1
|
Lucà S, Franco R, Napolitano A, Soria V, Ronchi A, Zito Marino F, Della Corte CM, Morgillo F, Fiorelli A, Luciano A, Palma G, Arra C, Battista S, Cerchia L, Fedele M. PATZ1 in Non-Small Cell Lung Cancer: A New Biomarker That Negatively Correlates with PD-L1 Expression and Suppresses the Malignant Phenotype. Cancers (Basel) 2023; 15:cancers15072190. [PMID: 37046851 PMCID: PMC10093756 DOI: 10.3390/cancers15072190] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Non-small cell lung cancer (NSCLC), the leading cause of cancer death worldwide, is still an unmet medical problem due to the lack of both effective therapies against advanced stages and markers to allow a diagnosis of the disease at early stages before its progression. Immunotherapy targeting the PD-1/PD-L1 checkpoint is promising for many cancers, including NSCLC, but its success depends on the tumor expression of PD-L1. PATZ1 is an emerging cancer-related transcriptional regulator and diagnostic/prognostic biomarker in different malignant tumors, but its role in lung cancer is still obscure. Here we investigated expression and role of PATZ1 in NSCLC, in correlation with NSCLC subtypes and PD-L1 expression. A cohort of 104 NSCLCs, including lung squamous cell carcinomas (LUSCs) and adenocarcinomas (LUADs), was retrospectively analyzed by immunohistochemistry for the expression of PATZ1 and PD-L1. The results were correlated with each other and with the clinical characteristics, showing on the one hand a positive correlation between the high expression of PATZ1 and the LUSC subtype and, on the other hand, a negative correlation between PATZ1 and PD-L1, validated at the mRNA level in independent NSCLC datasets. Consistently, two NSCLC cell lines transfected with a PATZ1-overexpressing plasmid showed PD-L1 downregulation, suggesting a role for PATZ1 in the negative regulation of PD-L1. We also showed that PATZ1 overexpression inhibits NSCLC cell proliferation, migration, and invasion, and that Patz1-knockout mice develop LUAD. Overall, this suggests that PATZ1 may act as a tumor suppressor in NSCLC.
Collapse
Affiliation(s)
- Stefano Lucà
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Antonella Napolitano
- Institute for Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80145 Naples, Italy
| | - Valeria Soria
- Institute for Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80145 Naples, Italy
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Carminia Maria Della Corte
- Department of Precision Medicine, Medical Oncology, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Floriana Morgillo
- Department of Precision Medicine, Medical Oncology, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Alfonso Fiorelli
- Translational Medical and Surgical Science, Thoracic Surgery, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Antonio Luciano
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Giuseppe Palma
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Claudio Arra
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Sabrina Battista
- Institute for Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80145 Naples, Italy
| | - Laura Cerchia
- Institute for Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80145 Naples, Italy
| | - Monica Fedele
- Institute for Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), 80145 Naples, Italy
| |
Collapse
|
2
|
Nair L, Mukherjee S, Kaur K, Murphy CM, Ravichandiran V, Roy S, Singh M. Multi compartmental 3D breast cancer disease model–recapitulating tumor complexity in in-vitro. Biochim Biophys Acta Gen Subj 2023; 1867:130361. [PMID: 37019341 DOI: 10.1016/j.bbagen.2023.130361] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023]
Abstract
Breast cancer is the most common ailment among women. In 2020, it had the highest incidence of any type of cancer. Many Phase II and III anti-cancer drugs fail due to efficacy, durability, and side effects. Thus, accelerated drug screening models must be accurate. In-vivo models have been used for a long time, but delays, inconsistent results, and a greater sense of responsibility among scientists toward wildlife have led to the search for in-vitro alternatives. Stromal components support breast cancer growth and survival. Multi-compartment Transwell models may be handy instruments. Co-culturing breast cancer cells with endothelium and fibroblasts improves modelling. The extracellular matrix (ECM) supports native 3D hydrogels in natural and polymeric forms. 3D Transwell cultured tumor spheroids mimicked in-vivo pathological conditions. Tumor invasion, migration, Trans-endothelial migration, angiogenesis, and spread are studied using comprehensive models. Transwell models can create a cancer niche and conduct high-throughput drug screening, promising future applications. Our comprehensive shows how 3D in-vitro multi compartmental models may be useful in producing breast cancer stroma in Transwell culture.
Collapse
Affiliation(s)
- Lakshmi Nair
- Department of Pharmaceutical Sciences, Assam Central University, Silchar, Assam 788011, India
| | - Souvik Mukherjee
- Department of Pharmaceutical Sciences, Guru Ghasidas University, Koni, Bilaspur,(C.G 495009, India
| | - Kulwinder Kaur
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons (RCSI), Dublin D02YN77, Ireland
| | - Ciara M Murphy
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons (RCSI), Dublin D02YN77, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin D02YN77, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India.
| | - Manjari Singh
- Department of Pharmaceutical Sciences, Assam Central University, Silchar, Assam 788011, India.
| |
Collapse
|
3
|
Benítez-Arvizu G, Castro-Jácome TP, Tovar-Pérez E, Alcántara-Quintana LE. [Antiproliferative, apoptotic, and antimigratory activities of kafirins on cervical cancer-derived cell lines]. REVISTA MEDICA DEL INSTITUTO MEXICANO DEL SEGURO SOCIAL 2023; 61:S4-S11. [PMID: 36378016 PMCID: PMC10395951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/27/2022] [Indexed: 06/16/2023]
Abstract
Background Cervical cancer is one of the leading causes of death in women worldwide, both in developed and developing countries. Therefore, effective treatment of cervical cancer with potential anti-tumor drugs is important. However, new treatments inspired by nutritional medicine are needed. Objective To use the human cervical cancer cell lines HeLa and SiHa to evaluate the antiproliferative, apoptotic, and migratory activity of sorghum (kafirins). Materials and methods The anticancer effects of the kafirins were examined by counting cells, MTT assays, apoptosis, and migration assays. Results This investigation showed that sorghum induced growth inhibition of HeLa and SiHa cells at a significant level. The growth inhibition is dose-dependent and irreversible. When HeLa and SiHa cells were treated with sorghum due to the activity of kafirins, morphological changes were observed, which were identified through the formation of apoptopic bodies. And the kafirins at concentrations of 37.5, 75, 150, and 300 μg/mL decreased the migration of HeLa cells and SiHa cells. Conclusion This paper demonstrates the induction of antiproliferative, apoptotic, and anti-migratory activity in HeLa and SiHa cells by kafirins. Sorghum may be used as a nutraceutical with potential cancer-prevention benefits.
