1
|
Aljabali AAA, Tambuwala MM, El-Tanani M, Hassan SS, Lundstrom K, Mishra V, Mishra Y, Hromić-Jahjefendić A, Redwan EM, Uversky VN. A comprehensive review of PRAME and BAP1 in melanoma: Genomic instability and immunotherapy targets. Cell Signal 2024; 124:111434. [PMID: 39326690 DOI: 10.1016/j.cellsig.2024.111434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
In a thorough review of the literature, the complex roles of PRAME (preferentially expressed Antigen of Melanoma) and BAP1 (BRCA1-associated protein 1) have been investigated in uveal melanoma (UM) and cutaneous melanoma. High PRAME expression in UM is associated with poor outcomes and correlated with extraocular extension and chromosome 8q alterations. BAP1 mutations in the UM indicate genomic instability and a poor prognosis. Combining PRAME and BAP1 immunohistochemical staining facilitates effective risk stratification. Mechanistically, both genes are associated with genomic instability, making them promising targets for cancer immunotherapy. Hypomethylation of PRAME, specifically in its promoter regions, is critical for UM progression and contributes to epigenetic reprogramming. Additionally, miR-211 regulation is crucial in melanoma and has therapeutic potential. The way PRAME changes signaling pathways provides clues about the cause of cancer due to genomic instability related to modifications in DNA repair. Inhibition of poly(ADP-ribose) polymerase-1 (PARP-1) and PARP-2 in cells expressing PRAME could lead to potential therapeutic applications. Pathway enrichment analysis underscores the significance of PRAME and BAP1 in melanoma pathogenesis.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan.
| | - Murtaza M Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, PO Box 11172, United Arab Emirates.
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, PO Box 11172, United Arab Emirates.
| | - Sk Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur, 721140, West Bengal, India.
| | | | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, 21934 Alexandria, Egypt.
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
2
|
Wen ZH, Chang L, Yang SN, Yu CL, Tung FY, Kuo HM, Lu IC, Wu CY, Shih PC, Chen WF, Chen NF. The anti-angiogenic and anti-vasculogenic mimicry effects of GN25 in endothelial and glioma cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119799. [PMID: 39043304 DOI: 10.1016/j.bbamcr.2024.119799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND AND PURPOSE Scientists have been exploring anti-angiogenic strategies to inhibit angiogenesis and prevent tumor growth. Vasculogenic mimicry (VM) in glioblastoma multiforme (GBM) poses a challenge, complicating anti-angiogenesis therapy. A novel drug, GN25 (3-[{1,4-dihydro-5,8-dimethoxy-1,4-dioxo-2-naphthalenyl}thio]-propanoic acid), can inhibit tumor formation. This study aims to investigate the microenvironmental effects and molecular mechanisms of GN25 in anti-angiogenesis and anti-VM. EXPERIMENTAL APPROACH MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) assay was used to evaluate the cell viability of different concentrations of GN25 in human umbilical vein endothelial cells (HUVEC) and Uppsala 87 malignant glioma (U87MG) cells. Functional assays were used to investigate the effects of GN25 on angiogenesis-related processes, whereas gelatin zymography, enzyme-linked immunosorbent assays, and Western blotting were utilized to assess the influence on matrix metalloproteinase (MMP)-2 and vascular endothelial growth factor (VEGF) secretion and related signaling pathways. KEY RESULTS GN25 suppressed migration, wound healing, and tube formation in HUVECs and disrupted angiogenesis in a rat aorta ring and zebrafish embryo model. GN25 dose-dependently reduced phosphatidylinositol 3-kinase/AKT and inhibited MMP-2/VEGF secretion in HUVECs. In U87MG cells, GN25 inhibited migration, wound healing, and VM, accompanied by a decrease in MMP-2 and VEGF secretion. The results indicate that GN25 effectively inhibits angiogenesis and VM formation in HUVECs and U87MG cells without affecting preexisting vascular structures. CONCLUSION AND IMPLICATIONS This study elaborated GN25's potential as an anti-angiogenic agent by elucidating its inhibitory effects on classical angiogenesis. VM provides valuable insights for developing novel therapeutic strategies against tumor progression and angiogenesis-related diseases. These results indicate the potential of GN25 as a promising candidate for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan; Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Long Chang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - San-Nan Yang
- Department of Pediatrics, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Chen-Ling Yu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Fang-Yu Tung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hsiao-Mei Kuo
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833301, Taiwan
| | - I-Chen Lu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Chang Shih
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833301, Taiwan.
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan.
| |
Collapse
|
3
|
Wang X, Xue X, Pang M, Yu L, Qian J, Li X, Tian M, Lyu A, Lu C, Liu Y. Epithelial-mesenchymal plasticity in cancer: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e659. [PMID: 39092293 PMCID: PMC11292400 DOI: 10.1002/mco2.659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Currently, cancer is still a leading cause of human death globally. Tumor deterioration comprises multiple events including metastasis, therapeutic resistance and immune evasion, all of which are tightly related to the phenotypic plasticity especially epithelial-mesenchymal plasticity (EMP). Tumor cells with EMP are manifest in three states as epithelial-mesenchymal transition (EMT), partial EMT, and mesenchymal-epithelial transition, which orchestrate the phenotypic switch and heterogeneity of tumor cells via transcriptional regulation and a series of signaling pathways, including transforming growth factor-β, Wnt/β-catenin, and Notch. However, due to the complicated nature of EMP, the diverse process of EMP is still not fully understood. In this review, we systematically conclude the biological background, regulating mechanisms of EMP as well as the role of EMP in therapy response. We also summarize a range of small molecule inhibitors, immune-related therapeutic approaches, and combination therapies that have been developed to target EMP for the outstanding role of EMP-driven tumor deterioration. Additionally, we explore the potential technique for EMP-based tumor mechanistic investigation and therapeutic research, which may burst vigorous prospects. Overall, we elucidate the multifaceted aspects of EMP in tumor progression and suggest a promising direction of cancer treatment based on targeting EMP.
Collapse
Affiliation(s)
- Xiangpeng Wang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoxia Xue
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Mingshi Pang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Liuchunyang Yu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Jinxiu Qian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoyu Li
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Meng Tian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Aiping Lyu
- School of Chinese MedicineHong Kong Baptist UniversityKowloonHong KongChina
| | - Cheng Lu
- Institute of Basic Research in Clinical MedicineChina Academy of Chinese Medical SciencesBeijingChina
| | - Yuanyan Liu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
4
|
Dong ZX, Chan SH, Chen SN, Li M, Zhang XD, Liu XQ. TJP1 promotes vascular mimicry in bladder cancer by facilitating VEGFA expression and transcriptional activity through TWIST1. Transl Oncol 2023; 32:101666. [PMID: 37031603 PMCID: PMC10119961 DOI: 10.1016/j.tranon.2023.101666] [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] [Received: 11/27/2022] [Revised: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 04/11/2023] Open
Abstract
Tight junction protein 1 (TJP1) is a recently identified prominent regulator of bladder cancer (BLCA) angiogenesis and tumorigenesis. Vascular mimicry (VM) is a newly described tumor feature and is correlated with an increased risk of tumor metastasis. However, the relationship between TJP1 expression and VM in bladder cancer remains elusive. In the present study, we report a novel function for TJP1 in accommodating VM to promote tumor progression. We found that the elevated TJP1 expression was positively related to VM in patients and xenograft tumor models in bladder cancer. Enforced expression of TJP1 increased VM of BLCA cells in vitro and in vivo by elevating Vascular endothelial growth factor A (VEGFA) levels. Furthermore, VM induced by TJP1 overexpression was significantly blocked by the VEGFA and VEGFR inhibitors (Bevacizumab and Sunitinib). Mechanistically, TJP1 promoted VEGFA transcriptional and protein level in a TWIST1-dependent manner. Taken together, our study reveals that TJP1-regulated VEGFA overexpression may indicate a potential therapeutic target for clinical intervention in the early tumor neovascularization of bladder cancer.
Collapse
Affiliation(s)
- Zhao-Xia Dong
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Sze-Hoi Chan
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Shu-Na Chen
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Miao Li
- Department of Hematology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
| | - Xing-Ding Zhang
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China.
| | - Xue-Qi Liu
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
5
|
Davalos V, Lovell CD, Von Itter R, Dolgalev I, Agrawal P, Baptiste G, Kahler DJ, Sokolova E, Moran S, Piqué L, Vega-Saenz de Miera E, Fontanals-Cirera B, Karz A, Tsirigos A, Yun C, Darvishian F, Etchevers HC, Osman I, Esteller M, Schober M, Hernando E. An epigenetic switch controls an alternative NR2F2 isoform that unleashes a metastatic program in melanoma. Nat Commun 2023; 14:1867. [PMID: 37015919 PMCID: PMC10073109 DOI: 10.1038/s41467-023-36967-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/24/2023] [Indexed: 04/06/2023] Open
Abstract
Metastatic melanoma develops once transformed melanocytic cells begin to de-differentiate into migratory and invasive melanoma cells with neural crest cell (NCC)-like and epithelial-to-mesenchymal transition (EMT)-like features. However, it is still unclear how transformed melanocytes assume a metastatic melanoma cell state. Here, we define DNA methylation changes that accompany metastatic progression in melanoma patients and discover Nuclear Receptor Subfamily 2 Group F, Member 2 - isoform 2 (NR2F2-Iso2) as an epigenetically regulated metastasis driver. NR2F2-Iso2 is transcribed from an alternative transcriptional start site (TSS) and it is truncated at the N-terminal end which encodes the NR2F2 DNA-binding domain. We find that NR2F2-Iso2 expression is turned off by DNA methylation when NCCs differentiate into melanocytes. Conversely, this process is reversed during metastatic melanoma progression, when NR2F2-Iso2 becomes increasingly hypomethylated and re-expressed. Our functional and molecular studies suggest that NR2F2-Iso2 drives metastatic melanoma progression by modulating the activity of full-length NR2F2 (Isoform 1) over EMT- and NCC-associated target genes. Our findings indicate that DNA methylation changes play a crucial role during metastatic melanoma progression, and their control of NR2F2 activity allows transformed melanocytes to acquire NCC-like and EMT-like features. This epigenetically regulated transcriptional plasticity facilitates cell state transitions and metastatic spread.
Collapse
Affiliation(s)
- Veronica Davalos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA.
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.
| | - Claudia D Lovell
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Richard Von Itter
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Igor Dolgalev
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Praveen Agrawal
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine/ Montefiore, Bronx, NY, 10461, USA
| | - Gillian Baptiste
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - David J Kahler
- High Throughput Biology Core, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Elena Sokolova
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Sebastian Moran
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Laia Piqué
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Eleazar Vega-Saenz de Miera
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Barbara Fontanals-Cirera
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Alcida Karz
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Chi Yun
- High Throughput Biology Core, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Farbod Darvishian
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | | | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- Centro de Investigacion Biomedica en Red, Cancer (CIBERONC), Madrid, Spain
| | - Markus Schober
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA.
