li H, Hai B. Tripartite motif-containing protein 6 facilitates growth and migration of breast cancer through degradation of STUB1. Eur J Histochem 2021; 65:3214. [PMID: 33728863 PMCID: PMC7967267 DOI: 10.4081/ejh.2021.3214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/02/2021] [Indexed: 12/23/2022] Open
Abstract
Proteins in the tripartite motif-containing protein (TRIM) family participates in carcinogenesis. However, little attention was focused on the role of TRIM6 on development of breast cancer. Expression level of TRIM6 was found to be markedly enhanced in breast cancer cells and tissues. Functional assays demonstrated that overexpression of TRIM6 promoted breast cancer progression through increase of YAP1 (Yes-associated Protein 1), while knockdown of TRIM6 suppressed in vitro breast cancer progression and in vivo tumor growth through decrease of YAP1. Co-Immunoprecipitation (co-IP) showed that TRIM6 interacted with STUB1 (stress induced phosphoprotein 1 homology and U-box containing protein 1). TRIM6 promoted ubiquitination-mediated degradation of STUB1 to promote YAP1 signaling. Overexpression of STUB1 attenuated TRIM6-induced promotion of breast cancer growth. In conclusion, TRIM6 contributed to breast cancer progression through ubiquitination-dependent proteasomal degradation of STUB1 and provocation of YAP1 pathway, providing potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Chuanchao Wei
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai.
| | - Jiayue Wu
- Department of Special Ward One, Shanghai Pulmonary Hospital, Shanghai.
| | - Weiyan Liu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai.
| | - Jingfeng Lu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai.
| | - Hongchang Li
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai.
| | - Benjun Hai
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai.
| |
Collapse
|
9
|
Korbecki J, Kojder K, Kapczuk P, Kupnicka P, Gawrońska-Szklarz B, Gutowska I, Chlubek D, Baranowska-Bosiacka I. The Effect of Hypoxia on the Expression of CXC Chemokines and CXC Chemokine Receptors-A Review of Literature. Int J Mol Sci 2021; 22:ijms22020843. [PMID: 33467722 PMCID: PMC7830156 DOI: 10.3390/ijms22020843] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Hypoxia is an integral component of the tumor microenvironment. Either as chronic or cycling hypoxia, it exerts a similar effect on cancer processes by activating hypoxia-inducible factor-1 (HIF-1) and nuclear factor (NF-κB), with cycling hypoxia showing a stronger proinflammatory influence. One of the systems affected by hypoxia is the CXC chemokine system. This paper reviews all available information on hypoxia-induced changes in the expression of all CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8 (IL-8), CXCL9, CXCL10, CXCL11, CXCL12 (SDF-1), CXCL13, CXCL14, CXCL15, CXCL16, CXCL17) as well as CXC chemokine receptors—CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CXCR7 and CXCR8. First, we present basic information on the effect of these chemoattractant cytokines on cancer processes. We then discuss the effect of hypoxia-induced changes on CXC chemokine expression on the angiogenesis, lymphangiogenesis and recruitment of various cells to the tumor niche, including myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), regulatory T cells (Tregs) and tumor-infiltrating lymphocytes (TILs). Finally, the review summarizes data on the use of drugs targeting the CXC chemokine system in cancer therapies.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland;
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Barbara Gawrońska-Szklarz
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
10
|
Premaratne ID, Toyoda Y, Celie KB, Brown KA, Spector JA. Tissue Engineering Models for the Study of Breast Neoplastic Disease and the Tumor Microenvironment. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:423-442. [DOI: 10.1089/ten.teb.2019.0347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ishani D. Premaratne
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Yoshiko Toyoda
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Karel-Bart Celie
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jason A. Spector
- Department of Surgery, Laboratory of Bioregenerative Medicine and Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, New York, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
11
|
Smit MJ, Schlecht-Louf G, Neves M, van den Bor J, Penela P, Siderius M, Bachelerie F, Mayor F. The CXCL12/CXCR4/ACKR3 Axis in the Tumor Microenvironment: Signaling, Crosstalk, and Therapeutic Targeting. Annu Rev Pharmacol Toxicol 2020; 61:541-563. [PMID: 32956018 DOI: 10.1146/annurev-pharmtox-010919-023340] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Elevated expression of the chemokine receptors CXCR4 and ACKR3 and of their cognate ligand CXCL12 is detected in a wide range of tumors and the tumor microenvironment (TME). Yet, the molecular mechanisms by which the CXCL12/CXCR4/ACKR3 axis contributes to the pathogenesis are complex and not fully understood. To dissect the role of this axis in cancer, we discuss its ability to impinge on canonical and less conventional signaling networks in different cancer cell types; its bidirectional crosstalk, notably with receptor tyrosine kinase (RTK) and other factors present in the TME; and the infiltration of immune cells that supporttumor progression. We discuss current and emerging avenues that target the CXCL12/CXCR4/ACKR3 axis. Coordinately targeting both RTKs and CXCR4/ACKR3 and/or CXCL12 is an attractive approach to consider in multitargeted cancer therapies. In addition, inhibiting infiltrating immune cells or reactivating the immune system along with modulating the CXCL12/CXCR4/ACKR3 axis in the TME has therapeutic promise.
Collapse
Affiliation(s)
- Martine J Smit
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France
| | - Maria Neves
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France.,Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Jelle van den Bor
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Petronila Penela
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Marco Siderius
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
12
|
Wang J, Tannous BA, Poznansky MC, Chen H. CXCR4 antagonist AMD3100 (plerixafor): From an impurity to a therapeutic agent. Pharmacol Res 2020; 159:105010. [PMID: 32544428 DOI: 10.1016/j.phrs.2020.105010] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/22/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
Abstract
AMD3100 (plerixafor), a CXCR4 antagonist, has opened a variety of avenues for potential therapeutic approaches in different refractory diseases. The CXCL12/CXCR4 axis and its signaling pathways are involved in diverse disorders including HIV-1 infection, tumor development, non-Hodgkin lymphoma, multiple myeloma, WHIM Syndrome, and so on. The mechanisms of action of AMD3100 may relate to mobilizing hematopoietic stem cells, blocking infection of X4 HIV-1, increasing circulating neutrophils, lymphocytes and monocytes, reducing myeloid-derived suppressor cells, and enhancing cytotoxic T-cell infiltration in tumors. Here, we first revisit the pharmacological discovery of AMD3100. We then review monotherapy of AMD3100 and combination use of AMD3100 with other agents in various diseases. Among those, we highlight the perspective of AMD3100 as an immunomodulator to regulate immune responses particularly in the tumor microenvironment and synergize with other therapeutics. All the pre-clinical studies support the clinical testing of the monotherapy and combination therapies with AMD3100 and further development for use in humans.
Collapse
Affiliation(s)
- Jingzhe Wang
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Medical School, Boston, MA, 02115, USA
| | - Huabiao Chen
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA; Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
13
|
Koens R, Tabata Y, Serrano JC, Aratake S, Yoshino D, Kamm RD, Funamoto K. Microfluidic platform for three-dimensional cell culture under spatiotemporal heterogeneity of oxygen tension. APL Bioeng 2020; 4:016106. [PMID: 32161836 PMCID: PMC7060087 DOI: 10.1063/1.5127069] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Cells in a tumor microenvironment are exposed to spatial and temporal variations in oxygen tension due to hyperproliferation and immature vascularization. Such spatiotemporal oxygen heterogeneity affects the behavior of cancer cells, leading to cancer growth and metastasis, and thus, it is essential to clarify the cellular responses of cancer cells to oxygen tension. Herein, we describe a new double-layer microfluidic device allowing the control of oxygen tension and the behavior of cancer cells under spatiotemporal oxygen heterogeneity. Two parallel gas channels were located above the media and gel channels to enhance gas exchange, and a gas-impermeable polycarbonate film was embedded in the device to prevent the diffusion of atmospheric oxygen. Variations in oxygen tension in the device with the experimental parameters and design variables were investigated computationally and validated by using oxygen-sensitive nanoparticles. The present device can generate a uniform hypoxic condition at oxygen levels down to 0.3% O2, as well as a linear oxygen gradient from 3% O2 to 17% O2 across the gel channel within 15 min. Moreover, human breast cancer cells suspended in type I collagen gel were introduced in the gel channel to observe their response under controlled oxygen tension. Hypoxic exposure activated the proliferation and motility of the cells, which showed a local maximum increase at 5% O2. Under the oxygen gradient condition, the increase in the cell number was relatively high in the central mild hypoxia region. These findings demonstrate the utility of the present device to study cellular responses in an oxygen-controlled microenvironment.