Collapse
Affiliation(s)
- Gamaliel Benítez-Arvizu
- Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Hospital de Especialidades “Dr. Bernardo Sepúlveda Gutiérrez”, Unidad Complementaria Banco de Sangre. Ciudad de México, MéxicoInstituto Mexicano del Seguro SocialMéxico
| | - Tania Patricia Castro-Jácome
- Instituto Tecnológico de Tepic, Laboratorio Integral de Investigación en Alimentos. Tepic, Nayarit, MéxicoInstituto Tecnológico de TepicMéxico
| | - Erik Tovar-Pérez
- Universidad Autónoma de Querétaro, Campus Amazcala, Facultad de Ingeniería. El Marqués, Querétaro, MéxicoUniversidad Autónoma de QuerétaroMéxico
| | - Luz Eugenia Alcántara-Quintana
- Universidad Autónoma de San Luis Potosí, Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología. San Luis Potosí, San Luis Potosí, México Universidad Autónoma de San Luis PotosíMéxico
| |
Collapse
|
4
|
Chen CJ, Shang HS, Huang YL, Tien N, Chen YL, Hsu SY, Wu RSC, Tang CL, Lien JC, Lee MH, Lu HF, Hsia TC. Bisdemethoxycurcumin suppresses human brain glioblastoma multiforme GBM 8401 cell migration and invasion via affecting NF-κB and MMP-2 and MMP-9 signaling pathway in vitro. ENVIRONMENTAL TOXICOLOGY 2022; 37:2388-2397. [PMID: 35735092 DOI: 10.1002/tox.23604] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Human glioblastoma (GBM) is one of the common cancer death in adults worldwide, and its metastasis will lead to difficult treatment. Finding compounds for future to develop treatment is urgent. Bisdemethoxycurcumin (BDMC), a natural product, was isolated from the rhizome of turmeric (Curcuma longa), which has been shown to against many human cancer cells. In the present study, we evaluated the antimetastasis activity of BDMC in human GBM cells. Cell proliferation, cell viability, cellular uptake, wound healing, migration and invasion, and western blotting were analyzed. Results indicated that BDMC at 1.5-3 μM significantly decreased the cell proliferation by MTT assay. BDMC showed the highest uptake by cells at 3 h. After treatment of BDMC at 12-48 h significantly inhibited cell motility in GBM 8401 cells by wound healing assay. BDMC suppressed cell migration and invasion at 24 and 48 h treatment by transwell chamber assay. BDMC significantly decreased the levels of proteins associated with PI3K/Akt, Ras/MEK/ERK pathways and resulted in the decrease in the expressions of NF-κB, MMP-2, MMP-9, and N-cadherin, leading to the inhibition of cell migration and invasion. These findings suggest that BDMC may be a potential candidate for the antimetastasis of human GBM cells in the future.
Collapse
Affiliation(s)
- Chiung-Ju Chen
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Clinical of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Li Huang
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung City, Taiwan
| | - Ni Tien
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yung-Liang Chen
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu, Taiwan
| | - Sheng-Yao Hsu
- Department of Ophthalmology, An Nan Hospital, China Medical University, Tainan, Taiwan
- Department of Optometry, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Rick Sai-Chuen Wu
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Chien-Lun Tang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Mei-Hui Lee
- Department of Genetic Counseling Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsu-Feng Lu
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung City, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Te-Chun Hsia
- Department of Respiratory Therapy, China Medical University, Taichung, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
5
|
Rae S, Spillane C, Blackshields G, Madden SF, Keenan J, Stordal B. The EMT-activator ZEB1 is unrelated to platinum drug resistance in ovarian cancer but is predictive of survival. Hum Cell 2022; 35:1547-1559. [PMID: 35794446 PMCID: PMC9374625 DOI: 10.1007/s13577-022-00744-y] [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: 04/19/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022]
Abstract
The IGROVCDDP cisplatin-resistant ovarian cancer cell line is an unusual model, as it is also cross-resistant to paclitaxel. IGROVCDDP, therefore, models the resistance phenotype of serous ovarian cancer patients who have failed frontline platinum/taxane chemotherapy. IGROVCDDP has also undergone epithelial-mesenchymal transition (EMT). We aim to determine if alterations in EMT-related genes are related to or independent from the drug-resistance phenotypes. EMT gene and protein markers, invasion, motility and morphology were investigated in IGROVCDDP and its parent drug-sensitive cell line IGROV-1. ZEB1 was investigated by qPCR, Western blotting and siRNA knockdown. ZEB1 was also investigated in publicly available ovarian cancer gene-expression datasets. IGROVCDDP cells have decreased protein levels of epithelial marker E-cadherin (6.18-fold, p = 1.58e-04) and higher levels of mesenchymal markers vimentin (2.47-fold, p = 4.43e-03), N-cadherin (4.35-fold, p = 4.76e-03) and ZEB1 (3.43-fold, p = 0.04). IGROVCDDP have a spindle-like morphology consistent with EMT. Knockdown of ZEB1 in IGROVCDDP does not lead to cisplatin sensitivity but shows a reversal of EMT-gene signalling and an increase in cell circularity. High ZEB1 gene expression (HR = 1.31, n = 2051, p = 1.31e-05) is a marker of poor overall survival in high-grade serous ovarian-cancer patients. In contrast, ZEB1 is not predictive of overall survival in high-grade serous ovarian-cancer patients known to be treated with platinum chemotherapy. The increased expression of ZEB1 in IGROVCDDP appears to be independent of the drug-resistance phenotypes. ZEB1 has the potential to be used as biomarker of overall prognosis in ovarian-cancer patients but not of platinum/taxane chemoresistance.
Collapse
Affiliation(s)
- Sophie Rae
- Department of Natural Sciences, Middlesex University London, London, UK
| | - Cathy Spillane
- Department of Histopathology, St James' Hospital and Trinity College Dublin, Dublin, Ireland
| | - Gordon Blackshields
- Department of Histopathology, St James' Hospital and Trinity College Dublin, Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Joanne Keenan
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Britta Stordal
- Department of Natural Sciences, Middlesex University London, London, UK.
| |
Collapse
|
6
|
Singh A, Erijman A, Noronha A, Kumar H, Peleg Y, Yarden Y, Shifman JM. Engineered variants of the Ras effector protein RASSF5 (NORE1A) promote anticancer activities in lung adenocarcinoma. J Biol Chem 2021; 297:101353. [PMID: 34717958 PMCID: PMC8605244 DOI: 10.1016/j.jbc.2021.101353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 11/24/2022] Open
Abstract
Within the superfamily of small GTPases, Ras appears to be the master regulator of such processes as cell cycle progression, cell division, and apoptosis. Several oncogenic Ras mutations at amino acid positions 12, 13, and 61 have been identified that lose their ability to hydrolyze GTP, giving rise to constitutive signaling and eventually development of cancer. While disruption of the Ras/effector interface is an attractive strategy for drug design to prevent this constitutive activity, inhibition of this interaction using small molecules is impractical due to the absence of a cavity to which such molecules could bind. However, proteins and especially natural Ras effectors that bind to the Ras/effector interface with high affinity could disrupt Ras/effector interactions and abolish procancer pathways initiated by Ras oncogene. Using a combination of computational design and in vitro evolution, we engineered high-affinity Ras-binding proteins starting from a natural Ras effector, RASSF5 (NORE1A), which is encoded by a tumor suppressor gene. Unlike previously reported Ras oncogene inhibitors, the proteins we designed not only inhibit Ras-regulated procancer pathways, but also stimulate anticancer pathways initiated by RASSF5. We show that upon introduction into A549 lung carcinoma cells, the engineered RASSF5 mutants decreased cell viability and mobility to a significantly greater extent than WT RASSF5. In addition, these mutant proteins induce cellular senescence by increasing acetylation and decreasing phosphorylation of p53. In conclusion, engineered RASSF5 variants provide an attractive therapeutic strategy able to oppose cancer development by means of inhibiting of procancer pathways and stimulating anticancer processes.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ariel Erijman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ashish Noronha
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Hemant Kumar
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yoav Peleg
- Life Sciences Core Facilities (LSCF) Structural Proteomics Unit (SPU), Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Julia M Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
7
|
Molecular and Cellular Mechanisms of Perineural Invasion in Oral Squamous Cell Carcinoma: Potential Targets for Therapeutic Intervention. Cancers (Basel) 2021; 13:cancers13236011. [PMID: 34885121 PMCID: PMC8656475 DOI: 10.3390/cancers13236011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Squamous cell carcinoma is the most common type of oral cavity cancer. It can spread along and invade nerves in a process called perineural invasion. Perineural invasion can increase the chances of tumor recurrence and reduce survival in patients with oral cancer. Understanding how oral cancer interacts with nerves to facilitate perineural invasion is an important area of research. Targeting key events that contribute to perineural invasion in oral cavity cancer may reduce tumor recurrence and improve survival. In this review, we describe the impact of perineural invasion in oral cancer and the mechanisms that contribute to perineural invasion. Highlighting the key events of perineural invasion is important for the identification and testing of novel therapies for oral cancer with perineural invasion. Abstract The most common oral cavity cancer is squamous cell carcinoma (SCC), of which perineural invasion (PNI) is a significant prognostic factor associated with decreased survival and an increased rate of locoregional recurrence. In the classical theory of PNI, cancer was believed to invade nerves directly through the path of least resistance in the perineural space; however, more recent evidence suggests that PNI requires reciprocal signaling interactions between tumor cells and nerve components, particularly Schwann cells. Specifically, head and neck SCC can express neurotrophins and neurotrophin receptors that may contribute to cancer migration towards nerves, PNI, and neuritogenesis towards cancer. Through reciprocal signaling, recent studies also suggest that Schwann cells may play an important role in promoting PNI by migrating toward cancer cells, intercalating, and dispersing cancer, and facilitating cancer migration toward nerves. The interactions of neurotrophins with their high affinity receptors is a new area of interest in the development of pharmaceutical therapies for many types of cancer. In this comprehensive review, we discuss diagnosis and treatment of oral cavity SCC, how PNI affects locoregional recurrence and survival, and the impact of adjuvant therapies on tumors with PNI. We also describe the molecular and cellular mechanisms associated with PNI, including the expression of neurotrophins and their receptors, and highlight potential targets for therapeutic intervention for PNI in oral SCC.