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Cell Biology, New York Grossman University School of Medicine, New York, NY, 10016, USA.
| | - Eva Hernando
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
6
|
Kang R, Song M, Fang Z, Liu K. Nano-composite hydrogels of Cu-Apa micelles for anti-vasculogenic mimicry. J Drug Target 2023; 31:166-178. [PMID: 35993258 DOI: 10.1080/1061186x.2022.2115047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Vasculogenic mimicry (VM) describes the phenomenon whereby fluid-conducting vessels are formed by highly invasive tumour cells, which supply blood to tumours during their early growth stages. Single antiangiogenic agents have limited inhibitory effects on VM, therefore, a multi-pathway anti-VM strategy is required. In this study, Apatinib (Apa) was coordinated with Cu2+ to form a Cu-Apa copper complex. The latter was loaded into oligo-hyaluronic acid (HA) polymeric micelles (HA-Chol) and subsequently embedded in Astragalus polysaccharide-based in situ hydrogels (APsGels) to generate Cu-Apa/HA-Chol@APsGels. In this system, Cu-Apa exerts the combined effects of Cu2+ and Apa to inhibit VM; HA-Chol micelles achieve targeted drug delivery and enhance endocytosis efficiency; APsGels realise sustained release of the drugs to ensure an anti-VM effect. This system demonstrated improved VM inhibition with low cytotoxicity and high biocompatibility, wound healing, and transwell invasion in three-dimensional cell cultured VM. Moreover, this system significantly inhibited VM formation and melanoma growth in a mouse tumour transplantation model. This study provides an effective strategy for inhibiting VM.
Collapse
Affiliation(s)
- Rui Kang
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, PR China
| | - Mengdi Song
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, PR China
| | - Zhou Fang
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, PR China
| | - Kehai Liu
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, PR China
| |
Collapse
|
7
|
Nisin delivery by nanosponges increases its anticancer activity against in-vivo melanoma model. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Olbryt M. Potential Biomarkers of Skin Melanoma Resistance to Targeted Therapy—Present State and Perspectives. Cancers (Basel) 2022; 14:cancers14092315. [PMID: 35565444 PMCID: PMC9102921 DOI: 10.3390/cancers14092315] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Around 5–10% of advanced melanoma patients progress early on anti-BRAF targeted therapy and 20–30% respond only with the stabilization of the disease. Presumably, these patients could benefit more from first-line immunotherapy. Resistance to BRAF/MEK inhibitors is generated by genetic and non-genetic factors inherent to a tumor or acquired during therapy. Some of them are well documented as a cause of treatment failure. They are potential predictive markers that could improve patients’ selection for both standard and also alternative therapy as some of them have therapeutic potential. Here, a summary of the most promising predictive and therapeutic targets is presented. This up-to-date knowledge may be useful for further study on implementing more accurate genetic/molecular tests in melanoma treatment. Abstract Melanoma is the most aggressive skin cancer, the number of which is increasing worldwide every year. It is completely curable in its early stage and fatal when spread to distant organs. In addition to new therapeutic strategies, biomarkers are an important element in the successful fight against this cancer. At present, biomarkers are mainly used in diagnostics. Some biological indicators also allow the estimation of the patient’s prognosis. Still, predictive markers are underrepresented in clinics. Currently, the only such indicator is the presence of the V600E mutation in the BRAF gene in cancer cells, which qualifies the patient for therapy with inhibitors of the MAPK pathway. The identification of response markers is particularly important given primary and acquired resistance to targeted therapies. Reliable predictive tests would enable the selection of patients who would have the best chance of benefiting from treatment. Here, up-to-date knowledge about the most promising genetic and non-genetic resistance-related factors is described. These are alterations in MAPK, PI3K/AKT, and RB signaling pathways, e.g., due to mutations in NRAS, RAC1, MAP2K1, MAP2K2, and NF1, but also other changes activating these pathways, such as the overexpression of HGF or EGFR. Most of them are also potential therapeutic targets and this issue is also addressed here.
Collapse
Affiliation(s)
- Magdalena Olbryt
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| |
Collapse
|
9
|
Combined Action of Hyper-Harmonized Hydroxylated Fullerene Water Complex and Hyperpolarized Light Leads to Melanoma Cell Reprogramming In Vitro. NANOMATERIALS 2022; 12:nano12081331. [PMID: 35458039 PMCID: PMC9033139 DOI: 10.3390/nano12081331] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 01/03/2023]
Abstract
(1) Background: Their unique structure and electron deficiency have brought fullerenes into the focus of research in many fields, including medicine. The hyper-harmonized hydroxylated fullerene water complex (3HFWC) formulation has solved the limitations of the poor solubility and bioavailability of fullerenes. To achieve better antitumor activity, 3HFWC was combined with short-term irradiation of cells with hyperpolarized light (HPL) generated by the application of a nanophotonic fullerene filter in a Bioptron® device. The benefits of HPL were confirmed in the microcirculation, wound healing and immunological function. (2) Methods: B16, B16-F10 and A375 melanoma cells were exposed to a wide spectrum of 3HFWC doses and to a single short-term HPL irradiation. (3) Results: Apart from the differences in the redox status and level of invasiveness, the effects of the treatments were quite similar. Decreased viability, morphological alteration, signs of melanocytic differentiation and cellular senescence were observed upon the successful internalization of the nanoquantum substance. (4) Conclusions: Overall, 3HFWC/HPL promoted melanoma cell reprogramming toward a normal phenotype.
Collapse
|
10
|
Reikvam H, Hatfield KJ, Wendelbo Ø, Lindås R, Lassalle P, Bruserud Ø. Endocan in Acute Leukemia: Current Knowledge and Future Perspectives. Biomolecules 2022; 12:biom12040492. [PMID: 35454082 PMCID: PMC9027427 DOI: 10.3390/biom12040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Endocan is a soluble dermatan sulfate proteoglycan expressed by endothelial cells and detected in serum/plasma. Its expression is increased in tumors/tumor vessels in several human malignancies, and high expression (high serum/plasma levels or tumor levels) has an adverse prognostic impact in several malignancies. The p14 endocan degradation product can also be detected in serum/plasma, but previous clinical studies as well as previously unpublished results presented in this review suggest that endocan and p14 endocan fragment levels reflect different biological characteristics, and the endocan levels seem to reflect the disease heterogeneity in acute leukemia better than the p14 fragment levels. Furthermore, decreased systemic endocan levels in previously immunocompetent sepsis patients are associated with later severe respiratory complications, but it is not known whether this is true also for immunocompromised acute leukemia patients. Finally, endocan is associated with increased early nonrelapse mortality in (acute leukemia) patients receiving allogeneic stem cell transplantation, and this adverse prognostic impact seems to be independent of the adverse impact of excessive fluid overload. Systemic endocan levels may also become important to predict cytokine release syndrome after immunotherapy/haploidentical transplantation, and in the long-term follow-up of acute leukemia survivors with regard to cardiovascular risk. Therapeutic targeting of endocan is now possible, and the possible role of endocan in acute leukemia should be further investigated to clarify whether the therapeutic strategy should also be considered.
Collapse
Affiliation(s)
- Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Kimberley Joanne Hatfield
- Department of Transfusion Medicine and Immunology, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Øystein Wendelbo
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Roald Lindås
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
| | - Philippe Lassalle
- Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR9017, University of Lille, 59000 Lille, France;
- Center for Infection and Immunity, le Centre Nationale de la Recherche Scientifique, Univeristy of Lille, 59000 Lille, France
- Centre d’Infection et d’Immunité de Lille, Equipe Immunité Pulmonaire, University of Lille, 59000 Lille, France
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway;
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (Ø.W.); (R.L.)
- Correspondence:
| |
Collapse
|
11
|
Simbulan-Rosenthal CM, Haribabu Y, Vakili S, Kuo LW, Clark H, Dougherty R, Alobaidi R, Carney B, Sykora P, Rosenthal DS. Employing CRISPR-Cas9 to Generate CD133 Synthetic Lethal Melanoma Stem Cells. Int J Mol Sci 2022; 23:2333. [PMID: 35216449 PMCID: PMC8877091 DOI: 10.3390/ijms23042333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/19/2022] Open
Abstract
Malignant melanoma is a lethal skin cancer containing melanoma-initiating cells (MIC) implicated in tumorigenesis, invasion, and drug resistance, and is characterized by the elevated expression of stem cell markers, including CD133. The siRNA knockdown of CD133 enhances apoptosis induced by the MEK inhibitor trametinib in melanoma cells. This study investigates the underlying mechanisms of CD133's anti-apoptotic activity in patient-derived BAKP and POT cells, harboring difficult-to-treat NRASQ61K and NRASQ61R drivers, after CRISPR-Cas9 CD133 knockout or Dox-inducible expression of CD133. MACS-sorted CD133(+) BAKP cells were conditionally reprogrammed to derive BAKR cells with sustained CD133 expression and MIC features. Compared to BAKP, CD133(+) BAKR exhibit increased cell survival and reduced apoptosis in response to trametinib or the chemotherapeutic dacarbazine (DTIC). CRISPR-Cas9-mediated CD133 knockout in BAKR cells (BAKR-KO) re-sensitized cells to trametinib. CD133 knockout in BAKP and POT cells increased trametinib-induced apoptosis by reducing anti-apoptotic BCL-xL, p-AKT, and p-BAD and increasing pro-apoptotic BAX. Conversely, Dox-induced CD133 expression diminished apoptosis in both trametinib-treated cell lines, coincident with elevated p-AKT, p-BAD, BCL-2, and BCL-xL and decreased activation of BAX and caspases-3 and -9. AKT1/2 siRNA knockdown or inhibition of BCL-2 family members with navitoclax (ABT-263) in BAKP-KO cells further enhanced caspase-mediated apoptotic PARP cleavage. CD133 may therefore activate a survival pathway where (1) increased AKT phosphorylation and activation induces (2) BAD phosphorylation and inactivation, (3) decreases BAX activation, and (4) reduces caspases-3 and -9 activity and caspase-mediated PARP cleavage, leading to apoptosis suppression and drug resistance in melanoma. Targeting nodes of the CD133, AKT, or BCL-2 survival pathways with trametinib highlights the potential for combination therapies for NRAS-mutant melanoma stem cells for the development of more effective treatments for patients with high-risk melanoma.
Collapse
Affiliation(s)
- Cynthia M. Simbulan-Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Yogameenakshi Haribabu
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Sahar Vakili
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Li-Wei Kuo
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Havens Clark
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Ryan Dougherty
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Ryyan Alobaidi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| | - Bonnie Carney
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
- Firefighters’ Burn and Surgical Laboratory, MedStar Health Research Institute, Washington, DC 20010, USA
| | - Peter Sykora
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
- Amelia Technologies, LLC, 1121 5th St. NW, Washington, DC 20001, USA
| | - Dean S. Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (C.M.S.-R.); (Y.H.); (S.V.); (L.-W.K.); (H.C.); (R.D.); (R.A.); (B.C.); (P.S.)
| |
Collapse
|
12
|
He W, Yang G, Liu S, Maghsoudloo M, Shasaltaneh MD, Kaboli PJ, Zhang C, Zhang J, Entezari M, Imani S, Wen Q. Comparative mRNA/micro-RNA co-expression network drives melanomagenesis by promoting epithelial-mesenchymal transition and vasculogenic mimicry signaling. Transl Oncol 2021; 14:101237. [PMID: 34626953 PMCID: PMC8512639 DOI: 10.1016/j.tranon.2021.101237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/17/2022] Open
Abstract
This study aimed to identify a novel disease-associated differentially co-expressed mRNA-microRNA (miRNA) that is associated with vasculogenic mimicry (VM) and epithelial-to-mesenchymal transition (EMT) network at different stages of melanoma. By applying weighted gene co-expression network analysis, we constructed a VM+EMT biological network with the available microarray dataset downloaded from a public database. Quantitative real-time PCR, immunohistochemical staining, and CD31-periodic acid solution dual staining were performed to confirm the expression of genes associated with EMT and VM formation in subjects with malignant melanoma (n = 18) and primary melanoma (n = 13) and in healthy subjects (n = 10). Our findings suggested that phosphatidylserine-specific phospholipase A1-alpha (PLA1A) and dermokine (DMKN) genes function as oncogenes that trigger VM and EMT processes during melanomagenesis on interaction with miR-370, miR-563, and miR-770-5p. PLA1A and DMKN genes can be considered potential VM+EMT network-based diagnostic biomarkers for distinguishing between melanoma patients. We postulate that a network with altered PLA1A/miR-563 and DMNK/miR-770-5p/miR-370 may contribute to melanomagenesis by triggering the EMT signaling pathway and VM formation. This study provides a potentially valuable approach for the early diagnosis and prognosis of melanoma progression.