Collapse
Affiliation(s)
- Rei Koens
- Graduate School of Biomedical Engineering, Tohoku University, 6-6-12 Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | | | - Jean C. Serrano
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA
| | | | | | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
14
|
Design of Phthalocyanine‐Nanoparticle Hybrids for Photodynamic Therapy Applications in Oxygen‐Deficient Tumour Environment. ChemistrySelect 2019. [DOI: 10.1002/slct.201901466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
Targeting the phosphoinositide 3-kinase/AKT pathways by small molecules and natural compounds as a therapeutic approach for breast cancer cells. Mol Biol Rep 2019; 46:4809-4816. [PMID: 31313132 DOI: 10.1007/s11033-019-04929-x] [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: 02/13/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022]
Abstract
The phosphoinositide 3-kinase/AKT/mTOR (PI3K/AkT/mTOR) pathway plays a pivotal role in the uncontrolled growth, migration and development of human breast cancer. The elevated expression of TGF-β1 increases the PI3K/AkT/mTOR activity in human breast cancer tissue and potentially motivates tumor metastasis and resistance to chemotherapy. Here, we investigated whether treatment with PI3K/AkT/mTOR dual inhibitor NVP-BEZ235 alone or in combination with caffeic acid phenyl ester (CAPE) could prevent TGF-β1 effects on breast cancer cells. MCF-7 human breast cancer cells were exposed to TGF-β1 for 14 days and then were treated with/without NVP-BEZ235 and/or CAPE. Cell viability, apoptosis, CXCR4 surface expression and mRNA levels of CXCR4 and TWIST-1 were analyzed in all treated groups. We found that treatment of human breast cancer cells with a combination of NVP-BEZ235 and CAPE increased induction of cellular death. Although flow cytometry analysis demonstrated that NVP-BEZ235 alone treatment reduced CXCR4 expression while increasing CXCR4 mRNA level; when NVP-BEZ235 was combined with CAPE, inhibition of CXCR4 surface expression and enhancement of CXCR4 mRNA expression was diminished. In addition, TWIST-1 mRNA expression was down regulated in samples treated with both NVP-BEZ235 and CAPE. These altogether signified that NVP-BEZ235 in combination with CAPE showed improved therapeutic efficacy in breast cancer cells by decreasing apoptotic resistance and reduction of CXCR4 and TWIST-1 expression at mRNA level could be one of mechanism of action.
Collapse
|
16
|
Hwang PY, Brenot A, King AC, Longmore GD, George SC. Randomly Distributed K14 + Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues. Cancer Res 2019; 79:1899-1912. [PMID: 30862718 DOI: 10.1158/0008-5472.can-18-2828] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/27/2018] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Collective cell migration is an adaptive, coordinated interactive process involving cell-cell and cell-extracellular matrix (ECM) microenvironmental interactions. A critical aspect of collective migration is the sensing and establishment of directional movement. It has been proposed that a subgroup of cells known as leader cells localize at the front edge of a collectively migrating cluster and are responsible for directing migration. However, it is unknown how and when leader cells arrive at the front edge and what environmental cues dictate leader cell development and behavior. Here, we addressed these questions by combining a microfluidic device design that mimics multiple tumor microenvironmental cues concurrently with biologically relevant primary, heterogeneous tumor cell organoids. Prior to migration, breast tumor leader cells (K14+) were present throughout a tumor organoid and migrated (polarized) to the leading edge in response to biochemical and biomechanical cues. Impairment of either CXCR4 (biochemical responsive) or the collagen receptor DDR2 (biomechanical responsive) abrogated polarization of leader cells and directed collective migration. This work demonstrates that K14+ leader cells utilize both chemical and mechanical cues from the microenvironment to polarize to the leading edge of collectively migrating tumors. SIGNIFICANCE: These findings demonstrate that pre-existing, randomly distributed leader cells within primary tumor organoids use CXCR4 and DDR2 to polarize to the leading edge and direct migration.
Collapse
Affiliation(s)
- Priscilla Y Hwang
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri.,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Audrey Brenot
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri.,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Ashley C King
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri.,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Gregory D Longmore
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri. .,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri.,Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, Missouri
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, California.
| |
Collapse
|
17
|
Curcumin Suppresses Hepatic Stellate Cell-Induced Hepatocarcinoma Angiogenesis and Invasion through Downregulating CTGF. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8148510. [PMID: 30800209 PMCID: PMC6360067 DOI: 10.1155/2019/8148510] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/16/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022]
Abstract
Microenvironment plays a vital role in tumor progression; we focused on elucidating the role of hepatic stellate cells (HSCs) in hepatocarcinoma (HCC) aggressiveness and investigated the potential protective effect of curcumin on HSC-driven hepatocarcinoma angiogenesis and invasion. Our data suggest that HSCs increase HCC reactive oxygen species (ROS) production to upregulate hypoxia-inducible factor-1α (HIF-1α) expression to promote angiogenesis, epithelial to mesenchymal transition (EMT) process and invasion. And HSCs could secrete soluble factors, such as interleukin-6 (IL-6), vascular endothelial cell growth factor (VEGF), and stromal-derived factor-1 (SDF-1) to facilitate HCC progression. Curcumin could significantly suppress the above HSC-induced effects in HCC and could abrogate ROS and HIF-1α expression in HCC. HIF-1α or connective tissue growth factor (CTGF) knockdown could abolish the aforementioned curcumin affection. Moreover, CTGF is a downstream gene of HIF-1α. In addition, nuclear factor E2-related factor 2 (Nrf2) and glutathione (GSH) are involved in curcumin protection of HCC. These data indicate that curcumin may induce ROS scavenging by upregulating Nrf2 and GSH, thus inhibiting HIF-1α stabilization to suppress CTGF expression to exhibit its protection on HCC. Curcumin has a promising therapeutic effect on HCC. CTGF is responsible for curcumin-induced protection in HCC.
Collapse
|
18
|
GPCR Modulation in Breast Cancer. Int J Mol Sci 2018; 19:ijms19123840. [PMID: 30513833 PMCID: PMC6321247 DOI: 10.3390/ijms19123840] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most prevalent cancer found in women living in developed countries. Endocrine therapy is the mainstay of treatment for hormone-responsive breast tumors (about 70% of all breast cancers) and implies the use of selective estrogen receptor modulators and aromatase inhibitors. In contrast, triple-negative breast cancer (TNBC), a highly heterogeneous disease that may account for up to 24% of all newly diagnosed cases, is hormone-independent and characterized by a poor prognosis. As drug resistance is common in all breast cancer subtypes despite the different treatment modalities, novel therapies targeting signaling transduction pathways involved in the processes of breast carcinogenesis, tumor promotion and metastasis have been subject to accurate consideration. G protein-coupled receptors (GPCRs) are the largest family of cell-surface receptors involved in the development and progression of many tumors including breast cancer. Here we discuss data regarding GPCR-mediated signaling, pharmacological properties and biological outputs toward breast cancer tumorigenesis and metastasis. Furthermore, we address several drugs that have shown an unexpected opportunity to interfere with GPCR-based breast tumorigenic signals.
Collapse
|
19
|
Nobre AR, Entenberg D, Wang Y, Condeelis J, Aguirre-Ghiso JA. The Different Routes to Metastasis via Hypoxia-Regulated Programs. Trends Cell Biol 2018; 28:941-956. [PMID: 30041830 PMCID: PMC6214449 DOI: 10.1016/j.tcb.2018.06.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/18/2022]
Abstract
Hypoxia is linked to metastasis; however, how it affects metastatic progression is not clear due to limited consensus in the literature. We posit that this lack of consensus is due to hypoxia being studied using different approaches, such as in vitro, primary tumor, or metastasis assays in an isolated manner. Here, we review the pros and cons of in vitro hypoxia assays, highlight in vivo studies that inform on physiological hypoxia, and review the evidence that primary tumor hypoxia might influence the fate of disseminated tumor cells (DTCs) in secondary organs. Our analysis suggests that consensus can be reached by using in vivo methods of study, which also allow better modeling of how hypoxia affects DTC fate and metastasis.
Collapse
Affiliation(s)
- Ana Rita Nobre
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; Abel Salazar School of Biomedicine, Porto University, Porto, Portugal; These authors contributed equally
| | - David Entenberg
- Department of Anatomy and Structural Biology, Gruss Lipper Biophotonics Center, Integrated Imaging Program, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA; These authors contributed equally
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Gruss Lipper Biophotonics Center, Integrated Imaging Program, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - John Condeelis
- Department of Anatomy and Structural Biology, Gruss Lipper Biophotonics Center, Integrated Imaging Program, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA.
| | - Julio A Aguirre-Ghiso
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
20
|
Drenckhan A, Freytag M, Supuran CT, Sauter G, Izbicki JR, Gros SJ. CAIX furthers tumour progression in the hypoxic tumour microenvironment of esophageal carcinoma and is a possible therapeutic target. J Enzyme Inhib Med Chem 2018; 33:1024-1033. [PMID: 29865880 PMCID: PMC6010094 DOI: 10.1080/14756366.2018.1475369] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The hypoxic tumour microenvironment of solid tumours represents an important starting point for modulating progression and metastatic spread. Carbonic anhydrase IX (CAIX) is a known HIF-1α-dependent key player in maintaining cell pH conditions under hypoxia. We show that CAIX is strongly expressed in esophageal carcinoma tissues. We hypothesize that a moderate CAIX expression facilitates metastases and thereby worsens prognosis. Selective inhibition of CAIX by specific CAIX inhibitors and a CAIX knockdown effectively inhibit proliferation and migration in vitro. In the orthotopic esophageal carcinoma model, the humanized HER2 antibody trastuzumab down-regulates CAIX, possibly through CAIX’s linkage with HER2 in the hypoxic microenvironment. Our results show CAIX to be an essential part of the tumour microenvironment and a possible master regulator of tumour progression. This makes CAIX a highly effective and feasible therapeutic target for selective cancer treatment.