Collapse
|
8
|
Chou SE, Lee KL, Wei PK, Cheng JY. Screening anti-metastasis drugs by cell adhesion-induced color change in a biochip. LAB ON A CHIP 2021; 21:2955-2970. [PMID: 34132296 DOI: 10.1039/d1lc00039j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Metastasis is a frequent complication of cancer and accounts for more than 60% of patients' mortality. Despite technological advancements, treatment options are still limited. Ion channels participate in the regulation of cell adhesion, whilst the regulation of cell adhesion further controls metastasis formation. However, to develop a new ion channel inhibitor targeting metastasis takes tremendous effort and resources; therefore, drug repurposing is an emerging strategy in oncology. In previous studies, we have developed a metal-based nanoslit surface plasmon resonance (SPR) platform to examine the influence of drugs on the cell adhesion process. In this work, we developed a scanner-based cell adhesion kinetic examination (CAKE) system that is capable of monitoring the cell adhesion process by measuring color changes of SPR biosensors. The system's performance was demonstrated by screening the anti-metastasis ability of compounds from a commercial ion-channel inhibitor library. Out of the 274 compounds from the inhibitor library, zinc pyrithione (ZPT) and terfenadine were demonstrated to influence CL1-5 cell adhesion. The cell responses to the two compounds were then compared with those by traditional cell adhesion assays where similar behavior was observed. Further investigation of the two compounds using wound healing and transwell assays was performed and inhibitions of both cell migration and invasion by the two compounds were also observed. The results indicate that ZPT and terfenadine are potential candidates for anti-metastasis drugs. Our work has demonstrated the label-free drug screening ability of our CAKE system for finding potential drugs for cancer treatment.
Collapse
Affiliation(s)
- Shih-En Chou
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan.
| | - Kuang-Li Lee
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan.
| | - Pei-Kuen Wei
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan. and Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Ji-Yen Cheng
- Research Center for Applied Sciences, Academia Sinica Taiwan, Taipei, 11529, Taiwan. and Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan and Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, Keelung, 20224, Taiwan and College of Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| |
Collapse
|
9
|
Alburquerque-González B, Bernabé-García Á, Bernabé-García M, Ruiz-Sanz J, López-Calderón FF, Gonnelli L, Banci L, Peña-García J, Luque I, Nicolás FJ, Cayuela-Fuentes ML, Luchinat E, Pérez-Sánchez H, Montoro-García S, Conesa-Zamora P. The FDA-Approved Antiviral Raltegravir Inhibits Fascin1-Dependent Invasion of Colorectal Tumor Cells In Vitro and In Vivo. Cancers (Basel) 2021; 13:cancers13040861. [PMID: 33670655 PMCID: PMC7921938 DOI: 10.3390/cancers13040861] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 01/29/2023] Open
Abstract
Simple Summary Colorectal cancer (CRC) is the third leading cause of cancer-related deaths worldwide. Serrated adenocarcinoma (SAC) has been recently recognized by the WHO as a histological CRC with bad prognosis. Consistent with previous evidence, our group identified Fascin1 as a protein directly related to the invasiveness of tumor cells, overexpressed and positively correlated with worse survival in various carcinomas, including SAC. Therefore, Fascin1 has emerged as an ideal target for cancer treatment. In the present study, virtual screening has been carried out from a library of 9591 compounds, thus identifying the FDA-approved anti-retroviral raltegravir (RAL) as a potential Fascin1 blocker. In vitro and in vivo results show that RAL exhibits Fascin1-binding activity and Fascin1-dependent anti-invasive and anti-metastatic properties against CRC cells both in vitro and in vivo. Abstract Background: Fascin1 is the key actin-bundling protein involved in cancer invasion and metastasis whose expression is associated with bad prognosis in tumor from different origins. Methods: In the present study, virtual screening (VS) was performed for the search of Fascin1 inhibitors and RAL, an FDA-approved inhibitor of human immunodeficiency virus-1 (HIV-1) integrase, was identified as a potential Fascin1 inhibitor. Biophysical techniques including nuclear magnetic resonance (NMR) and differential scanning fluorimetry (DSF) were carried out in order to confirm RAL as a Fascin1 blocker. The effect of RAL on actin-bundling activity Fascin1 was assessed by transmission electron microscopy (TEM), immunofluorescence, migration, and invasion assays on two human colorectal adenocarcinoma cell lines: HCT-116 and DLD-1. In addition, the anti-metastatic potential of RAL was in vivo evaluated by using the zebrafish animal model. Results: NMR and DSF confirmed in silico predictions and TEM demonstrated the RAL-induced disorganization of the actin structure compared to control conditions. The protrusion of lamellipodia in cancer cell line overexpressing Fascin1 (HCT-116) was abolished in the presence of this drug. By following the addition of RAL, migration of HCT-116 and DLD-1 cell lines was significantly inhibited. Finally, using endogenous and exogenous models of Fascin1 expression, the invasive capacity of colorectal tumor cells was notably impaired in the presence of RAL in vivo assays; without undesirable cytotoxic effects. Conclusion: The current data show the in vitro and in vivo efficacy of the antiretroviral drug RAL in inhibiting human colorectal cancer cells invasion and metastasis in a Fascin1-dependent manner.
Collapse
Affiliation(s)
- Begoña Alburquerque-González
- Department of Pathology and Histology, Campus de los Jerónimos, UCAM Universidad Católica San Antonio de Murcia, s/n, 30107 Murcia, Spain; (B.A.-G.); (F.F.L.-C.)
| | - Ángel Bernabé-García
- Laboratorio de Regeneración, Oncología Molecular y TGF-ß, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Carretera Madrid-Cartagena, El Palmar, 30120 Murcia, Spain; (Á.B.-G.); (F.J.N.)
| | - Manuel Bernabé-García
- Telomerase, Cancer and Aging Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (M.B.-G.); (M.L.C.-F.)
| | - Javier Ruiz-Sanz
- Department of Physical Chemistry, Institute of Biotechnology and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment, Spain Campus Fuentenueva s/n, University of Granada, 18071 Granada, Spain; (J.R.-S.); (I.L.)
| | - Fernando Feliciano López-Calderón
- Department of Pathology and Histology, Campus de los Jerónimos, UCAM Universidad Católica San Antonio de Murcia, s/n, 30107 Murcia, Spain; (B.A.-G.); (F.F.L.-C.)
| | - Leonardo Gonnelli
- CERM—Magnetic Resonance Center, Università degli Studi di Firenze, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy; (L.G.); (L.B.); (E.L.)