Collapse
Affiliation(s)
- WenFeng He
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Oncology, Anyue Hospital of Traditional Chinese Medicine, Second Ziyang Hospital of Traditional Chinese Medicine, Ziyang, Sichuan, China
| | - Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Oncology, Chengdu Jinniu District People's Hospital, Chengdu, Sichuan, China
| | - Mazaher Maghsoudloo
- Laboratory of Systems Biology and Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Cuiwei Zhang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - JingHeng Zhang
- Oncology Department, Luzhou People's Hospital, Luzhou, Sichuan, China
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - QingLian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
13
|
Dong S, Chen Z, Wang L, Liu Y, Stagos D, Lin X, Liu M. Marine Bromophenol Bis(2,3,6-Tribromo-4,5-Dihydroxybenzyl)ether Inhibits Angiogenesis in Human Umbilical Vein Endothelial Cells and Reduces Vasculogenic Mimicry in Human Lung Cancer A549 Cells. Mar Drugs 2021; 19:641. [PMID: 34822512 PMCID: PMC8617710 DOI: 10.3390/md19110641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis, including the growth of new capillary blood vessels from existing ones and the malignant tumors cells formed vasculogenic mimicry, is quite important for the tumor metastasis. Anti-angiogenesis is one of the significant therapies in tumor treatment, while the clinical angiogenesis inhibitors usually exhibit endothelial cells dysfunction and drug resistance. Bis(2,3,6-tribromo-4,5-dihydroxybenzyl)ether (BTDE), a marine algae-derived bromophenol compound, has shown various biological activities, however, its anti-angiogenesis function remains unknown. The present study illustrated that BTDE had anti-angiogenesis effect in vitro through inhibiting human umbilical vein endothelial cells migration, invasion, tube formation, and the activity of matrix metalloproteinases 9 (MMP9), and in vivo BTDE also blocked intersegmental vessel formation in zebrafish embryos. Moreover, BTDE inhibited the migration, invasion, and vasculogenic mimicry formation of lung cancer cell A549. All these results indicated that BTDE could be used as a potential candidate in anti-angiogenesis for the treatment of cancer.
Collapse
Affiliation(s)
- Songtao Dong
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Zhongyuan Chen
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
| | - Li Wang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
| | - Yankai Liu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
| | - Dimitrios Stagos
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Biopolis, 41500 Larissa, Greece;
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, 319 Zhongshan Road, Jiangyang, Luzhou 646000, China;
| | - Ming Liu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
14
|
Lantsova A, Golubeva I, Borisova L, Nikolaeva L, Ektova L, Dmitrieva M, Orlova O. A new indolocarbazole derivative in melanoma and carcinoma lung in vivo treatment. BMC Complement Med Ther 2021; 21:117. [PMID: 33838683 PMCID: PMC8037905 DOI: 10.1186/s12906-021-03294-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/31/2021] [Indexed: 11/23/2022] Open
Abstract
Objective The current scientific research direction is development of drugs with a targeted effect on malignant tumors. One of the promising groups is indolocarbazoles and their derivatives, which can initiate various tumor cell death pathways. Russian scientists from N. N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation has developed a new experimental drug form of the original compound LCS 1269 with cytotoxic and antiangiogenic properties, blocking vasculogenic mimicry in tumor. The study aim is the experimental drug form LCS 1269 antitumor activity on models of transplantable mouse tumors B-16 melanoma and Lewis epidermoid lung carcinoma (LLC) with different routes and modes of administration. Material and methods Female F1 hybrid mice (C57Bl/6 x DBA/2) and male and female linear mice C57BL/6 were used for management of tumor strains. Mice were obtained from N. N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation vivarium. The antitumor effect was assessed by tumor growth inhibition (TGI) and increase of treated animal’s life span (ILS) compared to the control. Results The experimental drug form showed high antitumor activity when administered intravenously once at doses of 100 and 120 mg/kg (TGI = 98–82% and TGI = 95–77%, respectively, ILS = 24%, p < 0.05) on melanoma B-16 mice. On LLC mice, the experimental drug form showed that the intravenous administration route was effective in the range of doses from 60 to 80 mg/kg with a 5 day administration regimen with an interval of 24 h. A dose of 70 mg/kg had maximum effect at the level of TGI = 96–77% (p < 0.05) with its retention for 20 days after the end of treatment. Conclusion The studies have shown that the new compound LCS 1269 in the original drug form, has a pronounced antitumor activity and significantly reduces the volume of tumor mass both on melanoma B-16 and on LLC. It allows us to recommend continue the search for sensitivity of animal transplantable tumors to LCS 1269.
Collapse
Affiliation(s)
- Anna Lantsova
- Research Institute of Experimental Diagnostics and Therapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoe shosse, 24, 115478, Moscow, Russia
| | - Irina Golubeva
- Research Institute of Experimental Diagnostics and Therapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoe shosse, 24, 115478, Moscow, Russia
| | - Larisa Borisova
- Research Institute of Experimental Diagnostics and Therapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoe shosse, 24, 115478, Moscow, Russia
| | - Lyudmila Nikolaeva
- Research Institute of Experimental Diagnostics and Therapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoe shosse, 24, 115478, Moscow, Russia. .,Department of Pharmaceutical Technology and Pharmacology, Sechenov University, 8/2 Trubeckaya str., 119991, Moscow, Russia.
| | - Lydia Ektova
- Research Institute of Experimental Diagnostics and Therapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoe shosse, 24, 115478, Moscow, Russia
| | - Maria Dmitrieva
- Research Institute of Experimental Diagnostics and Therapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoe shosse, 24, 115478, Moscow, Russia
| | - Olga Orlova
- Research Institute of Experimental Diagnostics and Therapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology, Kashirskoe shosse, 24, 115478, Moscow, Russia
| |
Collapse
|
15
|
Piperigkou Z, Kyriakopoulou K, Koutsakis C, Mastronikolis S, Karamanos NK. Key Matrix Remodeling Enzymes: Functions and Targeting in Cancer. Cancers (Basel) 2021; 13:1441. [PMID: 33809973 PMCID: PMC8005147 DOI: 10.3390/cancers13061441] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue functionality and integrity demand continuous changes in distribution of major components in the extracellular matrices (ECMs) under normal conditions aiming tissue homeostasis. Major matrix degrading proteolytic enzymes are matrix metalloproteinases (MMPs), plasminogen activators, atypical proteases such as intracellular cathepsins and glycolytic enzymes including heparanase and hyaluronidases. Matrix proteases evoke epithelial-to-mesenchymal transition (EMT) and regulate ECM turnover under normal procedures as well as cancer cell phenotype, motility, invasion, autophagy, angiogenesis and exosome formation through vital signaling cascades. ECM remodeling is also achieved by glycolytic enzymes that are essential for cancer cell survival, proliferation and tumor progression. In this article, the types of major matrix remodeling enzymes, their effects in cancer initiation, propagation and progression as well as their pharmacological targeting and ongoing clinical trials are presented and critically discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| |
Collapse
|
16
|
Sedlmeier G, Al‐Rawi V, Buchert J, Yserentant K, Rothley M, Steshina A, Gräßle S, Wu R, Hurrle T, Richer W, Decraene C, Thiele W, Utikal J, Abuillan W, Tanaka M, Herten D, Hill CS, Garvalov BK, Jung N, Bräse S, Sleeman JP. Id1 and Id3 Are Regulated Through Matrix‐Assisted Autocrine BMP Signaling and Represent Therapeutic Targets in Melanoma. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Georg Sedlmeier
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Vanessa Al‐Rawi
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Justyna Buchert
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Klaus Yserentant
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- College of Medical and Dental Sciences & School of Chemistry University of Birmingham Birmingham UK
- Centre of Membrane Proteins and Receptors (COMPARE) Universities of Birmingham and Nottingham UK
| | - Melanie Rothley
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Anastasia Steshina
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Simone Gräßle
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Ruo‐Lin Wu
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Thomas Hurrle
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
| | - Wilfrid Richer
- CNRS UMR144 Translational Research Department Institut Curie PSL Research University 26 rue d'Ulm Paris Cedex 05 75248 France
| | - Charles Decraene
- CNRS UMR144 Translational Research Department Institut Curie PSL Research University 26 rue d'Ulm Paris Cedex 05 75248 France
| | - Wilko Thiele
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Jochen Utikal
- Skin Cancer Unit German Cancer Research Center (DKFZ) Im Neuenheimer Feld 280 69120 Heidelberg Germany
- Department of Dermatology, Venereology and Allergology University Medical Center Mannheim Ruprecht‐Karl University of Heidelberg Theodor‐Kutzer‐Ufer 1–3 68167 Mannheim Germany
| | - Wasim Abuillan
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
| | - Motomu Tanaka
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- Center for Integrative Medicine and Physics Institute for Advanced Study Kyoto University Yoshida Ushinomiya‐cho Sakyo‐Ku Kyoto 606‐8501 Japan
- Center for Integrative Medicine and Physics Institute for Advanced Study, Kyoto University Kyoto 606‐8501 Japan
| | - Dirk‐Peter Herten
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- College of Medical and Dental Sciences & School of Chemistry University of Birmingham Birmingham UK
- Centre of Membrane Proteins and Receptors (COMPARE) Universities of Birmingham and Nottingham UK
| | | | - Boyan K. Garvalov
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Nicole Jung
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Stefan Bräse
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Jonathan P. Sleeman
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| |
Collapse
|
17
|
Proliferation index and pseudoprogression as predictors of the therapeutic efficacy of suicide gene therapy for canine melanoma. Melanoma Res 2020; 30:126-135. [PMID: 32142496 DOI: 10.1097/cmr.0000000000000567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In our veterinary clinical trials, the combination of systemic immunotherapy with local herpes simplex virus thymidine kinase/ganciclovir suicide gene (SG) treatment induced tumor pseudoprogression as part of a strong local antitumor response. This phenomenon could be owing to tumor inflammation, increased vascular permeability and to different tumor growth rates before, during and after SG therapy. The proliferation index (PI: the fraction of viable cells in S, G2/M, and hyperdiploid phases) would reflect the in-vivo and in-vitro proportion of proliferating melanoma cells in the absence of treatment (PIB) or in response to SG (PISG). The extent of in-vivo and in-vitro melanoma cells responses to SG exhibited a reverse correlation with PIB and a direct correlation with PISG. Then, the final SG outcome depended on the balance between PIB-dependent 'regrowth resistance' versus 'regrowth sensitivity' to SG treatment. In all the cell lines derived from canine tumors presenting partial responses to SG treatment, PISG prevailed over PIB. Conversely, as more aggressive was the tumor (greater PIB of the cell line), the more the balance displacement towards 'regrowth resistance' over SG 'regrowth sensitivity'. All these parameters could have a prognostic value for SG treatment response and provide a glimpse at the clinical benefit of this therapy.