Collapse
Affiliation(s)
- Astrid Drenckhan
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Morton Freytag
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Claudiu T Supuran
- b Department Neurofarba , Section of Pharmaceutical Sciences, University of Florence , Florence , Italy
| | - Guido Sauter
- c Department of Pathology , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Jakob R Izbicki
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Stephanie J Gros
- a Department of General, Visceral and Thoracic Surgery , University Medical Center Hamburg-Eppendorf , Hamburg , Germany.,d Department of Pediatric Surgery , Ûniversity Children's Hospital Basel , Basel , Switzerland
| |
Collapse
|
21
|
Zhang Z, Li P, Wang Y, Yan H. Hypoxia‑induced expression of CXCR4 favors trophoblast cell migration and invasion via the activation of HIF‑1α. Int J Mol Med 2018; 42:1508-1516. [PMID: 29786753 PMCID: PMC6089771 DOI: 10.3892/ijmm.2018.3701] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/17/2018] [Indexed: 12/17/2022] Open
Abstract
The placenta initially develops in a low‑oxygen environment up to week 8‑10 of gestation, and a low oxygen level is a critical factor in the regulation of trophoblast migration and invasion. CXC chemokine receptor 4 (CXCR4) is transcriptionally activated by hypoxia in cancer cells. However, whether CXCR4 is involved in hypoxia‑inducible factor (HIF)‑1α‑dependent trophoblastic migration and invasion in a physiologically hypoxic environment (3% O2) remains to be fully elucidated and requires further investigation. In the present study, the expression of CXCR4 in first‑trimester villi was investigated, as was the response of the trophoblast to hypoxia, and the role of CXCR4 and HIF‑1α in trophoblast migration and invasion. CXCR4 was significantly elevated in the first‑trimester villi compared with normal full‑term placentas. In vitro, the expression of CXCR4 at the mRNA and protein levels was increased in JEG3 cells exposed to 3% O2 in a time‑dependent manner, and the migratory and invasive abilities of the JEG3 cells were upregulated. In addition, CXCR4 knockdown by transfection with CXCR4‑specific small interfering (si)RNA decreased the migration and invasion of JEG3 cells exposed to 3% O2. Furthermore, synthetic siRNA specific for HIF‑1α significantly suppressed the expression of CXCR4 in JEG3 cells exposed to 3% O2, whereas pcDNA‑HIF‑1α significantly increased the expression of CXCR4. These results indicated that the hypoxia‑induced expression of CXCR4 promoted trophoblast cell migration and invasion via the activation of HIF‑1α, which is crucial during placentation.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Pengyun Li
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yan Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Huan Yan
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
22
|
Wang J, Huang Y, Zhang J, Xing B, Xuan W, Wang H, Huang H, Yang J, Tang J. High co-expression of the SDF1/CXCR4 axis in hepatocarcinoma cells is regulated by AnnexinA7 in vitro and in vivo. Cell Commun Signal 2018; 16:22. [PMID: 29783989 PMCID: PMC5963093 DOI: 10.1186/s12964-018-0234-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/15/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND SDF1/CXCR4 and AnnexinA7 play important roles in many physiological and pathological conditions, but the molecular association between them in cancer cells has not been studied thus far. METHODS The expression changes of SDF1/CXCR4 were detected by gene transcriptome sequencing, qRT-PCR, Western blotting, cytoimmunofluorescence and immunohistochemistry in mouse hepatocarcinoma F/P cells, AnnexinA7 downregulated expression F (FA7DOWN) cells, AnnexinA7 overexpression P (PA7UP) cells, AnnexinA7 unrelated sequence F (FSHUS) cells, empty vector P (PNCEV) cells and normal liver cells in vitro and in vivo. RESULTS SDF1 and CXCR4 were co-expressed in hepatocarcinoma cells. SDF1 was localized mainly in the cytoplasm of cells, while CXCR4 was mainly localized in the cell membrane. Both in vitro and in vivo, expression levels of SDF1/CXCR4 in F and P cells were higher than in normal liver cells, and expression levels of SDF1/CXCR4 in F cells with high lymphatic metastatic potential were higher than those in P cells with low lymphatic metastatic potential. Expression of SDF1 was higher than that of CXCR4 in P cells and normal liver cells, while expression of CXCR4 was higher than that of SDF1 in F cells. Expression levels of SDF1/CXCR4 were completely consistent with AnnexinA7 regulation. After the AnnexinA7 gene was downregulated or upregulated, expression levels of SDF1/CXCR4 in FA7DOWN/PA7UP cells were lower or higher than those in FSHUS/PNCEV cells. Furthermore, CXCR4 was more sensitively modulated by AnnexinA7 regulation than SDF1. CONCLUSIONS High co-expression of SDF1/CXCR4 is a molecular characteristic of hepatocarcinoma cells, especially those with high lymphatic metastatic potential. AnnexinA7 positively regulates expression levels of SDF1/CXCR4, in particular CXCR4, and AnnexinA7 is a functional regulator of SDF1/CXCR4.
Collapse
Affiliation(s)
- Jingwen Wang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Yuhong Huang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Jun Zhang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Boyi Xing
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Wei Xuan
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Honghai Wang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - He Huang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | | | - Jianwu Tang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China.
| |
Collapse
|
23
|
Basheer HA, Pakanavicius E, Cooper PA, Shnyder SD, Martin L, Hunter KD, Vinader V, Afarinkia K. Hypoxia modulates CCR7 expression in head and neck cancers. Oral Oncol 2018; 80:64-73. [PMID: 29706190 DOI: 10.1016/j.oraloncology.2018.03.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 03/05/2018] [Accepted: 03/23/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND The chemokine receptor CCR7 is expressed on lymphocytes and dendritic cells and is responsible for trafficking of these cells in and out of secondary lymphoid organs. It has recently been shown that CCR7 expression is elevated in a number of cancers, including head and neck cancers, and that its expression correlates to lymph node (LN) metastasis. However, little is known about the factors that can induce CCR7 expression in head and neck cancers. METHOD We compared the protein expression and functional responses of CCR7 under normoxia and hypoxia in head and neck cancer cell lines OSC-19, FaDu, SCC-4, A-253 and Detroit-562 cultured as monolayers, spheroids, and grown in vivo as xenografts in balb/c mice. In addition, we analysed the correlation between hypoxia marker HIF-1α and CCR7 expression in a tissue microarray comprising 80 clinical samples with various stages and grades of malignant tumour and normal tissue. RESULTS Under hypoxia, the expression of CCR7 is elevated in both in vitro and in vivo models. Furthermore, in malignant tissue, a correlation is observed between hypoxia marker HIF-1α and CCR7 across all clinical stages. This correlation is also strong in early histological grade of tumours. CONCLUSION Hypoxia plays a role in the regulation of the expression of CCR7 and it may contribute to the development of a metastatic phenotype in head and neck cancers through this axis.
Collapse
Affiliation(s)
- Haneen A Basheer
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, United Kingdom; Faculty of Pharmacy, Zarqa University, PO Box 132222, Zarqa 13132, Jordan
| | - Edvinas Pakanavicius
- Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
| | - Patricia A Cooper
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Steven D Shnyder
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Lisette Martin
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Keith D Hunter
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Victoria Vinader
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Kamyar Afarinkia
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, United Kingdom.
| |
Collapse
|
24
|
Rosa P, Catacuzzeno L, Sforna L, Mangino G, Carlomagno S, Mincione G, Petrozza V, Ragona G, Franciolini F, Calogero A. BK channels blockage inhibits hypoxia-induced migration and chemoresistance to cisplatin in human glioblastoma cells. J Cell Physiol 2018; 233:6866-6877. [PMID: 29319175 DOI: 10.1002/jcp.26448] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) cells express large-conductance, calcium-activated potassium (BK) channels, whose activity is important for several critical aspects of the tumor, such as migration/invasion and cell death. GBMs are also characterized by a heavy hypoxic microenvironment that exacerbates tumor aggressiveness. Since hypoxia modulates the activity of BK channels in many tissues, we hypothesized that a hypoxia-induced modulation of these channels may contribute to the hypoxia-induced GBM aggressiveness. In U87-MG cells, hypoxia induced a functional upregulation of BK channel activity, without interfering with their plasma membrane expression. Wound healing and transwell migration assays showed that hypoxia increased the migratory ability of U87-MG cells, an effect that could be prevented by BK channel inhibition. Toxicological experiments showed that hypoxia was able to induce chemoresistance to cisplatin in U87-MG cells and that the inhibition of BK channels prevented the hypoxia-induced chemoresistance. Clonogenic assays showed that BK channels are also used to increase the clonogenic ability of U87-MG GBM cells in presence, but not in absence, of cisplatin. BK channels were also found to be essential for the hypoxia-induced de-differentiation of GBM cells. Finally, using immunohistochemical analysis, we highlighted the presence of BK channels in hypoxic areas of human GBM tissues, suggesting that our findings may have physiopathological relevance in vivo. In conclusion, our data show that BK channels promote several aspects of the aggressive potential of GBM cells induced by hypoxia, such as migration and chemoresistance to cisplatin, suggesting it as a potential therapeutic target in the treatment of GBM.
Collapse
Affiliation(s)
- Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Giorgio Mangino
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Silvia Carlomagno
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Gabriella Mincione
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" Chieti-Pescara, Italy
| | - Vincenzo Petrozza
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| | - Giuseppe Ragona
- Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy.,Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy.,Istituto Chirurgico Ortopedico Traumatologico, ICOT, Latina, Italy
| |
Collapse
|
25
|
Kenney RM, Lloyd CC, Whitman NA, Lockett MR. 3D cellular invasion platforms: how do paper-based cultures stack up? Chem Commun (Camb) 2018. [PMID: 28621775 DOI: 10.1039/c7cc02357j] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cellular invasion is the gateway to metastasis, which is the leading cause of cancer-related deaths. Invasion is driven by a number of chemical and mechanical stresses that arise in the tumor microenvironment. In vitro assays are needed for the systematic study of cancer progress. To be truly predictive, these assays must generate tissue-like environments that can be experimentally controlled and manipulated. While two-dimensional (2D) monolayer cultures are easily assembled and evaluated, they lack the extracellular components needed to assess invasion. Three-dimensional (3D) cultures are better suited for invasion studies because they generate cellular phenotypes that are more representative of those found in vivo. This feature article provides an overview of four invasion platforms. We focus on paper-based cultures, an emerging 3D culture platform capable of generating tissue-like structures and quantifying cellular invasion. Paper-based cultures are as easily assembled and analyzed as monolayers, but provide an experimentally powerful platform capable of supporting: co-cultures and representative extracellular environments; experimentally controlled gradients; readouts capable of quantifying, discerning, and separating cells based on their invasiveness. With a series of examples we highlight the potential of paper-based cultures, and discuss how they stack up against other invasion platforms.