- Dipartimento di Chimica, Università degli Studi di Firenze, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Lucia Banci
- CERM—Magnetic Resonance Center, Università degli Studi di Firenze, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy; (L.G.); (L.B.); (E.L.)
- Dipartimento di Chimica, Università degli Studi di Firenze, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Jorge Peña-García
- Structural Bioinformatics and High Performance Computing (BIO-HPC) Research Group, Campus de los Jerónimos, s/n, UCAM Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain; (J.P.-G.); (H.P.-S.)
| | - Irene Luque
- Department of Physical Chemistry, Institute of Biotechnology and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment, Spain Campus Fuentenueva s/n, University of Granada, 18071 Granada, Spain; (J.R.-S.); (I.L.)
| | - Francisco José Nicolás
- Laboratorio de Regeneración, Oncología Molecular y TGF-ß, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Carretera Madrid-Cartagena, El Palmar, 30120 Murcia, Spain; (Á.B.-G.); (F.J.N.)
| | - María Luisa Cayuela-Fuentes
- Telomerase, Cancer and Aging Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (M.B.-G.); (M.L.C.-F.)
| | - Enrico Luchinat
- CERM—Magnetic Resonance Center, Università degli Studi di Firenze, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy; (L.G.); (L.B.); (E.L.)
- Consorzio per lo Sviluppo dei Sistemi a Grande Interfase—CSGI, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High Performance Computing (BIO-HPC) Research Group, Campus de los Jerónimos, s/n, UCAM Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain; (J.P.-G.); (H.P.-S.)
| | - Silvia Montoro-García
- Cell Culture Lab, Facultad de Ciencias de la Salud, Campus de los Jerónimos, s/n, UCAM Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain
- Correspondence: (S.M.-G.); (P.C.-Z.); Tel.: +34-9681-286-02 (Ext. 951615) (P.C.-Z.)
| | - Pablo Conesa-Zamora
- Laboratory Medicine Department, Group of Molecular Pathology and Pharmacogenetics, Biomedical Research Institute from Murcia (IMIB), Hospital Universitario Santa Lucía, c/Mezquita sn, 30202 Cartagena, Spain
- Correspondence: (S.M.-G.); (P.C.-Z.); Tel.: +34-9681-286-02 (Ext. 951615) (P.C.-Z.)
| |
Collapse
|
10
|
Moodad S, El Hajj R, Hleihel R, Hajjar L, Tawil N, Karam M, Hamie M, Abou Merhi R, El Sabban M, El Hajj H. Lenalidomide in Combination with Arsenic Trioxide: an Effective Therapy for Primary Effusion Lymphoma. Cancers (Basel) 2020; 12:E2483. [PMID: 32883022 PMCID: PMC7563318 DOI: 10.3390/cancers12092483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
Primary effusion lymphoma (PEL) is a rare aggressive subset of non-Hodgkin B cell lymphoma. PEL is secondary to Kaposi sarcoma herpes virus (KSHV) and predominantly develops in serous cavities. Conventional chemotherapy remains the treatment of choice for PEL and yields high response rates with no significant comorbidities. Yet, chemotherapy often fails in achieving or maintaining long-term remission. Lenalidomide (Lena), an immunomodulatory drug, displayed some efficacy in the treatment of PEL. On the other hand, arsenic trioxide (ATO) in combination with other agents effectively treated a number of blood malignancies, including PEL. In this study, we present evidence that the combination of ATO/Lena significantly enhanced survival of PEL mice, decreased the volume of exacerbated ascites in the peritoneum, and reduced tumor infiltration in organs of treated animals. In ex vivo treated PEL cells, ATO/Lena decreased the proliferation and downregulated the expression of KSHV latent viral proteins. This was associated with decreased NF-κB activation, resulting in reactivation of viral replication, downregulation of interleukin-6 (IL-6) and IL-10, inhibition of vascular endothelial growth factor, and apoptosis. Our results elucidate the mechanism of action of ATO/Lena and present it as a promising targeted therapeutic modality in PEL management, which warrants further clinical investigation.
Collapse
Affiliation(s)
- Sara Moodad
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (S.M.); (R.H.); (M.H.)
| | - Rana El Hajj
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon;
| | - Rita Hleihel
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (S.M.); (R.H.); (M.H.)
- Department of Anatomy, Cell Biology, and Physiology, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (L.H.); (M.E.S.)
| | - Layal Hajjar
- Department of Anatomy, Cell Biology, and Physiology, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (L.H.); (M.E.S.)
| | - Nadim Tawil
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (N.T.); (M.K.)
| | - Martin Karam
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (N.T.); (M.K.)
| | - Maguy Hamie
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (S.M.); (R.H.); (M.H.)
| | - Raghida Abou Merhi
- Department of Biology, Faculty of Sciences, GSBT laboratory, Lebanese University, Hadath 31143, Lebanon;
| | - Marwan El Sabban
- Department of Anatomy, Cell Biology, and Physiology, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (L.H.); (M.E.S.)
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 202627, Lebanon; (N.T.); (M.K.)
| |
Collapse
|
11
|
Ramadan Q, Zourob M. Organ-on-a-chip engineering: Toward bridging the gap between lab and industry. BIOMICROFLUIDICS 2020; 14:041501. [PMID: 32699563 PMCID: PMC7367691 DOI: 10.1063/5.0011583] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/22/2020] [Indexed: 05/03/2023]
Abstract
Organ-on-a-chip (OOC) is a very ambitious emerging technology with a high potential to revolutionize many medical and industrial sectors, particularly in preclinical-to-clinical translation in the pharmaceutical arena. In vivo, the function of the organ(s) is orchestrated by a complex cellular structure and physiochemical factors within the extracellular matrix and secreted by various types of cells. The trend in in vitro modeling is to simplify the complex anatomy of the human organ(s) to the minimal essential cellular structure "micro-anatomy" instead of recapitulating the full cellular milieu that enables studying the absorption, metabolism, as well as the mechanistic investigation of drug compounds in a "systemic manner." However, in order to reflect the human physiology in vitro and hence to be able to bridge the gap between the in vivo and in vitro data, simplification should not compromise the physiological relevance. Engineering principles have long been applied to solve medical challenges, and at this stage of organ-on-a-chip technology development, the work of biomedical engineers, focusing on device engineering, is more important than ever to accelerate the technology transfer from the academic lab bench to specialized product development institutions and to the increasingly demanding market. In this paper, instead of presenting a narrative review of the literature, we systemically present a synthesis of the best available organ-on-a-chip technology from what is found, what has been achieved, and what yet needs to be done. We emphasized mainly on the requirements of a "good in vitro model that meets the industrial need" in terms of the structure (micro-anatomy), functions (micro-physiology), and characteristics of the device that hosts the biological model. Finally, we discuss the biological model-device integration supported by an example and the major challenges that delay the OOC technology transfer to the industry and recommended possible options to realize a functional organ-on-a-chip system.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| | - Mohammed Zourob
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Yu Y, He JH, Hu LL, Jiang LL, Fang L, Yao GD, Wang SJ, Yang Q, Guo Y, Liu L, Shang T, Sato Y, Kawamura K, Hsueh AJ, Sun YP. Placensin is a glucogenic hormone secreted by human placenta. EMBO Rep 2020; 21:e49530. [PMID: 32329225 DOI: 10.15252/embr.201949530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/13/2020] [Accepted: 03/27/2020] [Indexed: 01/09/2023] Open
Abstract
FBN1 encodes asprosin, a glucogenic hormone, following furin cleavage of the C-terminus of profibrillin 1. Based on evolutionary conservation between FBN1 and FBN2, together with conserved furin cleavage sites, we identified a peptide hormone placensin encoded by FBN2 based on its high expression in trophoblasts of human placenta. In primary and immortalized murine hepatocytes, placensin stimulates cAMP production, protein kinase A (PKA) activity, and glucose secretion, accompanied by increased expression of gluconeogenesis enzymes. In situ perfusion of liver and in vivo injection with placensin also stimulate glucose secretion. Placensin is secreted by immortalized human trophoblastic HTR-8/SVneo cells, whereas placensin treatment stimulates cAMP-PKA signaling in these cells, accompanied by increases in MMP9 transcripts and activities, thereby promoting cell invasion. In pregnant women, levels of serum placensin increase in a stage-dependent manner. During third trimester, serum placensin levels of patients with gestational diabetes mellitus are increased to a bigger extent compared to healthy pregnant women. Thus, placensin represents a placenta-derived hormone, capable of stimulating glucose secretion and trophoblast invasion.