Collapse
|
18
|
Mabeta P. Paradigms of vascularization in melanoma: Clinical significance and potential for therapeutic targeting. Biomed Pharmacother 2020; 127:110135. [PMID: 32334374 DOI: 10.1016/j.biopha.2020.110135] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most aggressive form of skin cancer. Malignant melanoma in particular has a poor prognosis and although treatment has improved, drug resistance continues to be a challenge. Angiogenesis, the formation of blood vessels from existing microvessels, precedes the progression of melanoma from a radial growth phase to a malignant phenotype. In addition, melanoma cells can form networks of vessel-like fluid conducting channels through vasculogenic mimicry (VM). Both angiogenesis and VM have been postulated to contribute to the development of resistance to treatment and to enable metastasis. Also, the metastatic spread of melanoma is highly dependent on lymphangiogenesis, the formation of lymphatic vessels from pre-existing vessels. Interestingly, the design and clinical testing of drugs that target VM and lymphangiogenesis lag behind that of angiogenesis inhibitors. Despite this, antiangiogenic drugs have not significantly improved the overall survival of melanoma patients, thus necessitating the targeting of alternative mechanisms. In this article, I review the roles of the three paradigms of tissue perfusion, namely, angiogenesis, VM and lymphangiogenesis, in promoting melanoma progression and metastasis. This article also explores the latest development and potential opportunities in the therapeutic targeting of these processes.
Collapse
Affiliation(s)
- Peace Mabeta
- Angiogenesis Laboratory, Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
19
|
Zhang X, Zhang J, Zhou H, Liu G, Li Q. Rho kinase mediates transforming growth factor-β1-induced vasculogenic mimicry formation: involvement of the epithelial-mesenchymal transition and cancer stemness activity. Acta Biochim Biophys Sin (Shanghai) 2020; 52:411-420. [PMID: 32296834 DOI: 10.1093/abbs/gmaa014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/05/2020] [Accepted: 01/20/2020] [Indexed: 01/23/2023] Open
Abstract
Vasculogenic mimicry (VM), a newly defined pattern of tumor blood supply, has been identified in several malignant tumors, including hepatocellular carcinoma (HCC). Rho kinase (ROCK) plays an important role in various types of cancers. However, whether ROCK participates in transforming growth factor-β1 (TGF-β1)-induced VM formation is unclear. Here, we evaluated the role of ROCK in TGF-β1-induced VM formation in HCC. Our findings showed that the TGF-β1/ROCK signaling pathway is involved in VM formation by inducing the epithelial-mesenchymal transition. Furthermore, TGF-β1 and ROCK were found to play distinct roles in the cancer stem cell phenotype during VM formation. These results provide insights into potential antitumor therapies for inhibiting VM by targeting the TGF-β1/ROCK signaling pathway in HCC.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| | - Jigang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| | - Heming Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| | - Gaolin Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| | - Qin Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China
| |
Collapse
|
20
|
Zhu Y, Liu X, Zhao P, Zhao H, Gao W, Wang L. Celastrol Suppresses Glioma Vasculogenic Mimicry Formation and Angiogenesis by Blocking the PI3K/Akt/mTOR Signaling Pathway. Front Pharmacol 2020; 11:25. [PMID: 32116702 PMCID: PMC7025498 DOI: 10.3389/fphar.2020.00025] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
Angiogenesis and vasculogenic mimicry (VM) are thought to be the predominant processes ensuring tumor blood supply during the growth and metastasis of glioblastoma (GBM). Celastrol has potential anti-glioma effects, however the mechanisms underlying these effects remain unclarified. Recent studies have shown that the PI3K/Akt/mTOR signaling pathway is closely related to angiogenesis and VM formation. In the present study, we have demonstrated, for the first time, that celastrol eliminated VM formation by blocking this signaling pathway in glioma cells. By the treatment of celastrol, tumor growth was suppressed, tight junction and basal lamina structures in tumor microvasculature were disarranged in U87 glioma orthotopic xenografts in nude mice. Periodic acid Schiff (PAS)-CD31 staining revealed that celastrol inhibited both VM and angiogenesis in tumor tissues. Additionally, celastrol reduced the expression levels of the angiogenesis-related proteins CD31, vascular endothelial growth factor receptor (VEGFR) 2, angiopoietin (Ang) 2 and VEGFA, VM-related proteins ephrin type-A receptor (EphA) 2, and vascular endothelial (VE)-cadherin. Hypoxia inducible factor (HIF)-1α, phosphorylated PI3K, Akt, and mTOR were also downregulated by treatment with celastrol. In vitro, we further demonstrated that celastrol inhibited the growth, migration, and invasion of U87 and U251 cells, disrupted VM formation, and blocked the activity of PI3K, Akt, and mTOR. Collectively, our data suggest that celastrol inhibits VM formation and angiogenesis likely by regulating the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yingjun Zhu
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Xihong Liu
- Basic Discipline of Integrated Chinese and Western Medicine, Henan University of Chinese Medicine, Henan, China
| | - Peiyuan Zhao
- Basic Discipline of Integrated Chinese and Western Medicine, Henan University of Chinese Medicine, Henan, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China.,School of Pharmaceutical Sciences, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Heggannavar GB, Vijeth S, Kariduraganavar MY. Development of dual drug loaded PLGA based mesoporous silica nanoparticles and their conjugation with Angiopep-2 to treat glioma. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
22
|
López-Urrutia E, Bustamante Montes LP, Ladrón de Guevara Cervantes D, Pérez-Plasencia C, Campos-Parra AD. Crosstalk Between Long Non-coding RNAs, Micro-RNAs and mRNAs: Deciphering Molecular Mechanisms of Master Regulators in Cancer. Front Oncol 2019; 9:669. [PMID: 31404273 PMCID: PMC6670781 DOI: 10.3389/fonc.2019.00669] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer is a complex disease, and its study requires deep understanding of several biological processes and their regulation. It is an accepted fact that non-coding RNAs are vital components of the regulation and cross-talk among cancer-related signaling pathways that favor tumor aggressiveness and metastasis, such as neovascularization, angiogenesis, and vasculogenic mimicry. Both long non-coding RNAs (lncRNAs) and micro-RNAs (miRNAs) have been described as master regulators of cancer on their own; yet there is accumulating evidence that, besides regulating mRNA expression through independent mechanisms, these classes of non-coding RNAs interact with each other directly, fine-tuning the effects of their regulation. While still relatively scant, research on the lncRNA-miRNA-mRNA axis regulation is growing at a fast rate, it is only in the last 5 years, that lncRNA-miRNA interactions have been identified in tumor-related vascular processes. In this review, we summarize the current progress of research on the cross-talk between lncRNAs and miRNAs in the regulation of neovascularization, angiogenesis and vasculogenic mimicry.
Collapse
Affiliation(s)
- Eduardo López-Urrutia
- Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | | | | | - Carlos Pérez-Plasencia
- Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico.,Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Alma D Campos-Parra
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| |
Collapse
|
23
|
Sandomenico A, Ruvo M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr Med Chem 2019; 26:1994-2050. [PMID: 30207211 DOI: 10.2174/0929867325666180912104707] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Elucidating the mechanisms of recurrence of embryonic signaling pathways in tumorigenesis has led to the discovery of onco-fetal players which have physiological roles during normal development but result aberrantly re-activated in tumors. In this context, Nodal and Cripto-1 are recognized as onco-developmental factors, which are absent in normal tissues but are overexpressed in several solid tumors where they can serve as theranostic agents. OBJECTIVE To collect, review and discuss the most relevant papers related to the involvement of Nodal and Cripto-1 in the development, progression, recurrence and metastasis of several tumors where they are over-expressed, with a particular attention to their occurrence on the surface of the corresponding sub-populations of cancer stem cells (CSC). RESULTS We have gathered, rationalized and discussed the most interesting findings extracted from some 370 papers related to the involvement of Cripto-1 and Nodal in all tumor types where they have been detected. Data demonstrate the clear connection between Nodal and Cripto-1 presence and their multiple oncogenic activities across different tumors. We have also reviewed and highlighted the potential of targeting Nodal, Cripto-1 and the complexes that they form on the surface of tumor cells, especially of CSC, as an innovative approach to detect and suppress tumors with molecules that block one or more mechanisms that they regulate. CONCLUSION Overall, Nodal and Cripto-1 represent two innovative and effective biomarkers for developing potential theranostic anti-tumor agents that target normal as well as CSC subpopulations and overcome both pharmacological resistance and tumor relapse.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| |
Collapse
|
24
|
Molecular background of skin melanoma development and progression: therapeutic implications. Postepy Dermatol Alergol 2019; 36:129-138. [PMID: 31320844 PMCID: PMC6627250 DOI: 10.5114/ada.2019.84590] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/18/2018] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the most aggressive skin cancer with an increasing number of cases worldwide and curable mostly in its early stage. The improvement in patients' survival in advanced melanoma has been achieved only recently, due to development of new biological drugs for targeted therapies and immunotherapy. Further progress in the treatment of melanoma is clearly dependent on the better understanding of its complex biology. This review describes the most important molecular mechanisms and genetic events underlying skin melanoma development and progression, depicts the way of action of newly developed drugs and indicates new potential therapeutic targets.
Collapse
|
25
|
Sun H, Yao N, Cheng S, Li L, Liu S, Yang Z, Shang G, Zhang D, Yao Z. Cancer stem-like cells directly participate in vasculogenic mimicry channels in triple-negative breast cancer. Cancer Biol Med 2019; 16:299-311. [PMID: 31516750 PMCID: PMC6713644 DOI: 10.20892/j.issn.2095-3941.2018.0209] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective Vasculogenic mimicry (VM) channels that are lined by tumor cells are a functional blood supply in malignant tumors. However, the role of VM-initiating cells remains poorly understood. Cancer stem-like cells (CSCs) are positively correlated with VM. In this study, triple-negative breast cancer (TNBC) enriched with CSCs was used to investigate the relationship between VM and CSCs. Methods The expression of several CSC markers was detected by immunohistochemistry in 100 human breast cancer samples. The clinical significance of CSC markers and the relationship between VM, CSCs, breast cancer subtypes, and VM-associated proteins were analyzed. CD133+ and ALDH+ human and mouse TNBC cells were isolated by FACS to examine the ability of VM formation and the spatial relationship between VM and CSCs. Results CSCs were associated with TNBC subtype and VM in human invasive breast cancer. CSCs in TNBC MDA-MB-231 cells formed more VM channels and expressed more molecules promoting VM than the non-TNBC MCF-7 cells in vitro. MDA-MB-231 cells that encircled VM channels on Matrigel expressed CD133. Moreover, CSCs were located near VM channels in the 3D reconstructed blood supply system in human TNBC grafts. The CD133+ and ALDH+ cells isolated from TA2 mouse breast cancer formed more VM channels in vivo.
Conclusions CSCs line VM channels directly. Additionally, CSCs provide more VM-related molecules to synergize VM formation. The signaling pathways that control CSC differentiation may also be potential treatment targets for TNBC.