Collapse
Affiliation(s)
- Rachael M Kenney
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, 125 South Road, Chapel Hill, NC 27599-3290, USA.
| | | | | | | |
Collapse
|
26
|
Lecavalier-Barsoum M, Chaudary N, Han K, Koritzinsky M, Hill R, Milosevic M. Targeting the CXCL12/CXCR4 pathway and myeloid cells to improve radiation treatment of locally advanced cervical cancer. Int J Cancer 2018; 143:1017-1028. [PMID: 29417588 DOI: 10.1002/ijc.31297] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/10/2018] [Accepted: 02/01/2018] [Indexed: 12/17/2022]
Abstract
Cervical cancer is the fourth most commonly diagnosed cancer and the fourth leading cause of cancer death in women worldwide. Approximately half of cervical cancer patients present with locally advanced disease, for which surgery is not an option. These cases are nonetheless potentially curable with radiotherapy and cisplatin chemotherapy. Unfortunately, some tumours are resistant to treatment, and lymph node and distant recurrences are major problems in patients with advanced disease at diagnosis. New targeted treatments that can overcome treatment resistance and reduce metastases are urgently needed. The CXCL12/CXCR4 chemokine pathway is ubiquitously expressed in many normal tissues and cancers, including cervical cancer. Emerging evidence indicates that it plays a central role in cervical cancer pathogenesis, malignant progression, the development of metastases and radiation treatment response. Pre-clinical studies of standard-of-care fractionated radiotherapy and concurrent weekly cisplatin plus the CXCR4 inhibitor Plerixafor (AMD3100) in patient-derived orthotopic cervical cancer xenografts have shown improved primary tumour response and reduced lymph node metastases with no increase in early or late side effects. These studies have pointed the way forward to future clinical trials of radiotherapy/cisplatin plus Plerixafor or other newly emerging CXCL12 or CXCR4 inhibitors in women with cervical cancer.
Collapse
Affiliation(s)
- Magali Lecavalier-Barsoum
- Department of Oncology, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Canada
| | - Naz Chaudary
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Canada
| | - Kathy Han
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Marianne Koritzinsky
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Richard Hill
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Canada.,Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Michael Milosevic
- Radiation Medicine Program, University Health Network and Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
27
|
Mukherjee D, Lu H, Yu L, He C, Lahiri SK, Li T, Zhao J. Krüppel-like factor 8 activates the transcription of C-X-C cytokine receptor type 4 to promote breast cancer cell invasion, transendothelial migration and metastasis. Oncotarget 2018; 7:23552-68. [PMID: 26993780 PMCID: PMC5029647 DOI: 10.18632/oncotarget.8083] [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: 11/07/2015] [Accepted: 02/25/2016] [Indexed: 02/04/2023] Open
Abstract
Krüppel-like factor 8 (KLF8) has been strongly implicated in breast cancer metastasis. However, the underlying mechanisms remain largely unknown. Here we report a novel signaling from KLF8 to C-X-C cytokine receptor type 4 (CXCR4) in breast cancer. Overexpression of KLF8 in MCF-10A cells induced CXCR4 expression at both mRNA and protein levels, as determined by quantitative real-time PCR and immunoblotting. This induction was well correlated with increased Boyden chamber migration, matrigel invasion and transendothelial migration (TEM) of the cells towards the ligand CXCL12. On the other hand, knockdown of KLF8 in MDA-MB-231 cells reduced CXCR4 expression associated with decreased cell migration, invasion and TEM towards CXCL12. Histological and database mining analyses of independent cohorts of patient tissue microarrays revealed a correlation of aberrant co-elevation of KLF8 and CXCR4 with metastatic potential. Promoter analysis indicated that KLF8 directly binds and activates the human CXCR4 gene promoter. Interestingly, a CXCR4-dependent activation of focal adhesion kinase (FAK), a known upregulator of KLF8, was highly induced by CXCL12 treatment in KLF8-overexpressing, but not KLF8 deficient cells. This activation of FAK in turn induced a further increase in KLF8 expression. Xenograft studies showed that overexpression of CXCR4, but not a dominant-negative mutant of it, in the MDA-MB-231 cells prevented the invasive growth of primary tumor and lung metastasis from inhibition by knockdown of KLF8. These results collectively suggest a critical role for a previously unidentified feed-forward signaling wheel made of KLF8, CXCR4 and FAK in promoting breast cancer metastasis and shed new light on potentially more effective anti-cancer strategies.
Collapse
Affiliation(s)
- Debarati Mukherjee
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL, USA
| | - Heng Lu
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL, USA
| | - Lin Yu
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL, USA
| | - Chunjiang He
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Satadru K Lahiri
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL, USA
| | - Tianshu Li
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL, USA.,Current address: Cleveland Clinic, Cleveland, OH, USA
| | - Jihe Zhao
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL, USA
| |
Collapse
|
28
|
Yang R, Duan C, Yuan L, Engelbach JA, Tsien CI, Beeman SC, Perez-Torres CJ, Ge X, Rich KM, Ackerman JJH, Garbow JR. Inhibitors of HIF-1α and CXCR4 Mitigate the Development of Radiation Necrosis in Mouse Brain. Int J Radiat Oncol Biol Phys 2017; 100:1016-1025. [PMID: 29485043 DOI: 10.1016/j.ijrobp.2017.12.257] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/30/2017] [Accepted: 12/11/2017] [Indexed: 01/06/2023]
Abstract
PURPOSE There is mounting evidence that, in addition to angiogenesis, hypoxia-induced inflammation via the hypoxia-inducible factor 1α (HIF-1α)-CXC chemokine receptor 4 (CXCR4) pathway may contribute to the pathogenesis of late-onset, irradiation-induced necrosis. This study investigates the mitigative efficacy of an HIF-1α inhibitor, topotecan, and a CXCR4 antagonist, AMD3100, on the development of radiation necrosis (RN) in an intracranial mouse model. METHODS AND MATERIALS Mice received a single-fraction, 50-Gy dose of hemispheric irradiation from the Leksell Gamma Knife Perfexion and were then treated with either topotecan, an HIF-1α inhibitor, from 1 to 12 weeks after irradiation, or AMD3100, a CXCR4 antagonist, from 4 to 12 weeks after irradiation. The onset and progression of RN were monitored longitudinally via noninvasive, in vivo magnetic resonance imaging (MRI) from 4 to 12 weeks after irradiation. Conventional hematoxylin-eosin staining and immunohistochemistry staining were performed to evaluate the treatment response. RESULTS The progression of brain RN was significantly mitigated for mice treated with either topotecan or AMD3100 compared with control animals. MRI-derived lesion volumes were significantly smaller for both of the treated groups, and histologic findings correlated well with the MRI data. By hematoxylin-eosin staining, both treated groups demonstrated reduced irradiation-induced tissue damage compared with controls. Furthermore, immunohistochemistry results revealed that expression levels of vascular endothelial growth factor, CXC chemokine ligand 12, CD68, CD3, and tumor necrosis factor α in the lesion area were significantly lower in treated (topotecan or AMD3100) brains versus control brains, while ionized calcium-binding adapter molecule 1 (Iba1) and HIF-1α expression was similar, though somewhat reduced. CXCR4 expression was reduced only in topotecan-treated mice, while interleukin 6 expression was unaffected by either topotecan or AMD3100. CONCLUSIONS By reducing inflammation, both topotecan and AMD3100 can, independently, mitigate the development of RN in the mouse brain. When combined with first-line, antiangiogenic treatment, anti-inflammation therapy may provide an adjuvant therapeutic strategy for clinical, postirradiation management of tumors, with additional benefits in the mitigation of RN development.
Collapse
Affiliation(s)
- Ruimeng Yang
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Department of Radiology, Washington University, St Louis, Missouri
| | - Chong Duan
- Department of Chemistry, Washington University, St Louis, Missouri
| | - Liya Yuan
- Department of Neurosurgery, Washington University, St Louis, Missouri
| | - John A Engelbach
- Department of Radiology, Washington University, St Louis, Missouri
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University, St Louis, Missouri
| | - Scott C Beeman
- Department of Radiology, Washington University, St Louis, Missouri
| | | | - Xia Ge
- Department of Radiology, Washington University, St Louis, Missouri
| | - Keith M Rich
- Department of Neurosurgery, Washington University, St Louis, Missouri; Department of Radiation Oncology, Washington University, St Louis, Missouri
| | - Joseph J H Ackerman
- Department of Radiology, Washington University, St Louis, Missouri; Department of Chemistry, Washington University, St Louis, Missouri; Department of Medicine, Washington University, St Louis, Missouri; Alvin J. Siteman Cancer Center, Washington University, St Louis, Missouri
| | - Joel R Garbow
- Department of Radiology, Washington University, St Louis, Missouri; Alvin J. Siteman Cancer Center, Washington University, St Louis, Missouri.
| |
Collapse
|
29
|
Hypoxia-induced reactive oxygen species mediate N-cadherin and SERPINE1 expression, EGFR signalling and motility in MDA-MB-468 breast cancer cells. Sci Rep 2017; 7:15140. [PMID: 29123322 PMCID: PMC5680275 DOI: 10.1038/s41598-017-15474-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/26/2017] [Indexed: 12/30/2022] Open
Abstract
One of the hallmarks of the tumour microenvironment is hypoxia resulting from increased oxygen consumption by proliferative cancer cells and altered vasculature. Hypoxic tension initiates various cellular signals and can drive epithelial to mesenchymal transition (EMT), a process important in cancer progression. In this study, using the antioxidant N-acetylcysteine (NAC), we show that hypoxia-induced reactive oxygen species (ROS) in MDA-MB-468 breast cancer cells, selectively regulate hypoxia-induced increases in N-cadherin and SERPINE1, two proteins involved in cell adhesion. Treatment of cells with NAC also attenuated hypoxia-mediated activation of EGFR, but did not have any effect on hypoxia-mediated induction of HIF1α. Exogenous hydrogen peroxide phenocopied the effects of hypoxia on N-cadherin and SERPINE1 expression and EGFR activation, suggesting its possible involvement in these hypoxia-mediated events. Reflective of their effect on cell adhesion proteins and EGFR (associated with migratory phenotypes), NAC also reduced cell migration under hypoxic conditions, a crucial event in metastasis. Our findings suggest a selective role for redox signalling in the regulation of specific components of the responses to hypoxia and induction of EMT in breast cancer cells. This study provides new evidence supporting the potential of targeting ROS as a therapeutic strategy for the control of breast cancer metastasis.