Collapse
Affiliation(s)
- Yiping Yu
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jia-Huan He
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lin-Li Hu
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lin-Lin Jiang
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lanlan Fang
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gui-Dong Yao
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Si-Jia Wang
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingling Yang
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yanjie Guo
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lin Liu
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Trisha Shang
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yorino Sato
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Kazuhiro Kawamura
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Aaron Jw Hsueh
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ying-Pu Sun
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Montoro-García S, Alburquerque-González B, Bernabé-García Á, Bernabé-García M, Rodrigues PC, den-Haan H, Luque I, Nicolás FJ, Pérez-Sánchez H, Cayuela ML, Salo T, Conesa-Zamora P. Novel anti-invasive properties of a Fascin1 inhibitor on colorectal cancer cells. J Mol Med (Berl) 2020; 98:383-394. [PMID: 31996952 DOI: 10.1007/s00109-020-01877-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 12/05/2019] [Accepted: 01/15/2020] [Indexed: 01/01/2023]
Abstract
Tumor invasion and metastasis involve processes in which actin cytoskeleton rearrangement induced by Fascin1 plays a crucial role. Indeed, Fascin1 has been found overexpressed in tumors with worse prognosis. Migrastatin and its analogues target Fascin1 and inhibit its activity. However, there is need for novel and smaller Fascin1 inhibitors. The aim of this study was to assess the effect of compound G2 in colorectal cancer cell lines and compare it to migrastatin in in vitro and in vivo assays. Molecular modeling, actin-bundling, cell viability, inmunofluorescence, migration, and invasion assays were carried out in order to test anti-migratory and anti-invasive properties of compound G2. In addition, the in vivo effect of compound G2 was evaluated in a zebrafish model of invasion. HCT-116 cells exhibited the highest Fascin1 expression from eight tested colorectal cancer cell lines. Compound G2 showed important inhibitory effects on actin bundling, filopodia formation, migration, and invasion in different cell lines. Moreover, compound G2 treatment resulted in significant reduction of invasion of DLD-1 overexpressing Fascin1 and HCT-116 in zebrafish larvae xenografts; this effect being less evident in Fascin1 known-down HCT-116 cells. This study proves, for the first time, the in vitro and in vivo anti-tumoral activity of compound G2 on colorectal cancer cells and guides to design improved compound G2-based Fascin1 inhibitors. KEY MESSAGES: • Fascin is crucial for tumor invasion and metastasis and is overexpressed in bad prognostic tumors. • Several adverse tumors overexpress Fascin1 and lack targeted therapy. • Anti-fascin G2 is for the first time evaluated in colorectal carcinoma and compared with migrastatin. • Filopodia formation, migration activity, and invasion in vitro and in vivo assays were performed. • G2 blocks actin structures, migration, and invasion of colorectal cancer cells as fascin-dependent.
Collapse
Affiliation(s)
- Silvia Montoro-García
- Cell Culture Lab. Health Faculty, Universidad Católica de Murcia (UCAM), Campus de los Jerónimos, s/n, Guadalupe, 30107, Murcia, Spain.
| | - Begoña Alburquerque-González
- Pathology and Histology Department. Heatlh Faculty, Universidad Católica de Murcia (UCAM), Campus de los Jerónimos, s/n, Guadalupe, 30107, Murcia, Spain
| | - Ángel Bernabé-García
- Molecular Oncology and TGF-ß Lab, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Carretera Madrid-Cartagena. El Palmar, Murcia, Spain
| | - Manuel Bernabé-García
- Telomerase, Cancer and Aging Group, University Clinical Hospital "Virgen de la Arrixaca", Biomedical Research Institute of Murcia (IMIB-Arrixaca) Murcia, Murcia, Spain
| | - Priscila Campioni Rodrigues
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Aapistie 5A, FI-90220, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Helena den-Haan
- Eurofins Villapharma Research, Parque Tecnológico de Fuente Álamo. Ctra. El Estrecho-Lobosillo, Km 2,5. Av. Azul E, 30320, Murcia, Spain
| | - Irene Luque
- Department of Physical Chemistry and Institute of Biotechnology, University of Granada, Campus Fuentenueva s/n 18071 Granada, Granada, Spain
| | - Francisco José Nicolás
- Molecular Oncology and TGF-ß Lab, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Carretera Madrid-Cartagena. El Palmar, Murcia, Spain
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High Performance Computing (BIO-HPC) Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, Spain
| | - María Luisa Cayuela
- Telomerase, Cancer and Aging Group, University Clinical Hospital "Virgen de la Arrixaca", Biomedical Research Institute of Murcia (IMIB-Arrixaca) Murcia, Murcia, Spain
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Aapistie 5A, FI-90220, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.,Institute of Oral and Maxillofacial Disease, University of Helsinki, Helsinki, Finland.,HUSLAB, Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| | - Pablo Conesa-Zamora
- Pathology and Histology Department. Heatlh Faculty, Universidad Católica de Murcia (UCAM), Campus de los Jerónimos, s/n, Guadalupe, 30107, Murcia, Spain. .,Clinical Analysis Department, Group of Molecular Pathology and Pharmacogenetics, Biomedical Research Institute from Murcia (IMIB), Hospital Universitario Santa Lucía, c/Mezquita sn, 30202, Cartagena, Spain.
| |
Collapse
|
14
|
He C, Danes JM, Hart PC, Zhu Y, Huang Y, de Abreu AL, O'Brien J, Mathison AJ, Tang B, Frasor JM, Wakefield LM, Ganini D, Stauder E, Zielonka J, Gantner BN, Urrutia RA, Gius D, Bonini MG. SOD2 acetylation on lysine 68 promotes stem cell reprogramming in breast cancer. Proc Natl Acad Sci U S A 2019; 116:23534-23541. [PMID: 31591207 PMCID: PMC6876149 DOI: 10.1073/pnas.1902308116] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial superoxide dismutase (SOD2) suppresses tumor initiation but promotes invasion and dissemination of tumor cells at later stages of the disease. The mechanism of this functional switch remains poorly defined. Our results indicate that as SOD2 expression increases acetylation of lysine 68 ensues. Acetylated SOD2 promotes hypoxic signaling via increased mitochondrial reactive oxygen species (mtROS). mtROS, in turn, stabilize hypoxia-induced factor 2α (HIF2α), a transcription factor upstream of "stemness" genes such as Oct4, Sox2, and Nanog. In this sense, our findings indicate that SOD2K68Ac and mtROS are linked to stemness reprogramming in breast cancer cells via HIF2α signaling. Based on these findings we propose that, as tumors evolve, the accumulation of SOD2K68Ac turns on a mitochondrial pathway to stemness that depends on HIF2α and may be relevant for the progression of breast cancer toward poor outcomes.