Collapse
Affiliation(s)
- Huizhi Sun
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Nan Yao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Siqi Cheng
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Linqi Li
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Shiqi Liu
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Zhao Yang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Guanjie Shang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin 300070, China
| | - Zhi Yao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, China.,Department of Immunology, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
26
|
Xia Y, Cai XY, Fan JQ, Zhang LL, Ren JH, Li ZY, Zhang RG, Zhu F, Wu G. The role of sema4D in vasculogenic mimicry formation in non-small cell lung cancer and the underlying mechanisms. Int J Cancer 2018; 144:2227-2238. [PMID: 30374974 DOI: 10.1002/ijc.31958] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/06/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
Abstract
Vasculogenic mimicry (VM) is a special vascular pattern in malignant tumors, which is composed of highly aggressive tumor cells. This tumor cell-mediated blood supply pattern is closely associated with a poor prognosis in cancer patients. The interaction of axon guidance factor Sema4D and its high affinity receptor plexinB1 could activate small GTPase RhoA and its downstream ROCKs; this process has an active role in the migration of endothelial cells and tumor angiogenesis. Here, we have begun to uncover the role of this pathway in VM formation in non-small cell lung cancer (NSCLC). First, we confirmed this special form of vasculature in NSCLC tissues and found the existence of VM channels in tumor tissues was correlated with Sema4D expression. Further, we found that inhibition of Sema4D in the human NSCLC cells H1299 and HCC827 reduces VM formation both in vitro and in vivo. Moreover, we demonstrated that downregulating the expression of plexinB1 by siRNA expressing vectors and inhibiting the RhoA/ROCK signaling pathway using fasudil can reduce VM formation of H1299 and HCC827 cells. Finally, we found that suppression of Sema4D leads to less stress fibers and depleted the motility of H1299 and HCC827 cells. Collectively, our study implicates Sema4D plays an important role in the process of VM formation in NSCLC through activating the RhoA/ROCK pathway and regulating tumor cell plasticity and migration. Modulation of the Sema4D/plexinB1 and downstream RhoA/ROCK pathway may prevent the tumor blood supply through the VM pattern, which may eventually halt growth and metastasis of NSCLC.
Collapse
Affiliation(s)
- Yun Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian-Yi Cai
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Quan Fan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Ling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing-Hua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Yu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui-Guang Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Shao B, Zhao X, Liu T, Zhang Y, Sun R, Dong X, Liu F, Zhao N, Zhang D, Wu L, Wang Y, Wang M, Meng J, Lin X, Sun B. LOXL2 promotes vasculogenic mimicry and tumour aggressiveness in hepatocellular carcinoma. J Cell Mol Med 2018; 23:1363-1374. [PMID: 30506621 PMCID: PMC6349148 DOI: 10.1111/jcmm.14039] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/11/2018] [Accepted: 10/28/2018] [Indexed: 12/11/2022] Open
Abstract
Lysyl oxidase‐like 2 (LOXL2) has shown to promote metastasis and poor prognosis in hepatocellular carcinoma (HCC). Also, we have previously reported that vasculogenic mimicry (VM) is associated with invasion, metastasis and poor survival in HCC patients. In the present study, we investigated molecular function of LOXL2 in HCC and VM. We used the immunohistochemical and CD31/periodic acid‐Schiff double staining to detect the relationship between LOXL2 and VM formation. We performed the gain and loss of function studies and analysed the migratory, invasion and tube formation in HCC cell lines. We analysed the function of LOXL2 in VM formation and HCC metastasis both in vitro and in vivo. We have showed that LOXL2 was overexpression in HCC and was positively correlated with tumour grade, metastasis, VM formation and poor survival in 201 HCC patients. Secondly, our studies have showed that LOXL2 overexpression in HCC cells significantly promoted migration, invasion and tube formation. Finally, we found that LOXL2 may increase SNAIL expression, thereby enabling VM. Our study indicated that LOXL2 may promote VM formation and tumour metastasis by collaborating with SNAIL in HCC. What's more, the overexpression of LOXL2 indicated a poor prognosis in HCC patients.
Collapse
Affiliation(s)
- Bing Shao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Tieju Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Yanhui Zhang
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China
| | - Ran Sun
- Tianjin Nankai Hospital, Tianjin, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Fang Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Lili Wu
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Yong Wang
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Meili Wang
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Jie Meng
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Xian Lin
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
28
|
Wijeratne D, Rodger J, Stevenson A, Wallace H, Prêle CM, Wood FM, Fear MW. Ephrin-A2 affects wound healing and scarring in a murine model of excisional injury. Burns 2018; 45:682-690. [PMID: 30482614 DOI: 10.1016/j.burns.2018.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/14/2018] [Accepted: 10/04/2018] [Indexed: 12/18/2022]
Abstract
Ephrin ligand/Eph receptor signaling is important in both tissue development and homeostasis. There is increasing evidence that Ephrin/Eph signaling is important in the skin, involved in hair follicle cycling, epidermal differentiation, cutaneous innervation and skin cancer. However, there is currently limited information on the role of Ephrin/Eph signaling in cutaneous wound healing. Here we report the effects of the Ephrin-A2 and A5 ligands on wound healing. Using Ephrin-A2-/-, Ephrin-A5-/- and Ephrin-A2A5-/- transgenic mice, in vitro wound healing assays were conducted using isolated keratinocytes and fibroblasts. Ephrin-A2-/-, Ephrin-A2A5-/- and wild type mice with excisional wounds were used to analyze the impact of these ligands on wound closure, scar outcome, collagen orientation and re-innervation in vivo. The absence of the Ephrin-A2 and A5 ligands did not have any effect on dermal fibroblast proliferation or on fibroblast or keratinocyte migration. The loss of Ephrin-A2 and A5 ligands did not impact on the rate of wound closure or re-innervation after injury. However, changes in the gross morphology of the healed scar and in collagen histology of the scar dermis were observed in transgenic mice. Therefore Ephrin-A2 and A5 ligands may play an important role in final scar appearance associated with collagen deposition and structure.
Collapse
Affiliation(s)
- Dulharie Wijeratne
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia
| | - Jennifer Rodger
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Australia
| | - Andrew Stevenson
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia
| | - Hilary Wallace
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia
| | - Cecilia M Prêle
- The Institute for Respiratory Health, The University of Western Australia, Nedlands, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, Australia; The Fiona Wood Foundation, Perth, Western Australia, Australia; Burns Service of Western Australia, WA Department of Health, Perth, Western Australia, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Australia; The Institute for Respiratory Health, The University of Western Australia, Nedlands, Western Australia, Australia; The Fiona Wood Foundation, Perth, Western Australia, Australia.
| |
Collapse
|
29
|
Mertz DR, Ahmed T, Takayama S. Engineering cell heterogeneity into organs-on-a-chip. LAB ON A CHIP 2018; 18:2378-2395. [PMID: 30040104 PMCID: PMC6081245 DOI: 10.1039/c8lc00413g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Organ-on-a-chip development is an application that will benefit from advances in cell heterogeneity characterization because these culture models are intended to mimic in vivo microenvironments, which are complex and dynamic. Due in no small part to advances in microfluidic single cell analysis methods, cell-to-cell variability is an increasingly understood feature of physiological tissues, with cell types from as common as 1 out of every 2 cells to as rare as 1 out of every 100 000 cells having important roles in the biochemical and biological makeup of tissues and organs. Variability between neighboring cells can be transient or maintained, and ordered or stochastic. This review covers three areas of well-studied cell heterogeneity that are informative for organ-on-a-chip development efforts: tumors, the lung, and the intestine. Then we look at how recent single cell analysis strategies have enabled better understanding of heterogeneity within in vitro and in vivo tissues. Finally, we provide a few work-arounds for adapting current on-chip culture methods to better mimic physiological cell heterogeneity including accounting for crucial rare cell types and events.
Collapse
Affiliation(s)
- David R Mertz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering and Emory School of Medicine, USA.
| | | | | |
Collapse
|
30
|
Xiao T, Zhang Q, Zong S, Zhong WL, Qin Y, Bi Z, Chen S, Liu HJ, Wei JJ, Zhou BJ, Wang LM, Zhou HG, Liu YR, Sun T, Yang C. Protease-activated receptor-1 (PAR1) promotes epithelial-endothelial transition through Twist1 in hepatocellular carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:185. [PMID: 30081924 PMCID: PMC6091192 DOI: 10.1186/s13046-018-0858-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/12/2018] [Indexed: 01/13/2023]
Abstract
Background Tumor cells transfer into endothelial cells by epithelial–endothelial transition (EET), which is characterized by vasculagenic mimicry (VM) in morphology. VM can change tumor microcirculation, progression, and metastasis. However, the molecular mechanisms of endothelial-like transition remain unclear. EET is a subtype of epithelial–mesenchymal transition (EMT). Twist1, a transcriptional regulatory factor of EMT, is an important factor that induces EET in hepatocellular carcinoma(HCC), but the upstream signal of Twist1 is unclear. Methods Expression plasmids, Ca mobilization, and three-dimensional cultures were evaluated. Western blot assay, reporter gene assay, and immunofluorescence staining were conducted. A murine xenograft model was established. Analyses of immunohistochemistry, patient samples, and complementary DNA (cDNA) microarrays were also performed. Results This study demonstrated that protease-activated receptor-1 (PAR1) can increase the expression of endothelial markers and enhance VM formation by upregulating Twist1 both in vitro and in vivo through thrombin binding. Thrombin not only activates PAR1 but also promotes PAR1 internalization in a time-dependent manner. Clinical pathological analysis further confirms that PAR1 expression is directly correlated with the endothelial marker expression, VM formation, and metastasis and indicates poor survival rate of patients with tumors. Conclusion PAR1 promotes EET through Twist1 in HCC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0858-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ting Xiao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Qiang Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Shumin Zong
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Wei-Long Zhong
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Yuan Qin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Zhun Bi
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Shuang Chen
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Hui-Juan Liu
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Jun-Jie Wei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Bi-Jiao Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Lu-Meng Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Hong-Gang Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China
| | - Yan-Rong Liu
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China.
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China. .,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300350, China. .,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, No. 220 Dongting Road, Binhai District, Tianjin, 300457, China.
| |
Collapse
|
31
|
Heggannavar G, Hiremath CG, Achari DD, Pangarkar VG, Kariduraganavar MY. Development of Doxorubicin-Loaded Magnetic Silica-Pluronic F-127 Nanocarriers Conjugated with Transferrin for Treating Glioblastoma across the Blood-Brain Barrier Using an in Vitro Model. ACS OMEGA 2018; 3:8017-8026. [PMID: 30087932 PMCID: PMC6072239 DOI: 10.1021/acsomega.8b00152] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/05/2018] [Indexed: 05/14/2023]
Abstract
Brain glioma is the most lethal type of cancer, with extremely poor prognosis and high relapse. Unfortunately, the treatment of brain glioma is often limited because of the low permeability of anticancer drugs across the blood-brain barrier (BBB). To circumvent this, magnetic mesoporous nanoparticles were synthesized and loaded with doxorubicin as an anticancer agent. These nanoparticles were fabricated with Pluronic F-127 and subsequently conjugated with transferrin (Tf) to achieve the sustained release of the drug at the targeted site. The physicochemical properties of the conjugated nanoparticles were analyzed using different techniques. The magnetic saturation of the nanoparticles determined by a vibration sample magnetometer was found to be 26.10 emu/g. The cytotoxicity study was performed using the MTT assay at 48 and 96 h against the U87 cell line. The Tf-conjugated nanoparticles (DOX-MNP-MSN-PF-127-Tf) exhibited a significant IC50 value (0.570 μg/mL) as compared to the blank nanoparticles (121.98 μg/mL). To understand the transport mechanism of drugs across the BBB, an in vitro BBB model using human brain microvascular endothelial cells was developed. Among the nanoparticles, the Tf-conjugated nanoparticles demonstrated an excellent permeability across the BBB. This effect was predominant in the presence of an external magnetic field, suggesting that magnetic particles present in the matrix facilitated the uptake of drugs in U87 cells. Finally, it is concluded that nanoparticles conjugated with Tf effectively crossed the BBB. Thus, the developed nanocarriers can be considered as potential candidates to treat brain tumor.