Collapse
|
30
|
Resveratrol inhibits hepatocellular carcinoma progression driven by hepatic stellate cells by targeting Gli-1. Mol Cell Biochem 2017; 434:17-24. [DOI: 10.1007/s11010-017-3031-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/01/2017] [Indexed: 02/06/2023]
|
31
|
Ponente M, Campanini L, Cuttano R, Piunti A, Delledonne GA, Coltella N, Valsecchi R, Villa A, Cavallaro U, Pattini L, Doglioni C, Bernardi R. PML promotes metastasis of triple-negative breast cancer through transcriptional regulation of HIF1A target genes. JCI Insight 2017; 2:e87380. [PMID: 28239645 DOI: 10.1172/jci.insight.87380] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Elucidating the molecular basis of tumor metastasis is pivotal for eradicating cancer-related mortality. Triple-negative breast cancer (TNBC) encompasses a class of aggressive tumors characterized by high rates of recurrence and metastasis, as well as poor overall survival. Here, we find that the promyelocytic leukemia protein PML exerts a prometastatic function in TNBC that can be targeted by arsenic trioxide. We found that, in TNBC patients, constitutive HIF1A activity induces high expression of PML, along with a number of HIF1A target genes that promote metastasis at multiple levels. Intriguingly, PML controls the expression of these genes by binding to their regulatory regions along with HIF1A. This mechanism is specific to TNBC cells and does not occur in other subtypes of breast cancer where PML and prometastatic HIF1A target genes are underexpressed. As a consequence, PML promotes cell migration, invasion, and metastasis in TNBC cell and mouse models. Notably, pharmacological inhibition of PML with arsenic trioxide, a PML-degrading agent used to treat promyelocytic leukemia patients, delays tumor growth, impairs TNBC metastasis, and cooperates with chemotherapy by preventing metastatic dissemination. In conclusion, we report identification of a prometastatic pathway in TNBC and suggest clinical development toward the use of arsenic trioxide for TNBC patients.
Collapse
Affiliation(s)
- Manfredi Ponente
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute.,Vita-Salute San Raffaele University
| | - Letizia Campanini
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute.,Vita-Salute San Raffaele University
| | - Roberto Cuttano
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute
| | - Andrea Piunti
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute
| | | | - Nadia Coltella
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute
| | - Roberta Valsecchi
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute
| | - Alessandra Villa
- Department of Experimental Oncology and Molecular Medicine Program, European Institute of Oncology, Milan, Italy
| | - Ugo Cavallaro
- Department of Experimental Oncology and Molecular Medicine Program, European Institute of Oncology, Milan, Italy
| | - Linda Pattini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy
| | - Claudio Doglioni
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosa Bernardi
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute
| |
Collapse
|
32
|
Cui X, Guo W, Sun Y, Sun B, Hu S, Sun D, Lam RHW. A microfluidic device for isolation and characterization of transendothelial migrating cancer cells. BIOMICROFLUIDICS 2017; 11:014105. [PMID: 28798840 PMCID: PMC5533502 DOI: 10.1063/1.4974012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/02/2017] [Indexed: 05/30/2023]
Abstract
Transendothelial migration of cancer cells is a critical stage in cancer, including breast cancer, as the migrating cells are generally believed to be highly metastatic. However, it is still challenging for many existing platforms to achieve a fully covering endothelium and to ensure transendothelial migration capability of the extracted cancer cells for analyses with high specificity. Here, we report a microfluidic device containing multiple independent cell collection microchambers underneath an embedded endothelium such that the transendothelial-migrated cells can be selectively collected from only the microchambers with full coverage of an endothelial layer. In this work, we first optimize the pore size of a microfabricated supporting membrane for the endothelium formation. We quantify transendothelial migration rates of a malignant human breast cell type (MDA-MB-231) under different shear stress levels. We investigate characteristics of the migrating cells including morphology, cytoskeletal structures, and migration (speed and persistence). Further implementation of this endothelium-embedded microfluidic device can provide important insights into migration and intracellular characteristics related to cancer metastasis and strategies for effective cancer therapy.
Collapse
Affiliation(s)
- Xin Cui
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Weijin Guo
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01002, USA
| | - Baoce Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Shuhuan Hu
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | | | | |
Collapse
|
33
|
Bamdad S, Khademi B, Chenari N, Taseh A, Razmkhah M. Stromal cell derived factor-1, CXCR4 and CXCR7 gene transcripts in pterygia. J Curr Ophthalmol 2016; 29:28-32. [PMID: 28367523 PMCID: PMC5362384 DOI: 10.1016/j.joco.2016.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/26/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Pterygium is a pathologic process with angiogenic and tumor cell like characteristics. Chemokine and chemokine receptors may contribute to the formation and growth of pterygia. The aim of this study was to assess the expression of stromal cell derived factor (SDF)-1, as an angiogenic chemokine, and its receptors, CXCR4 and CXCR7, gene transcripts in pterygia. METHODS RNA was extracted from tissue samples of 33 patients with primary pterygium and 35 volunteers with conjunctiva as the control group. Then the mRNA expression of SDF-1, CXCR4, and CXCR7 was assessed through quantitative Real Time PCR method using appropriate primers. RESULTS SDF-1 and both receptors transcripts had significantly higher expression in pterygia samples compared to the control group (P < 0.05). The ratio of CXCR7 transcript expression to CXCR4 was 26.4 in patients while it was 11 in controls. CONCLUSION As SDF-1 and its receptors, CXCR4 and CXCR7, were up-regulated in pterygia, SDF-1/CXCR4/CXCR7 axis may contribute to pterygium formation which can be possibly restrained by down-regulating this signaling pathway.
Collapse
Affiliation(s)
- Shahram Bamdad
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behzad Khademi
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nooshin Chenari
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atta Taseh
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
34
|
An N, Luo X, Zhang M, Yu R. MicroRNA-376b promotes breast cancer metastasis by targeting Hoxd10 directly. Exp Ther Med 2016; 13:79-84. [PMID: 28123472 PMCID: PMC5245143 DOI: 10.3892/etm.2016.3942] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/05/2016] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most common malignant disease in women, and metastasis formed at distant anatomic sites was the major cause of cancer-related mortality. Thus, a novel therapy target and progression biomarker for breast cancer metastasis was necessary. microRNA (miR)-376b has been demonstrated to regulate angiogenesis; however, its role in cancer metastasis remains elusive. In the present study, the expression of miR-376b in normal breast tissue, JC and 4T1 cells was determined by qPCR. Furthermore, in vitro and in vivo experiments were performed to determine the effect of miR-376b on breast cancer metastasis. The direct target of miR-376b was determined by the luciferase assay and western blotting. The results indicated that silencing of miR-376b by the miR-376-mimic significantly inhibited 4T1 cell migration and invasion in vitro. Lung metastasis was also evidently decreased after silencing of miR-376b in 4T1 cells. Moreover, the luciferase assay and western blotting identified that Hoxd10 is the direct target of miR-376b during the regulation of breast cancer metastasis. To the best of our knowledge, the present study was the first to demonstrate the promoting breast cancer metastasis role of miR-376b by directly targeting Hoxd10. Therefore, it would be a novel therapy target and prognostic biomarker for breast cancer.
Collapse
Affiliation(s)
- Ning An
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xinmei Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ming Zhang
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Ruilian Yu
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
35
|
ZHANG YING, CABARCAS STEPHANIEM, ZHENG JI, SUN LEI, MATHEWS LESLEYA, ZHANG XIAOHU, LIN HONGSHENG, FARRAR WILLIAML. Cryptotanshinone targets tumor-initiating cells through down-regulation of stemness genes expression. Oncol Lett 2016; 11:3803-3812. [PMID: 27313698 PMCID: PMC4888080 DOI: 10.3892/ol.2016.4444] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 01/19/2016] [Indexed: 01/06/2023] Open
Abstract
Recent evidence indicates that tumor-initiating cells (TICs), also called cancer stem cells (CSCs), are responsible for tumor initiation and progression, therefore representing an important cell population that may be used as a target for the development of future anticancer therapies. In the present study, Cryptotanshinone (CT), a traditional Chinese herbal medicine, was demonstrated to regulate the behaviors of LNCaP prostate cells and prostate LNCaP TICs. The results demonstrate that treatment with CT alters cellular proliferation, cell cycle status, migration, viability, colony formation and notably, sphere formation and down-regulation of stemness genes (Nanog, OCT4, SOX2, β-catenin, CXCR4) in TICs. The present study demonstrates that CT targets the LNCaP CD44+CD24- population that is representative of prostate TICs and also affects total LNCaP cells as well via down-regulation of stemness genes. The strong effect with which CT has on prostate TICs suggests that CT may potentially function as a novel natural anticancer agent that specifically targets TICs.