Collapse
Affiliation(s)
- Chenxia He
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jeanne M Danes
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Peter C Hart
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612
| | - Yueming Zhu
- Department of Radiation Oncology, Northwestern University, Chicago, IL 60657
| | - Yunping Huang
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI 53226
| | | | - Joseph O'Brien
- Department of Radiation Oncology, Northwestern University, Chicago, IL 60657
| | - Angela J Mathison
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Binwu Tang
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | - Jonna M Frasor
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612
| | - Lalage M Wakefield
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | - Douglas Ganini
- Free Radical Metabolism Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
| | - Erich Stauder
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Benjamin N Gantner
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Raul A Urrutia
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - David Gius
- Department of Radiation Oncology, Northwestern University, Chicago, IL 60657
| | - Marcelo G Bonini
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, Milwaukee, WI 53226;
| |
Collapse
|
15
|
Zhang N, Gao L, Ren W, Li S, Zhang D, Song X, Zhao C, Zhi K. Fucoidan affects oral squamous cell carcinoma cell functions in vitro by regulating FLNA-derived circular RNA. Ann N Y Acad Sci 2019; 1462:65-78. [PMID: 31495936 DOI: 10.1111/nyas.14190] [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: 01/21/2019] [Revised: 05/17/2019] [Accepted: 06/18/2019] [Indexed: 12/31/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common cancer types, with a high annual incidence. Although chemotherapy contributes to suppressing OSCC tumorigenesis, the available treatments result in poor prognosis because of local recurrence and regional lymph node metastasis. Thus, it is necessary to discover novel and safe drugs with greater effectiveness and fewer side effects. Fucoidan is a component of the cell wall of brown seaweed that has been shown to produce a wide range of biological activities. The present study aimed to investigate the effectiveness of fucoidan in treating OSCC. In in vitro studies, we found that fucoidan inhibited OSCC growth and suppressed migration and invasion of OSCC cells. In addition, the potential interaction between fucoidan and filamin A (FLNA)-derived circular RNA (circFLNA) was predicted using bioinformatics databases and then confirmed in OSCC samples and cell lines. Indeed, fucoidan increased the expression of circFLNA in OSCC cell lines. Furthermore, both fucoidan and circFLNA could mediate the expression of key proteins related to cell growth, apoptosis, migration, and invasion. In conclusion, our research demonstrated that fucoidan might be considered as a potential natural drug in the treatment of OSCC patients by targeting circFLNA.
Collapse
Affiliation(s)
- Nanyang Zhang
- Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ling Gao
- Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Implantology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Oral Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenhao Ren
- Department of Implantology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Oral Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shaoming Li
- Department of Implantology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Oral Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Demeng Zhang
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao Brightmoon Seaweed Group Co Ltd, Qingdao, Shandong, China
| | - Xianqin Song
- Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chenyang Zhao
- Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Keqian Zhi
- Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Implantology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Oral Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
16
|
Ardisia crispa root hexane fraction suppressed angiogenesis in human umbilical vein endothelial cells (HUVECs) and in vivo zebrafish embryo model. Biomed Pharmacother 2019; 118:109221. [PMID: 31545225 DOI: 10.1016/j.biopha.2019.109221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/08/2023] Open
Abstract
Ardisia crispa Thunb. A. DC. (Primulaceae) has been used extensively as folk-lore medicine in South East Asia including China and Japan to treat various inflammatory related diseases. Ardisia crispa root hexane fraction (ACRH) has been thoroughly studied by our group and it has been shown to exhibit anti-inflammatory, anti-hyperalgesic, anti-arthritic, anti-ulcer, chemoprevention and suppression against inflammation-induced angiogenesis in various animal model. Nevertheless, its effect against human endothelial cells in vitro has not been reported yet. Hence, the aim of the study is to investigate the potential antiangiogenic property of ACRH in human umbilical vein endothelial cells (HUVECs) and zebrafish embryo model. ACRH was separated from the crude ethanolic extract of the plant's root in prior to experimental studies. MTT assay revealed that ACRH exerted a concentration-dependent antiproliferative effect on HUVEC with the IC50 of 2.49 ± 0.04 μg/mL. At higher concentration (10 μg/mL), apoptosis was induced without affecting the cell cycle distribution. Angiogenic properties including migration, invasion and differentiation of HUVECs, evaluated via wound healing, trans-well invasion and tube formation assay respectively, were significantly suppressed by ACRH in a concentration-dependent manner. Noteworthily, significant antiangiogenic effects were observed even at the lowest concentration used (0.1 μg/mL). Expression of proMMP-2, vascular endothelial growth factor (VEGF)-C, VEGF-D, Angiopoietin-2, fibroblast growth factor (FGF)-1, FGF-2, Follistatin, and hepatocyte growth factor (HGF) were significantly reduced in various degrees by ACRH. The ISV formation in zebrafish embryo was significantly suppressed by ACRH at the concentration of 5 μg/mL. These findings revealed the potential of ACRH as antiangiogenic agent by suppressing multiple proangiogenic proteins. Thus, it can be further verified via the transcription of these proteins from their respective DNA, in elucidating their exact pathways.
Collapse
|
17
|
Ampelopsin E Reduces the Invasiveness of the Triple Negative Breast Cancer Cell Line, MDA-MB-231. Molecules 2019; 24:molecules24142619. [PMID: 31323836 PMCID: PMC6680398 DOI: 10.3390/molecules24142619] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the most common and the second leading cause of cancer-related deaths in women. It has two distinctive hallmarks: rapid abnormal growth and the ability to invade and metastasize. During metastasis, cancer cells are thought to form actin-rich protrusions, called invadopodia, which degrade the extracellular matrix. Current breast cancer treatments, particularly chemotherapy, comes with adverse effects like immunosuppression, resistance development and secondary tumour formation. Hence, naturally-occurring molecules claimed to be less toxic are being studied as new drug candidates. Ampelopsin E, a natural oligostilbene extracted from Dryobalanops species, has exhibited various pharmacological properties, including anticancer and anti-inflammatory activities. However, there is yet no scientific evidence of the effects of ampelopsin E towards metastasis. Scratch assay, transwell migration and invasion assays, invadopodia and gelatin degradation assays, and ELISA were used to determine the effects of ampelopsin E towards the invasiveness of MDA-MB-231 cells. Strikingly in this study, ampelopsin E was able to halt migration, transmigration and invasion in MDA-MB-231 cells by reducing formation of invadopodia and its degradation capability through significant reduction (p < 0.05) in expression levels of PDGF, MMP2, MMP9 and MMP14. In conclusion, ampelopsin E reduced the invasiveness of MDA-MB-231 cells and was proven to be a potential alternative in treating TNBC.
Collapse
|
18
|
Lee G, Han SB, Lee JH, Kim HW, Kim DH. Cancer Mechanobiology: Microenvironmental Sensing and Metastasis. ACS Biomater Sci Eng 2019; 5:3735-3752. [PMID: 33405888 DOI: 10.1021/acsbiomaterials.8b01230] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cellular microenvironment plays an important role in regulating cancer progress. Cancer can physically and chemically remodel its surrounding extracellular matrix (ECM). Critical cellular behaviors such as recognition of matrix geometry and rigidity, cell polarization and motility, cytoskeletal reorganization, and proliferation can be changed as a consequence of these ECM alternations. Here, we present an overview of cancer mechanobiology in detail, focusing on cancer microenvironmental sensing of exogenous cues and quantification of cancer-substrate interactions. In addition, mechanics of metastasis classified with tumor progression will be discussed. The mechanism underlying cancer mechanosensation and tumor progression may provide new insights into therapeutic strategies to alleviate cancer malignancy.