Collapse
Affiliation(s)
| | - Chinmay G. Hiremath
- Department
of Studies in Chemistry, Karnatak University, Dharwad 580 003, India
| | - Divya D. Achari
- Department
of Studies in Chemistry, Karnatak University, Dharwad 580 003, India
| | | | | |
Collapse
|
32
|
Balestrieri E, Argaw-Denboba A, Gambacurta A, Cipriani C, Bei R, Serafino A, Sinibaldi-Vallebona P, Matteucci C. Human Endogenous Retrovirus K in the Crosstalk Between Cancer Cells Microenvironment and Plasticity: A New Perspective for Combination Therapy. Front Microbiol 2018; 9:1448. [PMID: 30013542 PMCID: PMC6036167 DOI: 10.3389/fmicb.2018.01448] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/11/2018] [Indexed: 12/24/2022] Open
Abstract
Abnormal activation of human endogenous retroviruses (HERVs) has been associated with several diseases such as cancer, autoimmunity, and neurological disorders. In particular, in cancer HERV activity and expression have been specifically associated with tumor aggressiveness and patient outcomes. Cancer cell aggressiveness is intimately linked to the acquisition of peculiar plasticity and heterogeneity based on cell stemness features, as well as on the crosstalk between cancer cells and the microenvironment. The latter is a driving factor in the acquisition of aggressive phenotypes, associated with metastasis and resistance to conventional cancer therapies. Remarkably, in different cell types and stages of development, HERV expression is mainly regulated by epigenetic mechanisms and is subjected to a very precise temporal and spatial regulation according to the surrounding microenvironment. Focusing on our research experience with HERV-K involvement in the aggressiveness and plasticity of melanoma cells, this perspective aims to highlight the role of HERV-K in the crosstalk between cancer cells and the tumor microenvironment. The implications for a combination therapy targeted at HERVs with standard approaches are discussed.
Collapse
Affiliation(s)
- Emanuela Balestrieri
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | - Ayele Argaw-Denboba
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | - Alessandra Gambacurta
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | - Chiara Cipriani
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Annalucia Serafino
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
33
|
Wang H, Huang B, Li BM, Cao KY, Mo CQ, Jiang SJ, Pan JC, Wang ZR, Lin HY, Wang DH, Qiu SP. ZEB1-mediated vasculogenic mimicry formation associates with epithelial-mesenchymal transition and cancer stem cell phenotypes in prostate cancer. J Cell Mol Med 2018; 22:3768-3781. [PMID: 29754422 PMCID: PMC6050489 DOI: 10.1111/jcmm.13637] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/10/2018] [Indexed: 01/18/2023] Open
Abstract
The zinc finger E‐box‐binding homeobox 1 (ZEB1) induced the epithelial–mesenchymal transition (EMT) and altered ZEB1 expression could lead to aggressive and cancer stem cell (CSC) phenotypes in various cancers. Tissue specimens from 96 prostate cancer patients were collected for immunohistochemistry and CD34/periodic acid–Schiff double staining. Prostate cancer cells were subjected to ZEB1 knockdown or overexpression and assessment of the effects on vasculogenic mimicry formation in vitro and in vivo. The underlying molecular events of ZEB1‐induced vasculogenic mimicry formation in prostate cancer were then explored. The data showed that the presence of VM and high ZEB1 expression was associated with higher Gleason score, TNM stage, and lymph node and distant metastases as well as with the expression of vimentin and CD133 in prostate cancer tissues. Furthermore, ZEB1 was required for VM formation and altered expression of EMT‐related and CSC‐associated proteins in prostate cancer cells in vitro and in vivo. ZEB1 also facilitated tumour cell migration, invasion and clonogenicity. In addition, the effects of ZEB1 in prostate cancer cells were mediated by Src signalling; that is PP2, a specific inhibitor of the Src signalling, dose dependently reduced the p‐Src527 level but not p‐Src416 level, while ZEB1 knockdown also down‐regulated the level of p‐Src527 in PC3 and DU‐145 cells. PP2 treatment also significantly reduced the expression of VE‐cadherin, vimentin and CD133 in these prostate cancer cells. Src signalling mediated the effects of ZEB1 on VM formation and gene expression.
Collapse
Affiliation(s)
- Hua Wang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bin Huang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bai Mou Li
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Kai Yuan Cao
- Research Center for Clinical Laboratory Standard, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chen Qiang Mo
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuang Jian Jiang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jin Cheng Pan
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zong Ren Wang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huan Yi Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dao Hu Wang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shao Peng Qiu
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Department of Urology, Hui Ya hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
34
|
Sun B, Zhang D, Zhao N, Zhao X. Epithelial-to-endothelial transition and cancer stem cells: two cornerstones of vasculogenic mimicry in malignant tumors. Oncotarget 2018; 8:30502-30510. [PMID: 27034014 PMCID: PMC5444760 DOI: 10.18632/oncotarget.8461] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/14/2016] [Indexed: 01/11/2023] Open
Abstract
Vasculogenic mimicry (VM) is a functional microcirculation pattern in malignant tumors accompanied by endothelium-dependent vessels and mosaic vessels. VM has been identified in more than 15 solid tumor types and is associated with poor differentiation, late clinical stage and poor prognosis. Classic anti-angiogenic agents do not target endothelium-dependent vessels and are not efficacious against tumors exhibiting VM. Further insight into the molecular signaling that triggers and promotes VM formation could improve anti-angiogenic therapeutics. Recent studies have shown that cancer stem cells (CSCs) and epithelium-to-endothelium transition (EET), a subtype of epithelial-to-mesenchymal transition (EMT), accelerate VM formation by stimulating tumor cell plasticity, remodeling the extracellular matrix (ECM) and connecting VM channels with host blood vessels. VM channel-lining cells originate from CSCs due to expression of EMT inducers such as Twist1, which promote EET and ECM remodeling. Hypoxia and high interstitial fluid pressure in the tumor microenvironment induce a specific type of cell death, linearly patterned programmed cell necrosis (LPPCN), which spatially guides VM and endothelium-dependent vessel networks. This review focuses on the roles of CSCs and EET in VM, and on possible novel anti-angiogenic strategies against alternative tumor vascularization.
Collapse
Affiliation(s)
- Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China.,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
35
|
Etoposide-Bevacizumab a new strategy against human melanoma cells expressing stem-like traits. Oncotarget 2018; 7:51138-51149. [PMID: 27303923 PMCID: PMC5239464 DOI: 10.18632/oncotarget.9939] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 05/01/2016] [Indexed: 12/20/2022] Open
Abstract
Tumors contain a sub-population of self-renewing and expanding cells known as cancer stem cells (CSCs). Putative CSCs were isolated from human melanoma cells of a different aggressiveness, Hs294T and A375 cell lines, grown under hypoxia using “sphere-forming assay”, CD133 surface expression and migration ability. We found that a cell sub-population enriched for P1 sphere-initiating ability and CD133 expression also express larger amount of VEGF-R2. Etoposide does not influence phenotype of this sub-population of melanoma cells, while a combined treatment with Etoposide and Bevacizumab significantly abolished P1 sphere-forming ability, an effect associated with apoptosis of this subset of cells. Hypoxic melanoma cells sorted for VEGF-R2/CD133 positivity also undergo apoptosis when exposed to Etoposide and Bevacizumab. When Etoposide and Bevacizumab-treated hypoxic cells were injected intravenously into immunodeficient mice revealed a reduced capacity to induce lung colonies, which also appear with a longer latency period. Hence, our study indicates that a combined exposure to Etoposide and Bevacizumab targets melanoma cells endowed with stem-like properties and might be considered a novel approach to treat cancer-initiating cells.
Collapse
|
36
|
Flores-Téllez TNJ, Villa-Treviño S, Piña-Vázquez C. Road to stemness in hepatocellular carcinoma. World J Gastroenterol 2017; 23:6750-6776. [PMID: 29085221 PMCID: PMC5645611 DOI: 10.3748/wjg.v23.i37.6750] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/27/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
Carcinogenic process has been proposed to relay on the capacity to induce local tissue damage and proliferative repair. Liver has a great regeneration capacity and currently, most studies point towards the dominant role of hepatocytes in regeneration at all levels of liver damage. The most frequent liver cancer is hepatocellular carcinoma (HCC). Historical findings originally led to the idea that the cell of origin of HCC might be a progenitor cell. However, current linage tracing studies put the progenitor hypothesis of HCC origin into question. In agreement with their dominant role in liver regeneration, mature hepatocytes are emerging as the cell of origin of HCC, although, the specific hepatocyte subpopulation of origin is yet to be determined. The relationship between the cancer cell of origin (CCO) and cancer-propagating cells, known as hepatic cancer stem cell (HCSC) is unknown. It has been challenging to identify the definitive phenotypic marker of HCSC, probably due to the existence of different cancer stem cells (CSC) subpopulations with different functions within HCC. There is a dynamic interconversion among different CSCs, and between CSC and non-CSCs. Because of that, CSC-state is currently defined as a description of a highly adaptable and dynamic intrinsic property of tumor cells, instead of a static subpopulation of a tumor. Altered conditions could trigger the gain of stemness, some of them include: EMT-MET, epigenetics, microenvironment and selective stimulus such as chemotherapy. This CSC heterogeneity and dynamism makes them out reach from therapeutic protocols directed to a single target. A further avenue of research in this line will be to uncover mechanisms that trigger this interconversion of cell populations within tumors and target it.
Collapse
Affiliation(s)
- Teresita NJ Flores-Téllez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN No. 2508 Col. San Pedro Zacatenco CP 07360, Ciudad de México, México
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN No. 2508 Col. San Pedro Zacatenco CP 07360, Ciudad de México, México
| | - Carolina Piña-Vázquez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN No. 2508 Col. San Pedro Zacatenco CP 07360, Ciudad de México, México
| |
Collapse
|
37
|
Zhou X, Gu R, Han X, Wu G, Liu J. Cyclin-dependent kinase 5 controls vasculogenic mimicry formation in non-small cell lung cancer via the FAK-AKT signaling pathway. Biochem Biophys Res Commun 2017; 492:447-452. [DOI: 10.1016/j.bbrc.2017.08.076] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/20/2017] [Indexed: 12/14/2022]
|
38
|
RhoA/ROCK pathway inhibition by fasudil suppresses the vasculogenic mimicry of U2OS osteosarcoma cells in vitro. Anticancer Drugs 2017; 28:514-521. [DOI: 10.1097/cad.0000000000000490] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Sun J, Sun B, Sun R, Zhu D, Zhao X, Zhang Y, Dong X, Che N, Li J, Liu F, Zhao N, Wang Y, Zhang D. HMGA2 promotes vasculogenic mimicry and tumor aggressiveness by upregulating Twist1 in gastric carcinoma. Sci Rep 2017; 7:2229. [PMID: 28533522 PMCID: PMC5440402 DOI: 10.1038/s41598-017-02494-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 04/12/2017] [Indexed: 01/29/2023] Open
Abstract
High mobility group protein A2 (HMGA2) is a transcription factor that plays an important role in the invasion and metastasis of gastric carcinoma (GC). The term vasculogenic mimicry (VM) refers to the unique ability of aggressive tumour cells to mimic the pattern of embryonic vasculogenic networks. However, the relationship between HMGA2 and VM formation remains unclear. In the present study, we examined concomitant HMGA2 expression and VM in 228 human GC samples and 4 GC cell lines. Our data indicate that HMGA2 is not only significantly associated with VM formation but also influences the prognosis of patients with gastric carcinoma. Overexpression of HMGA2 significantly increased cell motility, invasiveness, and VM formation both in vitro and in vivo. A luciferase reporter assay, Co-IP and ChIP demonstrated that HMGA2 induced the expression of Twist1 and VE-cadherin by binding to the Twist1 promoter. Moreover, we observed a decrease in VE-cadherin following Twist1 knockdown in cells overexpressing HMGA2. This study indicates that HMGA2 promotes VM in GC via Twist1-VE-cadherin signalling and influences the prognosis of patients with GC.