Collapse
Affiliation(s)
- YING ZHANG
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
- Oncology Department, Guang An Men Hospital of China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - STEPHANIE M. CABARCAS
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
| | - JI ZHENG
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
| | - LEI SUN
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
| | - LESLEY A. MATHEWS
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
| | - XIAOHU ZHANG
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
| | - HONGSHENG LIN
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
- Oncology Department, Guang An Men Hospital of China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - WILLIAM L. FARRAR
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Cancer Institute-Frederick, Center for Cancer Research, Frederick, MD 21702, USA
| |
Collapse
|
36
|
Rieger CT, Fiegl M. Microenvironmental oxygen partial pressure in acute myeloid leukemia: Is there really a role for hypoxia? Exp Hematol 2016; 44:578-82. [PMID: 27118044 DOI: 10.1016/j.exphem.2016.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/16/2016] [Accepted: 04/18/2016] [Indexed: 02/02/2023]
Abstract
Reduced oxygen partial pressure (pO2) has been recognized as being relevant in hematopoiesis and the pathophysiology of malignant diseases. Although hypoxic (meaning insufficient supply of oxygen) and anoxic areas are present and of pathophysiologic importance (by hypoxia-induced pathways such as HiF1α) in solid tumors, this may not be true for (malignant) hematologic cells. Hematopoiesis occurs in the stem cell niche, which is characterized, among other things, by extremely low pO2. However, in contrast to solid tumors, in this context, the low pO2 is physiological and this feature, among others, is shared by the malignant stem cell niche harboring leukemia-initiating cells. Upon differentiation, hematopoietic cells are constantly exposed to changes in pO2 as they travel throughout the human body and encounter arterial and venous blood and migrate into oxygen-carrier-free tissue with low pO2. Hematologic malignancies such as acute myeloid leukemia (AML) make little difference in this respect and, whereas low oxygen is the usual environment of AML cells, recent evidence suggests no role for real hypoxia. Although there is no evidence that AML pathophysiology is related to hypoxia, leukemic blasts still show several distinct biological features when exposed to reduced pO2: they down- or upregulate membrane receptors such as CXCR4 or FLT3, activate or inhibit intracellular signaling pathways such as PI3K, and specifically secrete cytokines (IL-8). In summary, reduced pO2 should not be mistaken for hypoxia (nor should it be so called), and it does not automatically induce hypoxia-response mechanisms; therefore, a strict distinction should be made between physiologically low pO2 (physoxia) and hypoxia.
Collapse
Affiliation(s)
- Christina T Rieger
- Department of Internal Medicine III, Klinikum der Universität München, 81377 Munich, Germany
| | - Michael Fiegl
- Department of Internal Medicine III, Klinikum der Universität München, 81377 Munich, Germany.
| |
Collapse
|
37
|
Liang W, Ni Y, Chen F. Tumor resistance to vascular disrupting agents: mechanisms, imaging, and solutions. Oncotarget 2016; 7:15444-59. [PMID: 26812886 PMCID: PMC4941252 DOI: 10.18632/oncotarget.6999] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/14/2016] [Indexed: 01/04/2023] Open
Abstract
The emergence of vascular disrupting agents (VDAs) is a significant advance in the treatment of solid tumors. VDAs induce rapid and selective shutdown of tumor blood flow resulting in massive necrosis. However, a viable marginal tumor rim always remains after VDA treatment and is a major cause of recurrence. In this review, we discuss the mechanisms involved in the resistance of solid tumors to VDAs. Hypoxia, tumor-associated macrophages, and bone marrow-derived circulating endothelial progenitor cells all may contribute to resistance. Resistance can be monitored using magnetic resonance imaging markers. The various solutions proposed to manage tumor resistance to VDAs emphasize combining these agents with other approaches including antiangiogenic agents, chemotherapy, radiotherapy, radioimmunotherapy, and sequential dual-targeting internal radiotherapy.
Collapse
Affiliation(s)
- Wenjie Liang
- Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yicheng Ni
- Radiology Section, University Hospitals, University of Leuven, Leuven, Belgium
| | - Feng Chen
- Department of Radiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
38
|
Sarkar R, Mukherjee S, Biswas J, Roy M. Phenethyl isothiocyanate, by virtue of its antioxidant activity, inhibits invasiveness and metastatic potential of breast cancer cells: HIF-1α as a putative target. Free Radic Res 2015; 50:84-100. [DOI: 10.3109/10715762.2015.1108520] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Murakami JL, Xu B, Franco CB, Hu X, Galli SJ, Weissman IL, Chen CC. Evidence that β7 Integrin Regulates Hematopoietic Stem Cell Homing and Engraftment Through Interaction with MAdCAM-1. Stem Cells Dev 2015; 25:18-26. [PMID: 26422691 DOI: 10.1089/scd.2014.0551] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
α4β7 integrin is a cell adhesion receptor that is crucial for the migration of hematopoietic progenitors and mature effector cells in the periphery, but its role in adult hematopoiesis is controversial. We identified a subset of hematopoietic stem cells (HSCs) in the bone marrow (BM) that expressed β7 integrin. These β7(+) HSCs were capable of multilineage, long-term reconstitution and had an inherent competitive advantage over β7(-) HSCs. On the other hand, HSCs that lacked β7 integrin (β7KO) had reduced engraftment potential. Interestingly, quantitative RT-PCR and flow cytometry revealed that β7KO HSCs expressed lower levels of the chemokine receptor CXCR4. Accordingly, β7KO HSCs exhibited impaired migration abilities in vitro and BM homing capabilities in vivo. Lethal irradiation induced expression of the α4β7 integrin ligand-mucosal addressin cell adhesion molecule-1 (MAdCAM-1) on BM endothelial cells. Moreover, blocking MAdCAM-1 reduced the homing of HSCs and impaired the survival of recipient mice. Altogether, these data indicate that β7 integrin, when expressed by HSCs, interacted with its endothelial ligand MAdCAM-1 in the BM microenvironment, thereby promoting HSC homing and engraftment.
Collapse
Affiliation(s)
- Jodi L Murakami
- 1 Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of City of Hope , Duarte, California.,2 City of Hope Irell & Manella Graduate School of Biological Sciences , Duarte, California.,3 Gehr Family Center for Leukemia Research at City of Hope , Duarte, California
| | - Baohui Xu
- 4 Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Christopher B Franco
- 5 Department of Pathology, Stanford University School of Medicine , Stanford, California.,6 Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Xingbin Hu
- 1 Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of City of Hope , Duarte, California.,7 Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, People's Republic of China
| | - Stephen J Galli
- 5 Department of Pathology, Stanford University School of Medicine , Stanford, California.,8 Department of Microbiology and Immunology, Stanford University School of Medicine , Stanford, California
| | - Irving L Weissman
- 5 Department of Pathology, Stanford University School of Medicine , Stanford, California.,6 Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Ching-Cheng Chen
- 1 Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of City of Hope , Duarte, California.,2 City of Hope Irell & Manella Graduate School of Biological Sciences , Duarte, California.,3 Gehr Family Center for Leukemia Research at City of Hope , Duarte, California
| |
Collapse
|
40
|
Rivas-Fuentes S, Salgado-Aguayo A, Pertuz Belloso S, Gorocica Rosete P, Alvarado-Vásquez N, Aquino-Jarquin G. Role of Chemokines in Non-Small Cell Lung Cancer: Angiogenesis and Inflammation. J Cancer 2015; 6:938-52. [PMID: 26316890 PMCID: PMC4543754 DOI: 10.7150/jca.12286] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/23/2015] [Indexed: 12/12/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of aggressive cancer. The tumor tissue, which shows an active angiogenesis, is composed of neoplastic and stromal cells, and an abundant inflammatory infiltrate. Angiogenesis is important to support tumor growth, while infiltrating cells contribute to the tumor microenvironment through the secretion of growth factors, cytokines and chemokines, important molecules in the progression of the disease. Chemokines are important in development, activation of the immune response, and physiological angiogenesis. Chemokines have emerged as important regulators in the pathophysiology of cancer. These molecules are involved in the angiogenesis/angiostasis balance and in the recruitment of tumor infiltrating hematopoietic cells. In addition, chemokines promote tumor cell survival, as well as the directing and establishment of tumor cells to metastasis sites. The findings summarized here emphasize the central role of chemokines as modulators of tumor angiogenesis and their potential role as therapeutic targets in the inflammatory process of NSCLC angiogenesis.