Collapse
Affiliation(s)
- GeonHui Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, South Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, South Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, South Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| |
Collapse
|
19
|
Stojanović N, Dekanić A, Paradžik M, Majhen D, Ferenčak K, Ruščić J, Bardak I, Supina C, Tomicic MT, Christmann M, Osmak M, Ambriović-Ristov A. Differential Effects of Integrin αv Knockdown and Cilengitide on Sensitization of Triple-Negative Breast Cancer and Melanoma Cells to Microtubule Poisons. Mol Pharmacol 2018; 94:1334-1351. [DOI: 10.1124/mol.118.113027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/20/2018] [Indexed: 01/03/2023] Open
|
20
|
Kabała-Dzik A, Rzepecka-Stojko A, Kubina R, Wojtyczka RD, Buszman E, Stojko J. Caffeic Acid Versus Caffeic Acid Phenethyl Ester in the Treatment of Breast Cancer MCF-7 Cells: Migration Rate Inhibition. Integr Cancer Ther 2018; 17:1247-1259. [PMID: 30246565 PMCID: PMC6247537 DOI: 10.1177/1534735418801521] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Epithelium mammary carcinoma is a cancer with a high death rate among women. One factor having a significant impact on metastasis is cell migration. The aim of this study was to compare migration rate inhibition of caffeic acid (CA) and its phenethyl ester (CAPE) on MCF-7 breast cancer cells. Microscopic evaluation was used to determine the morphology of carcinoma cells, before and after 24-hour treatment with CA and CAPE using a dose of 50 µM. The cytotoxic effect was measured by XTT-NR-SRB assay (tetrazolium hydroxide-neutral red-Sulforhodamine B) for 24-hour and 48-hour periods, using CA and CAPE, with doses of 50 and 100 µM. These doses were used to determine cell migration inhibition using a wound closure assay for 0-hour, 8-hour, 16-hour, and 24-hour periods. Both CA and CAPE treatments displayed cytotoxic activity in a dose- and time-dependent trend. CAPE displayed IC50 values more than twice as low as CA. IC50 values for the XTT assay were as follows: CA was 102.98 µM for 24 hours and 59.12 µM for 48 hours, while CAPE was 56.39 µM for 24 hours and 28.10 µM for 48 hours. For the NR assay: CA was 84.87 µM at 24 hours and 65.05 µM at 48 hours, while CAPE was 69.05 µM at 24 hours and 29.05 µM at 48 hours. For the SRB assay: At 24 hours, CA was 83.47 µM and 53.46 µM at 48 hours, while CAPE was 38.53 µM at 24 hours and 20.15 µM at 48 hours. Both polyphenols induced migration inhibition, resulting in practically halting the wound closure. CAPE produced better results than CA with the same doses and experiment times, though both CA and CAPE displayed cytotoxic activity against MCF-7 cells, as well as inhibited migration.
Collapse
Affiliation(s)
- Agata Kabała-Dzik
- 1 Medical University of Silesia in Katowice, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Poland
| | - Anna Rzepecka-Stojko
- 1 Medical University of Silesia in Katowice, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Poland
| | - Robert Kubina
- 1 Medical University of Silesia in Katowice, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Poland
| | - Robert Dariusz Wojtyczka
- 1 Medical University of Silesia in Katowice, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Poland
| | - Ewa Buszman
- 1 Medical University of Silesia in Katowice, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Poland
| | - Jerzy Stojko
- 1 Medical University of Silesia in Katowice, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Poland
| |
Collapse
|
21
|
Development of pipette tip gap closure migration assay (s-ARU method) for studying semi-adherent cell lines. Cytotechnology 2018; 70:1685-1695. [PMID: 30069611 DOI: 10.1007/s10616-018-0245-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 07/25/2018] [Indexed: 10/28/2022] Open
Abstract
This work presents a pipette tip gap closure migration assay prototype tool (semi-adherent relative upsurge-s-ARU-method) to study cell migration or wound healing in semi-adherent cell lines, such as lymph node carcinoma of the prostate (LNCaP). Basically, it consists of a 6-well cover plate modification, where pipette tips with the filter are shortened and fixed vertically to the inner surface of the cover plate, with their heights adjusted to touch the bottom of the well center. This provides a barrier for the inoculated cells to grow on, creating a cell-free gap. Such a uniform gap formed can be used to study migration assay for both adherent as well as semi-adherent cells. After performing time studies, effective measurement of gap area can be carried out conveniently through image analysis software. Here, the prototype was tested for LNCaP cells, treated with testosterone and flutamide as well as with bacteriophages T4 and M13. A scratch assay using PC3 adherent cells was also performed for comparison. It was observed that s-ARU method is suitable for studying LNCaP cells migration assay, as observed from our results with testosterone, flutamide, and bacteriophages (T4 and M13). Our method is a low-cost handmade prototype, which can be an alternative to the other migration assay protocol(s) for both adherent and semi-adherent cell cultures in oncological research along with other biological research applications.
Collapse
|
22
|
Gillette KM, Forbes K, Sehgal I. Detection of Matrix Metalloproteinases (MMP), Tissue Inhibitor of Metalloproteinase-2, Urokinase and Plasminogen Activator Inhibitor-1 within Matrigel and Growth Factor-Reduced Matrigel Basement Membrane. TUMORI JOURNAL 2018; 89:421-5. [PMID: 14606648 DOI: 10.1177/030089160308900415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Matrigel (MG) basement membrane is commonly used for in vitro studies of cellular migration, invasion and angiogenesis. It contains structural molecules such as collagen IV and laminin plus several growth factors which are diminished in growth factor-reduced Matrigel (GFR-MG). A less appreciated but important aspect of MG is the presence of matrix enzymes and their inhibitors. For relevant interpretation of data using MG/GFR-MG models, it may be necessary to know the enzymes or inhibitors contributed by these basement membranes themselves. Methods Immunoblot and zymography were used to detect the presence or absence of MMP-1 and 7, tissue inhibitor of metalloproteinase 1 and 2 (TIMP-1, TIMP-2), plasminogen activator activity and plasminogen activator inhibitor-1 (PAI-1). Growth and invasion assays using prostate cancer cells were used to assess the effects of TIMP-2 presence or absence. Results We detected MMP-7, urokinase plasminogen activator (uPA) and PAI-1 in both Matrigels, TIMP-2 was detected only in regular Matrigel and no MMP-1 or TIMP-1 was detected in either matrix. Invasion assays comparing regular MG and GFR-MG indicated cell line variability with regard to invasion efficiency as two tested prostate cancer lines were unaffected by the MG type while one was significantly more invasive in regular MG. Growth experiments suggest that the presence of TIMP-2 in regular MG may retard growth but overall proliferation is still greater in regular MG than in GFR-MG. Conclusions These data provide a useful reference for interpretation of in vitro Matrigel assays.
Collapse
Affiliation(s)
- Karin M Gillette
- Center for Protease Research, North Dakota State University College of Pharmacy, Fargo, North Dakota, USA
| | | | | |
Collapse
|
23
|
Abstract
Tumor-induced angiogenesis is a key event for neoplastic progression. In vitro assays are important for identification of potential angiogenic agents and rapid sceening for pharmacological inhibitors. The increased interest in this field of study has generated several in vitro assays that recapitulate the steps of endothelial cell activation and differentiation. In this short report we emphasize the utility of Matrigel, a reconstituted basement membrane, to define two different steps in the angiogenic process: invasion in response to growth factors and organization of microvessels into a network with branching morphology on a Matrigel substrate.