Collapse
Affiliation(s)
- Junying Sun
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China. .,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China. .,Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060, China.
| | - Ran Sun
- Department of Surgery, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Dongwang Zhu
- Department of Prosthodontics, Affiliated Stomatological Hospital, Tianjin Medical University, Tianjin, 300070, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yanhui Zhang
- Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Na Che
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Jing Li
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Fang Liu
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yong Wang
- Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052, China
| |
Collapse
|
40
|
Bodenstine TM, Chandler GS, Seftor REB, Seftor EA, Hendrix MJC. Plasticity underlies tumor progression: role of Nodal signaling. Cancer Metastasis Rev 2016; 35:21-39. [PMID: 26951550 DOI: 10.1007/s10555-016-9605-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The transforming growth factor beta (TGFβ) superfamily member Nodal is an established regulator of early embryonic development, with primary roles in endoderm induction, left-right asymmetry, and primitive streak formation. Nodal signals through TGFβ family receptors at the plasma membrane and induces signaling cascades leading to diverse transcriptional regulation. While conceptually simple, the regulation of Nodal and its molecular effects are profoundly complex and context dependent. Pioneering work by developmental biologists has characterized the signaling pathways, regulatory components, and provided detailed insight into the mechanisms by which Nodal mediates changes at the cellular and organismal levels. Nodal is also an important factor in maintaining pluripotency of embryonic stem cells through regulation of core transcriptional programs. Collectively, this work has led to an appreciation for Nodal as a powerful morphogen capable of orchestrating multiple cellular phenotypes. Although Nodal is not active in most adult tissues, its reexpression and signaling have been linked to multiple types of human cancer, and Nodal has emerged as a driver of tumor growth and cellular plasticity. In vitro and in vivo experimental evidence has demonstrated that inhibition of Nodal signaling reduces cancer cell aggressive characteristics, while clinical data have established associations with Nodal expression and patient outcomes. As a result, there is great interest in the potential targeting of Nodal activity in a therapeutic setting for cancer patients that may provide new avenues for suppressing tumor growth and metastasis. In this review, we evaluate our current understanding of the complexities of Nodal function in cancer and highlight recent experimental evidence that sheds light on the therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Thomas M Bodenstine
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Grace S Chandler
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Richard E B Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Elisabeth A Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Mary J C Hendrix
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA.
| |
Collapse
|
41
|
Abstract
Malignant melanoma of the skin is the most aggressive human cancer given that a primary tumor a few millimeters in diameter frequently has full metastatic competence. In view of that, revealing the genetic background of this potential may also help to better understand tumor dissemination in general. Genomic analyses have established the molecular classification of melanoma based on the most frequent driver oncogenic mutations (BRAF, NRAS, KIT) and have also revealed a long list of rare events, including mutations and amplifications as well as genetic microheterogeneity. At the moment, it is unclear whether any of these rare events have role in the metastasis initiation process since the major drivers do not have such a role. During lymphatic and hematogenous dissemination, the clonal selection process is evidently reflected by differences in oncogenic drivers in the metastases versus the primary tumor. Clonal selection is also evident during lymphatic progression, though the genetic background of this immunoselection is less clear. Genomic analyses of metastases identified further genetic alterations, some of which may correspond to metastasis maintenance genes. The natural genetic progression of melanoma can be modified by targeted (BRAF or MEK inhibitor) or immunotherapies. Some of the rare events in primary tumors may result in primary resistance, while further new genetic lesions develop during the acquired resistance to both targeted and immunotherapies. Only a few genetic lesions of the primary tumor are constant during natural or therapy-modulated progression. EGFR4 and NMDAR2 mutations, MITF and MET amplifications and PTEN loss can be considered as metastasis drivers. Furthermore, BRAF and MITF amplifications as well as PTEN loss are also responsible for resistance to targeted therapies, whereas NRAS mutation is the only founder genetic lesion showing any association with sensitivity to immunotherapies. Unfortunately, there are hardly any data on the possible organ-specific metastatic drivers in melanoma. These observations suggest that clinical management of melanoma patients must rely on the genetic analysis of the metastatic lesions to be able to monitor progression-associated changes and to personalize therapies.
Collapse
|
42
|
Williamson SC, Metcalf RL, Trapani F, Mohan S, Antonello J, Abbott B, Leong HS, Chester CPE, Simms N, Polanski R, Nonaka D, Priest L, Fusi A, Carlsson F, Carlsson A, Hendrix MJC, Seftor REB, Seftor EA, Rothwell DG, Hughes A, Hicks J, Miller C, Kuhn P, Brady G, Simpson KL, Blackhall FH, Dive C. Vasculogenic mimicry in small cell lung cancer. Nat Commun 2016; 7:13322. [PMID: 27827359 PMCID: PMC5105195 DOI: 10.1038/ncomms13322] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/22/2016] [Indexed: 02/08/2023] Open
Abstract
Small cell lung cancer (SCLC) is characterized by prevalent circulating tumour cells (CTCs), early metastasis and poor prognosis. We show that SCLC patients (37/38) have rare CTC subpopulations co-expressing vascular endothelial-cadherin (VE-cadherin) and cytokeratins consistent with vasculogenic mimicry (VM), a process whereby tumour cells form 'endothelial-like' vessels. Single-cell genomic analysis reveals characteristic SCLC genomic changes in both VE-cadherin-positive and -negative CTCs. Higher levels of VM are associated with worse overall survival in 41 limited-stage patients' biopsies (P<0.025). VM vessels are also observed in 9/10 CTC patient-derived explants (CDX), where molecular analysis of fractionated VE-cadherin-positive cells uncovered copy-number alterations and mutated TP53, confirming human tumour origin. VE-cadherin is required for VM in NCI-H446 SCLC xenografts, where VM decreases tumour latency and, despite increased cisplatin intra-tumour delivery, decreases cisplatin efficacy. The functional significance of VM in SCLC suggests VM regulation may provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Stuart C. Williamson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Robert L. Metcalf
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Francesca Trapani
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Sumitra Mohan
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Jenny Antonello
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Benjamin Abbott
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Hui Sun Leong
- Computational Biology Support Team, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Christopher P. E. Chester
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Nicole Simms
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Radoslaw Polanski
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Daisuke Nonaka
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Lynsey Priest
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Alberto Fusi
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Fredrika Carlsson
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Anders Carlsson
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Mary J. C. Hendrix
- Stanley Manne Children's Research Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Richard E. B. Seftor
- Stanley Manne Children's Research Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Elisabeth A. Seftor
- Stanley Manne Children's Research Institute, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Dominic G. Rothwell
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Andrew Hughes
- The Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
| | - James Hicks
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Crispin Miller
- RNA Biology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Peter Kuhn
- University of Southern California Dornsife, Los Angeles, California 90089-3301, USA
| | - Ged Brady
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Kathryn L. Simpson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
| | - Fiona H. Blackhall
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
- The Institute of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
- Cancer Research UK, Lung Cancer Centre of Excellence, Manchester M20 4BX, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester M20 4BX, UK
- Cancer Research UK, Lung Cancer Centre of Excellence, Manchester M20 4BX, UK
| |
Collapse
|
43
|
The combination of bleomycin with suicide or interferon-β gene transfer is able to efficiently eliminate human melanoma tumor initiating cells. Biomed Pharmacother 2016; 83:290-301. [PMID: 27399807 DOI: 10.1016/j.biopha.2016.06.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/23/2016] [Accepted: 06/16/2016] [Indexed: 12/30/2022] Open
Abstract
We explored the potential of a chemogene therapy combination to eradicate melanoma tumor initiating cells, key producers of recurrence and metastatic spread. Three new human melanoma cell lines, two obtained from lymph nodes and one from spleen metastasis were established and characterized. They were cultured as monolayers and spheroids and, in both spatial configurations they displayed sensitivity to single treatments with bleomycin (BLM) or human interferon-β (hIFNβ) gene or herpes simplex virus thymidine kinase/ganciclovir suicide gene (SG) lipofection. However, the combination of bleomycin with SG or hIFNβ gene transfer displayed greater antitumor efficacy. The three cell lines exhibited a proliferative behavior consistent with melan A and gp100 melanoma antigens expression, and BRAF V600E mutation. BLM and both genetic treatments increased the fraction of more differentiated and treatment-sensitive cells. Simultaneously, they significantly decreased the sub-population of tumor initiating cells. There was a significant correlation between the cytotoxicity of treatments with BLM and gene transfer and the fraction of cells exhibiting (i) high proliferation index, and (ii) high intracellular levels of reactive oxygen species. Conversely, the fraction of cells surviving to our treatments closely paralleled their (i) colony and (ii) melanosphere forming capacity. A very significant finding was that the combination of BLM with SG or hIFNβ gene almost abrogated the clonogenic capacity of the surviving cells. Altogether, the results presented here suggest that the combined chemo-gene treatments are able to eradicate tumor initiating cells, encouraging further studies aimed to apply this strategy in the clinic.
Collapse
|
44
|
Yao L, Zhang D, Zhao X, Sun B, Liu Y, Gu Q, Zhang Y, Zhao X, Che N, Zheng Y, Liu F, Wang Y, Meng J. Dickkopf-1-promoted vasculogenic mimicry in non-small cell lung cancer is associated with EMT and development of a cancer stem-like cell phenotype. J Cell Mol Med 2016; 20:1673-85. [PMID: 27240974 PMCID: PMC4988283 DOI: 10.1111/jcmm.12862] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/03/2016] [Indexed: 01/04/2023] Open
Abstract
To characterize the contributions of Dickkopf‐1 (DKK1) towards the induction of vasculogenic mimicry (VM) in non‐small cell lung cancer (NSCLC), we evaluated cohorts of primary tumours, performed in vitro functional studies and generated xenograft mouse models. Vasculogenic mimicry was observed in 28 of 205 NSCLC tumours, while DKK1 was detected in 133 cases. Notably, DKK1 was positively associated with VM. Statistical analysis showed that VM and DKK1 were both related to aggressive clinical course and thus were indicators of a poor prognosis. Moreover, expression of epithelial‐mesenchymal transition (EMT)‐related proteins (vimentin, Slug, and Twist), cancer stem‐like cell (CSC)‐related proteins (nestin and CD44), VM‐related proteins (MMP2, MMP9, and vascular endothelial‐cadherin), and β‐catenin‐nu were all elevated in VM‐positive and DKK1‐positive tumours, whereas the epithelial marker (E‐cadherin) was reduced in the VM‐positive and DKK1‐positive groups. Non‐small cell lung cancer cell lines with overexpressed or silenced DKK1 highlighted its role in the restoration of mesenchymal phenotypes and development of CSC characteristics. Moreover, DKK1 significantly promotes NSCLC tumour cells to migrate, invade and proliferate. In vivo animal studies demonstrated that DKK1 enhances the growth of transplanted human tumours cells, as well as increased VM formation, mesenthymal phenotypes and CSC properties. Our results suggest that DKK1 can promote VM formation via induction of the expression of EMT and CSC‐related proteins. As such, we feel that DKK1 may represent a novel target of NSCLC therapy.