Collapse
Affiliation(s)
- Selma Rivas-Fuentes
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Alfonso Salgado-Aguayo
- 2. Laboratory of Research on Rheumatic Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Silvana Pertuz Belloso
- 3. Department of Comparative Biology, Faculty of Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Patricia Gorocica Rosete
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Noé Alvarado-Vásquez
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Guillermo Aquino-Jarquin
- 4. Laboratory of Research on Genomics, Genetics and Bioinformatics. Tower of Haemato-oncology, Children´s Hospital of Mexico “Federico Gomez”, Mexico City, Mexico
| |
Collapse
|
41
|
Guan G, Zhang Y, Lu Y, Liu L, Shi D, Wen Y, Yang L, Ma Q, Liu T, Zhu X, Qiu X, Zhou Y. The HIF-1α/CXCR4 pathway supports hypoxia-induced metastasis of human osteosarcoma cells. Cancer Lett 2014; 357:254-264. [PMID: 25444927 DOI: 10.1016/j.canlet.2014.11.034] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 12/11/2022]
Abstract
HIF-1α mediates hypoxia-induced expression of the chemokine receptor CXCR4 and contributes to metastasis in many different cancers. We have previously shown that hypoxia promotes migration of human osteosarcoma cells by activating the HIF-1α/CXCR4 pathway. Here, immunohistochemical analysis showed that unlike control osteochondroma samples, osteosarcoma specimens were characterized by elevated expression levels of HIF-1α and CXCR4. Moreover, we found that hypoxia-induced invasiveness was more pronounced in high metastatic potential F5M2 osteosarcoma cells than in low metastatic potential F4 cells, and that this induction was sensitive to treatment with the CXCR4 antagonist AMD3100 and the HIF-1α inhibitor KC7F2. Interestingly, hypoxia-induced CXCR4 expression persisted after cultured osteosarcoma cells were returned to normoxic conditions. These observations were confirmed by experiments in a mouse model of osteosarcoma lung metastasis showing that hypoxia stimulation of pulmonary metastasis was greater in F5M2 than in F4 cells, and was sensitive to treatment with AMD3100. Our study provides further evidence of the contributions of hypoxia and the HIF-1α/CXCR4 pathway to the progression of osteosarcoma, and suggests that this axis might be efficiently leveraged in the development of novel osteosarcoma therapeutics.
Collapse
Affiliation(s)
- Guofeng Guan
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Yinglong Zhang
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Yao Lu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Lijuan Liu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Doufei Shi
- Department of Geriatrics, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Yanhua Wen
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Lianjia Yang
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Qiong Ma
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Tao Liu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Xiaodong Zhu
- Department of Microsurgery, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, China.
| | - Xiuchun Qiu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| | - Yong Zhou
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| |
Collapse
|
42
|
Exploring the competition between proliferative and invasive cancer phenotypes in a continuous spatial model. PLoS One 2014; 9:e103191. [PMID: 25099885 PMCID: PMC4123877 DOI: 10.1371/journal.pone.0103191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 06/27/2014] [Indexed: 12/02/2022] Open
Abstract
Tumor is characterized by extensive heterogeneity with respect to its microenvironment and its genetic composition. We extend a previously developed monoclonal continuous spatial model of tumor growth to account for polyclonal cell populations and investigate the interplay between a more proliferative and a more invasive phenotype under different conditions. The model simulations demonstrate a transition from the dominance of the proliferative to the dominance of the invasive phenotype resembling malignant tumor progression and show a time period where both subpopulations are abundant. As the dominant phenotype switches from proliferative to invasive, the geometry of tumor changes from a compact and almost spherical shape to a more diffusive and fingered morphology with the proliferative phenotype to be restricted in the tumor bulk and the invasive to dominate at tumor edges. Different micro-environmental conditions and different phenotypic properties can promote or inhibit invasion demonstrating their mutual importance. The model provides a computational framework to investigate tumor heterogeneity and the constant interplay between the environment and the specific characteristics of phenotypes that should be taken into account for the prediction of tumor evolution, morphology and effective treatment.
Collapse
|
43
|
Lucansky V, Krmencikova-Fliegl M, Stanek L, Vonka V. Administration of a plasmid that expresses SDF-1α affects the oncogenic potential of mouse bcr-abl-transformed cells. Mol Med Rep 2014; 10:2116-22. [PMID: 25070183 DOI: 10.3892/mmr.2014.2425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/06/2013] [Indexed: 11/06/2022] Open
Abstract
Stromal-derived factor 1α (SDF‑1α, also known as CXCL12) is a chemokine that exerts its effects through the G-protein coupled receptors, C-X-C chemokine receptor type 4 (CXCR4) and 7 (CXCR7). There is marked evidence that the SDF-1/CXCR4 axis is involved in the pathogenesis of leukemia and therapies that target this axis are under development. The present study aimed to increase the efficacy of a DNA-based bcr-abl vaccine by simultaneously immunizing mice with a plasmid carrying the whole SDF-1α gene. Bcr-abl‑transformed 12B1 cells were used to challenge the mice. These cells have the oncogenic potential to induce both leukemia following intravenous inoculation and lymphoma-type solid tumors after subcutaneous inoculation. Administering an SDF‑1 carrying plasmid together with the bcr-abl vaccine resulted in increased survival following a challenge with subcutaneously administered 12B1 cells, although the difference was not statistically significant. However, there was a difference when the animals that developed subcutaneous tumors were only taken into consideration. In doubly-treated mice, significantly more mice failed to develop solid tumors than mice that had only received the bcr-abl vaccine. By contrast, the occurrence of fatal leukemia was significantly higher in the mice that were treated with the SDF-1 plasmid, regardless of whether they were immunized with the bcr-abl-vaccine. No humoral or cellular immune responses against SDF‑1 were detected in the treated mice, which suggested that the changes in oncogenic potential of 12B1 cells were due to the activity of SDF-1 itself.
Collapse
Affiliation(s)
- Vincent Lucansky
- Department of Experimental Virology, Institute of Hematology and Blood Transfusion, CZ-128 20 Prague 2, Czech Republic
| | - Monika Krmencikova-Fliegl
- Department of Experimental Virology, Institute of Hematology and Blood Transfusion, CZ-128 20 Prague 2, Czech Republic
| | - Libor Stanek
- Department of Experimental Virology, Institute of Hematology and Blood Transfusion, CZ-128 20 Prague 2, Czech Republic
| | - Vladimir Vonka
- Department of Experimental Virology, Institute of Hematology and Blood Transfusion, CZ-128 20 Prague 2, Czech Republic
| |
Collapse
|
44
|
Boyle ST, Kochetkova M. Breast cancer stem cells and the immune system: promotion, evasion and therapy. J Mammary Gland Biol Neoplasia 2014; 19:203-11. [PMID: 24997735 DOI: 10.1007/s10911-014-9323-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/16/2014] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells are believed to be a subset of heterogeneous tumour cells responsible for tumour initiation, growth, local invasion, and metastasis. In breast cancer, numerous factors have been implicated in regulation of cancer stem cells, but there is still a paucity of information regarding precise molecular and cellular mechanisms guiding their pathobiology. Components of both the adaptive and the innate immune system have been shown to play a crucial role in supporting breast cancer growth and spread, and recently some immune mediators, both molecules and cells, have been reported to influence breast cancer stem cell biology. This review summarises a small, pioneering body of evidence for the potentially important function of the "immuniche" in maintaining and supporting breast cancer stem cells.
Collapse
Affiliation(s)
- Sarah T Boyle
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | | |
Collapse
|
45
|
Arnolds KL, Spencer JV. CXCR4: a virus's best friend? INFECTION GENETICS AND EVOLUTION 2014; 25:146-56. [PMID: 24793563 DOI: 10.1016/j.meegid.2014.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 10/25/2022]
Abstract
Viruses are dependent on their hosts for replication and dispersal in the environment; thus, the most successful viruses are those that co-evolve with their hosts. CXCR4 is a cellular chemokine receptor that plays central roles in development, hematopoiesis, and immune surveillance through signaling induced by its ligand, CXCL12. The CXCR4-CXCL12 axis has been besieged by many pathogens that employ a range of strategies to modify or exploit CXCR4 activity. While CXCR4 was identified as a critical co-factor for entry of HIV into CD4+ T cells early on, other viruses may utilize CXCR4 to gain cell entry as well. Moreover, several viruses have been found to modulate CXCR4 expression or alter its functional activity, with direct effects on cell trafficking, immune responses, cell proliferation, and cell survival. Because CXCR4 is targeted by a diverse group of viral pathogens, modification of host CXCR4 signaling activity is emerging as a common theme in virus persistence and is likely to be important for subversion of the host immune system. This review highlights major viral pathogens that use and abuse CXCR4 and explores the possible reasons why this chemokine receptor has become "a virus's best friend".
Collapse
Affiliation(s)
- Kathleen L Arnolds
- Department of Biology, University of San Francisco, 2130 Fulton Street, San Francisco, CA 94403, United States
| | - Juliet V Spencer
- Department of Biology, University of San Francisco, 2130 Fulton Street, San Francisco, CA 94403, United States.
| |
Collapse
|
46
|
Guo M, Cai C, Zhao G, Qiu X, Zhao H, Ma Q, Tian L, Li X, Hu Y, Liao B, Ma B, Fan Q. Hypoxia promotes migration and induces CXCR4 expression via HIF-1α activation in human osteosarcoma. PLoS One 2014; 9:e90518. [PMID: 24618817 PMCID: PMC3949690 DOI: 10.1371/journal.pone.0090518] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 02/03/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Cellular adaptation to a hypoxic microenvironment is essential for tumor progression and is largely mediated by HIF-1α through coordinated regulation of hypoxia-responsive genes. The chemokine SDF-1α and its unique receptor CXCR4 have been implicated in organ-specific metastases of many cancers. In this study, we investigated the response of osteosarcoma cells to hypoxia and the expression of CXCR4 and HIF-1α in human osteosarcoma specimens and explored the roles of CXCR4 and HIF-1α in the cell migration process. METHODOLOGY/PRINCIPAL FINDINGS We performed immunohistochemistry, immunocytochemistry, quantitative real-time PCR, Western blots and fluorescent reporter assays to evaluate the correlation between CXCR4 and HIF-1α expression in human osteosarcoma specimens or SOSP-9607 cells under normoxic and hypoxic conditions. Transwell assays were used to assess cell migration under different conditions. Exposure of SOSP-9607 cells to hypoxic conditions resulted in significantly increased migration. When SOSP-9607 cells were subjected to hypoxic conditions, the mRNA and protein levels of CXCR4 were significantly increased in a time-dependent manner. Moreover, siHIF-1α significantly decreased the mRNA and protein levels of CXCR4 under hypoxia, whereas pcDNA-HIF-1α significantly increased the mRNA and protein levels of CXCR4 under normoxia. A luciferase reporter gene study showed that siHIF-1α reduced pGL3-CXCR4 luciferase activity. Furthermore, coexpression of HIF-1α and CXCR4 was significantly higher in patients with distant metastasis compared with those without metastasis. CONCLUSIONS/SIGNIFICANCE The hypoxia-HIF-1α-CXCR4 pathway plays a crucial role during the migration of human osteosarcoma cells, and targeting this pathway might represent a novel therapeutic strategy for patients suffering from osteosarcoma.
Collapse
Affiliation(s)
- Mingjun Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chengkui Cai
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Guangyi Zhao
- Department of Orthopaedics, Bethune International Peace Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Xiuchun Qiu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Haien Zhao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Qiong Ma
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Liying Tian
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Xuelian Li
- Department of Surgery, Xi'an Hospital of TCM, Shaanxi, People's Republic of China
| | - Yunsheng Hu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Bo Liao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Baoan Ma
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
- * E-mail: (BM); (QF)
| | - Qingyu Fan
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
- * E-mail: (BM); (QF)
| |
Collapse
|
47
|
Pinto S, Martínez-Romero A, O'Connor JE, Gil-Benso R, San-Miguel T, Terrádez L, Monteagudo C, Callaghan RC. Intracellular coexpression of CXC- and CC- chemokine receptors and their ligands in human melanoma cell lines and dynamic variations after xenotransplantation. BMC Cancer 2014; 14:118. [PMID: 24559071 PMCID: PMC3943502 DOI: 10.1186/1471-2407-14-118] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 02/14/2014] [Indexed: 12/05/2022] Open
Abstract
Background Chemokines have been implicated in tumor progression and metastasis. In melanoma, chemokine receptors have been implicated in organ selective metastasis by regulating processes such as chemoattraction, adhesion and survival. Methods In this study we have analyzed, using flow cytometry, the systems formed by the chemokine receptors CXCR3, CXCR4, CXCR7, CCR7 and CCR10 and their ligands in thirteen human melanoma cell lines (five established from primary tumors and eight established from metastasis from different tissues). WM-115 and WM-266.4 melanoma cell lines (obtained from a primary and a metastatic melanoma respectively) were xenografted in nude mice and the tumors and cell lines derived from them were also analyzed. Results Our results show that the melanoma cell lines do not express or express in a low degree the chemokine receptors on their cell surface. However, melanoma cell lines show intracellular expression of all the aforementioned receptors and most of their respective ligands. When analyzing the xenografts and the cell lines obtained from them we found variations in the intracellular expression of chemokines and chemokine receptors that differed between the primary and metastatic cell lines. However, as well as in the original cell lines, minute or no expression of the chemokine receptors was observed at the cell surface. Conclusions Coexpression of chemokine receptors and their ligands was found in human melanoma cell lines. However, this expression is intracellular and receptors are not found at the cell membrane nor chemokines are secreted to the cell medium. The levels of expressed chemokine receptors and their ligands show dynamic variations after xenotransplantation that differ depending on the origin of the cell line (from primary tumor or from metastasis).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robert C Callaghan
- Cytomics Laboratory, Mixed Unit CIPF-UVEG, Príncipe Felipe Research Centre, Valencia, Avda Autopista del Saler, 16, 46012 Valencia, Spain.
| |
Collapse
|
48
|
Lei J, Huo X, Duan W, Xu Q, Li R, Ma J, Li X, Han L, Li W, Sun H, Wu E, Ma Q. α-Mangostin inhibits hypoxia-driven ROS-induced PSC activation and pancreatic cancer cell invasion. Cancer Lett 2014; 347:129-38. [PMID: 24513179 DOI: 10.1016/j.canlet.2014.02.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/22/2014] [Accepted: 02/03/2014] [Indexed: 12/18/2022]
Abstract
Recent advances indicating a key role of microenvironment for tumor progression, we investigated the role of PSCs and hypoxia in pancreatic cancer aggressiveness, and examined the potential protective effect of α-mangostin on hypoxia-driven pancreatic cancer progression. Our data indicate that hypoxic PSCs exploit their oxidative stress due to hypoxia to secrete soluble factors favouring pancreatic cancer invasion. α-Mangostin suppresses hypoxia-induced PSC activation and pancreatic cancer cell invasion through the inhibition of HIF-1α stabilization and GLI1 expression. Increased generation of hypoxic ROS is responsible for HIF-1α stabilization and GLI1 upregulation. Therefore, α-mangostin may be beneficial in preventing hypoxia-induced pancreatic cancer progression.
Collapse
Affiliation(s)
- Jianjun Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Xiongwei Huo
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Qinhong Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Rong Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Jiguang Ma
- Department of Oncology, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Xuqi Li
- Department of General Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Liang Han
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Wei Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China
| | - Hao Sun
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China.
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, USA
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, Shaanxi Province, China.
| |
Collapse
|
49
|
Zhang HW, Sun XF, He YN, Li JT, Guo XH, Liu H. Bioinformatics analysis of breast cancer bone metastasis related gene-CXCR4. ASIAN PAC J TROP MED 2014; 6:732-8. [PMID: 23827153 DOI: 10.1016/s1995-7645(13)60128-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/15/2013] [Accepted: 08/15/2013] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To analyze breast cancer bone metastasis related gene-CXCR4. METHODS This research screened breast cancer bone metastasis related genes by high-flux gene chip. RESULTS It was found that the expressions of 396 genes were different including 165 up-regulations and 231 down-regulations. The expression of chemokine receptor CXCR4 was obviously up-regulated in the tissue with breast cancer bone metastasis. Compared with the tissue without bone metastasis, there was significant difference, which indicated that CXCR4 played a vital role in breast cancer bone metastasis. CONCLUSIONS The bioinformatics analysis of CXCR4 can provide a certain basis for the occurrence and diagnosis of breast cancer bone metastasis, target gene therapy and evaluation of prognosis.
Collapse
Affiliation(s)
- Heng-Wei Zhang
- Department of Breast, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | | | | | | | | | | |
Collapse
|
50
|
Fakhari S, Kalantar E, Nikzaban M, Hakhamneshi MS, Fathi F, Nikkhoo B, Rahmani MR, Beiraghdar M, Jalili A. Effect of Helicobacter pylori infection on stromal-derived factor-1/CXCR4 axis in bone marrow-derived mesenchymal stem cells. Adv Biomed Res 2014; 3:19. [PMID: 24592369 PMCID: PMC3929140 DOI: 10.4103/2277-9175.124650] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/10/2013] [Indexed: 02/06/2023] Open
Abstract
Background: Recent studies have demonstrated that during chronic Helicobacter pylori (H. pylori) infection bone marrow-derived-mesenchymal stem cells (BMD-MSCs) migrate to the gastric tissue and could be also the origin of gastric adenocarcinoma. The chemokine CXCR4 through binding to its ligand stromal-derived factor (SDF-1) plays a crucial role in migration of inflammatory and stem cells. However, the possible effect of H. pylori infection on the SDF-1/CXCR4 axis has not yet been elucidated. Materials and Methods: Gastric epithelial cell line, AGS, and BMD-MSCs were cocultured with H. pylori for 24 h. The expression of CXCR4 was examined in BMD-MSCs by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and flow cytometry, and SDF-1 expression in AGS cells was detected by qRT-PCR and enzyme-linked immunosorbent assay. Further, migration of BMD-MSCs toward SDF-1 was evaluated by chemotaxis assay. Results: We found that coculture of H. pylori with BMD-MSCs or AGS: (i) enhanced CXCR4 expression on the cell surface of BMD-MSCs and (ii) increased SDF-1 secretion by AGS cells. Consistently, we observed that H. pylori-treated BMD-MSCs showed a higher capability to migrate toward SDF-1 gradient compared with untreated cells. Conclusion: We found that H. pylori upregulates CXCR4 expression in BMD-MSCs and enhance their migration toward SDF-1. This study provides the first evidence that H. pylori infection may enhance BMD-MSC migration through acting on the SDF-1/CXCR4 axis.
Collapse
Affiliation(s)
- Shohreh Fakhari
- Kurdistan Molecular & Cellular Research Center, Kurdistan University of Medical Sciences, Sanadaj, Iran
| | - Enayat Kalantar
- Department of Microbiology, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehrnoush Nikzaban
- Kurdistan Molecular & Cellular Research Center, Kurdistan University of Medical Sciences, Sanadaj, Iran
| | - Mohammad Said Hakhamneshi
- Kurdistan Molecular & Cellular Research Center, Kurdistan University of Medical Sciences, Sanadaj, Iran
| | - Fardin Fathi
- Kurdistan Molecular & Cellular Research Center, Kurdistan University of Medical Sciences, Sanadaj, Iran
| | - Bahram Nikkhoo
- Kurdistan Molecular & Cellular Research Center, Kurdistan University of Medical Sciences, Sanadaj, Iran
| | - Mohammad Reza Rahmani
- Kurdistan Molecular & Cellular Research Center, Kurdistan University of Medical Sciences, Sanadaj, Iran
| | - Mina Beiraghdar
- Department of Physiology, Isfahan Payamnoor University, Isfahan, Iran
| | - Ali Jalili
- Kurdistan Molecular & Cellular Research Center, Kurdistan University of Medical Sciences, Sanadaj, Iran
| |
Collapse
|