Collapse
Affiliation(s)
- Lingli Yao
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, China
| | - Yanrong Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Qiang Gu
- Department of Pathology, Tianjin Medical University, Tianjin, China.,Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, China
| | - Yanhui Zhang
- Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, China
| | - Xueming Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Na Che
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Yanjun Zheng
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Fang Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Yong Wang
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | - Jie Meng
- Department of Pathology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
45
|
Deltex-3-like (DTX3L) stimulates metastasis of melanoma through FAK/PI3K/AKT but not MEK/ERK pathway. Oncotarget 2016; 6:14290-9. [PMID: 26033450 PMCID: PMC4546467 DOI: 10.18632/oncotarget.3742] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/08/2015] [Indexed: 12/19/2022] Open
Abstract
Deltex-3-like (DTX3L), an E3 ligase, is a member of the Deltex (DTX) family and is also called B-lymphoma and BAL-associated protein (BBAP). Previously, we established RFP/RET-transgenic mice, in which systemic hyperpigmented skin, benign melanocytic tumor(s) and melanoma(s) develop stepwise. Here we showed that levels of Dtx3l/DTX3L in spontaneous melanoma in RFP/RET-transgenic mice and human melanoma cell lines were significantly higher than those in benign melanocytic cells and primarily cultured normal human epithelial melanocytes, respectively. Immunohistochemical analysis of human tissues showed that more than 80% of the melanomas highly expressed DTX3L. Activity of FAK/PI3K/AKT signaling, but not that of MEK/ERK signaling, was decreased in Dtx3l/DTX3L-depleted murine and human melanoma cells. In summary, we demonstrated not only increased DTX3L level in melanoma cells but also DTX3L-mediated regulation of invasion and metastasis in melanoma through FAK/PI3K/AKT but not MEK/ERK signaling. Our analysis in human BRAFV600E inhibitor-resistant melanoma cells showed about 80% decreased invasion in the DTX3L-depleted cells compared to that in the DTX3L-intact cells. Thus, DTX3L is clinically a potential therapeutic target as well as a potential biomarker for melanoma.
Collapse
|
46
|
Mo J, He L, Ma B, Chen T. Tailoring Particle Size of Mesoporous Silica Nanosystem To Antagonize Glioblastoma and Overcome Blood-Brain Barrier. ACS APPLIED MATERIALS & INTERFACES 2016; 8:6811-6825. [PMID: 26911360 DOI: 10.1021/acsami.5b11730] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The blood-brain barrier (BBB) is the main bottleneck to prevent some macromolecular substance entering the cerebral circulation, resulting the failure of chemotherapy in the treatment of glioma. Cancer nanotechnology displays potent applications in glioma therapy owing to their penetration across BBB and accumulation into the tumor core. In this study, we have tailored the particle size of mesoporous silica nanoparticles (MSNs) through controlling the hydrolysis rate and polycondensation degree of reactants, and optimized the nanosystem that could effectively penetrate BBB and target the tumor tissue to achieve enhanced antiglioma efficacy. The nanoparticle was conjugated with cRGD peptide to enhance its cancer targeting effect, and then used to load antineoplastic doxorubicin. Therefore, the functionalized nanosystem (DOX@MSNs) selectively recognizes and binds to the U87 cells with higher expression level of ανβ3 integrin, sequentially enhancing the cellular uptake and inhibition to glioma cells, especially the particle size at 40 nm. This particle could rapidly enter cancer cells and was difficult to excrete outside the cells, thus leading to high drug accumulation. Furthermore, DOX@MSNs exhibited much higher selectivity and anticancer activity than free DOX and induced the glioma cells apoptosis through triggering ROS overproduction. Interestingly, DOX@MSNs at about 40 nm exhibited stronger permeability across the BBB, and could disrupt the VM-capability of glioma cells by regulating the expression of E-cadherin, FAK, and MMP-2, thus achieving satisfactory antiglioblastoma efficacy and avoiding the unwanted toxic side effects to normal brain tissue. Taken together, these results suggest that tailoring the particle size of MSNs nanosystem could be an effective strategy to antagonize glioblastoma and overcome BBB.
Collapse
Affiliation(s)
- Jianbin Mo
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| | - Lizhen He
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| | - Bin Ma
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| |
Collapse
|
47
|
Cui YF, Liu AH, An DZ, Sun RB, Shi Y, Shi YX, Shi M, Zhang Q, Wang LL, Feng Q, Pan GL, Wang Q. Claudin-4 is required for vasculogenic mimicry formation in human breast cancer cells. Oncotarget 2016; 6:11087-97. [PMID: 25871476 PMCID: PMC4484441 DOI: 10.18632/oncotarget.3571] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/22/2015] [Indexed: 11/29/2022] Open
Abstract
Vasculogenic mimicry (VM) refers to the unique capability of aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks. Claudins are aberrantly expressed in aggressive breast cancer. However, the relationship between claudins and VM formation is not clear. We examined VM in two human breast cancer cell lines with different aggressive capabilities (MDA-MB-231 and MCF-7 cells) and one human umbilical vein endothelial cell line (HUVEC). Both HUVEC and MDA-MB-231 cells formed vascular channels in Matrigel cultures, while MCF-7 cells did not. Western blot analysis revealed a possible correlation between claudin-4 and -6 expression in breast cancer cell lines and tumor aggressiveness, with protein levels correlating with the ability to form vascular channels. Treatment of MDA-MB-231 and HUVEC cells with claudin-4 monoclonal antibodies completely inhibited the ability of cells to form vascular channels. Moreover, knockdown of claudin-4 by short hairpin RNA completely inhibited tubule formation in MDA-MB-231 cells. Overexpression of claudin-4 in MCF-7 cells induced formation of vascular channels. Immunocytochemistry revealed that membranous claudin-4 protein was significantly associated with vascular channel formation. Collectively, these results indicate that claudin-4 may play a critical role in VM in human breast cancer cells, opening new opportunities to improve aggressive breast cancer therapy.
Collapse
Affiliation(s)
- Yong-Feng Cui
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - An-Heng Liu
- Cardiovascular Medicine, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Dai-Zhi An
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Ru-Bao Sun
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Yun Shi
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Yun-Xiang Shi
- Department of Physiology, BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Miao Shi
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Qiang Zhang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Li-Li Wang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Qiong Feng
- Department of Physiology, BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Gui-Lan Pan
- Department of Physiology, BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Qiang Wang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
48
|
Yassine H, De Freitas Caires N, Depontieu F, Scherpereel A, Awad A, Tsicopoulos A, Leboeuf C, Janin A, Duez C, Grigoriu B, Lassalle P. The non glycanated endocan polypeptide slows tumor growth by inducing stromal inflammatory reaction. Oncotarget 2015; 6:2725-35. [PMID: 25575808 PMCID: PMC4413613 DOI: 10.18632/oncotarget.2614] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/21/2014] [Indexed: 01/08/2023] Open
Abstract
Endocan expression is increasingly studied in various human cancers. Experimental evidence showed that human endocan, through its glycan chain, is implicated in various processes of tumor growth. We functionally characterize mouse endocan which is also a chondroitin sulfate proteoglycan but much less glycanated than human endocan. Distant domains from the O-glycanation site, located within exons 1 and 2 determine the glycanation pattern of endocan. In opposite to the human homologue, overexpression of mouse endocan in HT-29 cells delayed the tumor appearance and reduced the tumor growth rate. This tumor growth inhibition is supported by non glycanated form of mouse endocan. Non glycanated human endocan overexpressed in HT-29, A549 or K1000 cells also exhibited an anti-tumor effect. Moreover, systemic delivery of non glycanated human endocan also results in HT-29 tumor growth delay. In vitro, endocan polypeptide did not affect HT-29 cell proliferation, nor cell viability. In tumor tissue sections, a stromal inflammatory reaction was observed only in tumors overexpressing endocan polypeptide, and depletion of CD122+ cells was able to delete partially the anti-tumor effect of endocan polypeptide. These results reveal a novel pathway for endocan in the control of tumor growth, which involves inflammatory cells of the innate immunity.
Collapse
Affiliation(s)
- Hanane Yassine
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France
| | - Nathalie De Freitas Caires
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France.,Lunginnov, Lille, France
| | - Florence Depontieu
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France
| | - Arnaud Scherpereel
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France.,CHRU Lille, Hôpital Calmette, Lille, France
| | - Ali Awad
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France
| | - Anne Tsicopoulos
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France
| | - Christophe Leboeuf
- Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Anne Janin
- Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Catherine Duez
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France
| | - Bogdan Grigoriu
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France.,Regional Institute of Oncology, Iasi, Romania.,University of Medicine and Pharmacy "Gr.T.Popa" Iasi, Iasi, Romania
| | - Philippe Lassalle
- Institut Pasteur de Lille, Center for Infection and Immunity of Lille, Lille, France.,Univ Lille Nord de France, Lille, France.,CNRS, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale, Lille, France
| |
Collapse
|
49
|
Rycaj K, Tang DG. Cell-of-Origin of Cancer versus Cancer Stem Cells: Assays and Interpretations. Cancer Res 2015; 75:4003-11. [PMID: 26292361 DOI: 10.1158/0008-5472.can-15-0798] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 05/18/2015] [Indexed: 12/14/2022]
Abstract
A tumor originates from a normal cell that has undergone tumorigenic transformation as a result of genetic mutations. This transformed cell is the cell-of-origin for the tumor. In contrast, an established clinical tumor is sustained by subpopulations of self-renewing cancer cells operationally called cancer stem cells (CSC) that can generate, intraclonally, both tumorigenic and nontumorigenic cells. Identifying and characterizing tumor cell-of-origin and CSCs should help elucidate tumor cell heterogeneity, which, in turn, should help understand tumor cell responses to clinical treatments, drug resistance, tumor relapse, and metastatic spread. Both tumor transplantation and lineage-tracing assays have been helpful in characterizing these cancer cell populations, although each system has its strengths and caveats. In this article, we briefly review and summarize advantages and limitations of both assays in support of a combinatorial approach to accurately define the roles of both cancer-initiating and cancer-propagating cells. As an aside, we also wish to clarify the definitions of cancer cell-of-origin and CSCs, which are often interchangeably used by mistake.
Collapse
Affiliation(s)
- Kiera Rycaj
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas
| | - Dean G Tang
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas. Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
50
|
Abstract
BACKGROUND The histogenesis and clinical behavior of combined cutaneous tumors (CCTs) in which the mesenchymal component consists of melanoma remain unclear. OBJECTIVE We sought to characterize the clinical, histologic, and molecular findings in CCTs with an epithelial and a melanoma component. METHODS We retrospectively reviewed the records from 2 institutions for CCTs. Fluorescence in situ hybridization was performed to assess chromosomal copy number alterations in both components. RESULTS Sixteen CCTs were included. The most common subtype was the squamomelanocytic tumor (11), followed by the basomelanocytic tumor (3) and the trichoblastomelanoma (2). CCTs were more common in men (87%), on the head and neck (57%), and had extensive solar elastosis (81%). The median follow-up was 25 months (range, 8-167 months). One case had an adverse outcome. Fluorescence in situ hybridization revealed chromosomal alterations in approximately 55% of the cases. Five cases showed chromosomal gains only in the melanocytic component. One case showed 11q13 gains in both the epithelial and melanocytic components. LIMITATIONS Our study is retrospective and the sample is small. CONCLUSIONS The low incidence of adverse outcomes suggests that CCT may be more indolent than noncombined tumors. 11q13 amplification in both components supports the theory of dual differentiation from a common progenitor cell.
Collapse
|