Collapse
Affiliation(s)
- R Benelli
- Molecular Oncology Service, Advanced Biotechnologies Center, Genova, Italy
| | | |
Collapse
|
24
|
Nuclear Phosphatidylinositol-Phosphate Type I Kinase α-Coupled Star-PAP Polyadenylation Regulates Cell Invasion. Mol Cell Biol 2018; 38:MCB.00457-17. [PMID: 29203642 PMCID: PMC5809686 DOI: 10.1128/mcb.00457-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/27/2017] [Indexed: 01/15/2023] Open
Abstract
Star-PAP, a nuclear phosphatidylinositol (PI) signal-regulated poly(A) polymerase (PAP), couples with type I PI phosphate kinase α (PIPKIα) and controls gene expression. We show that Star-PAP and PIPKIα together regulate 3′-end processing and expression of pre-mRNAs encoding key anti-invasive factors (KISS1R, CDH1, NME1, CDH13, FEZ1, and WIF1) in breast cancer. Consistently, the endogenous Star-PAP level is negatively correlated with the cellular invasiveness of breast cancer cells. While silencing Star-PAP or PIPKIα increases cellular invasiveness in low-invasiveness MCF7 cells, Star-PAP overexpression decreases invasiveness in highly invasive MDA-MB-231 cells in a cellular Star-PAP level-dependent manner. However, expression of the PIPKIα-noninteracting Star-PAP mutant or the phosphodeficient Star-PAP (S6A mutant) has no effect on cellular invasiveness. These results strongly indicate that PIPKIα interaction and Star-PAP S6 phosphorylation are required for Star-PAP-mediated regulation of cancer cell invasion and give specificity to target anti-invasive gene expression. Our study establishes Star-PAP–PIPKIα-mediated 3′-end processing as a key anti-invasive mechanism in breast cancer.
Collapse
|
25
|
Fatima T, Haque RA, Iqbal MA, Ahmad A, Hassan LEA, Taleb-Agha M, Ahamed MBK, Majid AA, Razali MR. Tetra N -heterocyclic carbene dinuclear silver(I) complexes as potential anticancer agents: Synthesis and in vitro anticancer studies. J Organomet Chem 2017. [DOI: 10.1016/j.jorganchem.2017.10.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
Blaha L, Zhang C, Cabodi M, Wong JY. A microfluidic platform for modeling metastatic cancer cell matrix invasion. Biofabrication 2017; 9:045001. [PMID: 28812983 DOI: 10.1088/1758-5090/aa869d] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Invasion of the extracellular matrix is a critical step in the colonization of metastatic tumors. The invasion process is thought to be driven by both chemokine signaling and interactions between invading cancer cells and physical components of the metastatic niche, including endothelial cells that line capillary walls and serve as a barrier to both diffusion and invasion of the underlying tissue. Transwell chambers, a tool for generating artificial chemokine gradients to induce cell migration, have facilitated recent work to investigate the chemokine contributions to matrix invasion. These chambers, however, are poorly designed for imaging, which limits their use in investigating the physical cell-cell and cell-matrix interactions driving matrix invasion. Microfluidic devices offer a promising model in which the invasion process can be imaged. Many current designs, however, have limited surface areas and possess intricate geometries that preclude the use of standard staining protocols to visualize cells and matrix proteins. In this work, we present a novel microfluidic platform for imaging cell-cell and cell-matrix interactions driving metastatic cancer cell matrix invasion. Our model is applied to investigate how endothelial cell-secreted matrix proteins and the physical endothelial monolayer itself interact with invading metastatic breast cancer cells to facilitate invasion of an underlying type I collagen gel. The results show that matrix invasion of metastatic breast cancer cells is significantly enhanced in the presence of live endothelial cells. Probing this interaction further, our platform revealed that, while the fibronectin-rich matrix deposited by endothelial cells was not sufficient to drive invasion alone, metastatic breast cancer cells were able to exploit components of energetically inactivated endothelial cells to gain entry into the underlying matrix. These findings reveal novel cell-cell interactions driving a key step in the colonization of metastatic tumors and have important implications for designing drugs targeted at preventing cancer metastasis.
Collapse
Affiliation(s)
- Laura Blaha
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States of America
| | | | | | | |
Collapse
|
27
|
He Y, Xue H, Zhang W, Wang L, Xiang G, Li L, Shang X. Discovery of a series of ruthenium(II) derivatives with α -dicarbonylmonoxime as novel inhibitors of cancer cells invasion and metastasis. J Organomet Chem 2017. [DOI: 10.1016/j.jorganchem.2017.05.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
28
|
Wang D, Xin Y, Tian Y, Li W, Sun D, Yang Y. Pseudolaric acid B inhibits gastric cancer cell metastasis in vitro and in haematogenous dissemination model through PI3K/AKT, ERK1/2 and mitochondria-mediated apoptosis pathways. Exp Cell Res 2017; 352:34-44. [PMID: 28132880 DOI: 10.1016/j.yexcr.2017.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 01/15/2017] [Accepted: 01/25/2017] [Indexed: 12/30/2022]
Abstract
Pseudolaric acid B (PAB) is the major bioactive constituent in the root bark of Pseudolarix kaempferi and has been reported to have cytotoxicity against tumor cells. Our in vivo experiments showed that PAB could inhibit gastric cancer cell lung metastasis in a nude mouse haematogenous dissemination model. To evaluate the anti-metastasis mechanism of PAB in gastric cancer cells, cytological experiments were performed. The results showed that PAB could inhibit the adhesion ability to matrigel, migration, invasion and colony formation ability of BGC-823 and MKN-45 cells. Western blot further confirmed that the inhibitory effects of PAB on anti-metastasis may involve regulating the expression of the metastasis-related proteins MMP-9, HIF-1α, VEGF, VEGFR2, E-Cadherin and Ezrin. We obtained further proof that PAB which could be used as a multi-targeted agent to inhibit the PI3K/AKT, ERK1/2 and mitochondria-mediated apoptosis pathways and consequently suppress tumor growth and metastasis. Our experiments suggest that PAB-induced effects may have novel therapeutic applications for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Dan Wang
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China; Colledge of Pharmacy, Liaoning University, Shenyang, China.
| | - Yan Xin
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China
| | - Yanqiu Tian
- Colledge of Life Science, Liaoning University, Shenyang, China
| | - Wenhui Li
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China
| | - Dan Sun
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China
| | - Yi Yang
- Laboratory Animal Center, China Medical University, Shenyang, China
| |
Collapse
|
29
|
Kwong WL, Lam KY, Lok CN, Lai YT, Lee PY, Che CM. A Macrocyclic Ruthenium(III) Complex Inhibits Angiogenesis with Down-Regulation of Vascular Endothelial Growth Factor Receptor-2 and Suppresses Tumor Growth In Vivo. Angew Chem Int Ed Engl 2016; 55:13524-13528. [DOI: 10.1002/anie.201608094] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/07/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Wai-Lun Kwong
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Kar-Yee Lam
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Yau-Tsz Lai
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Pui-Yan Lee
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
- HKU Shenzhen Institute of Research and Innovation; Shenzen 518053 P.R. China
| |
Collapse
|
30
|
Kwong WL, Lam KY, Lok CN, Lai YT, Lee PY, Che CM. A Macrocyclic Ruthenium(III) Complex Inhibits Angiogenesis with Down-Regulation of Vascular Endothelial Growth Factor Receptor-2 and Suppresses Tumor Growth In Vivo. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201608094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Wai-Lun Kwong
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Kar-Yee Lam
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Yau-Tsz Lai
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Pui-Yan Lee
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
- HKU Shenzhen Institute of Research and Innovation; Shenzen 518053 P.R. China
| |
Collapse
|
31
|
Aptamer targeting EGFRvIII mutant hampers its constitutive autophosphorylation and affects migration, invasion and proliferation of glioblastoma cells. Oncotarget 2016; 6:37570-87. [PMID: 26461476 PMCID: PMC4741949 DOI: 10.18632/oncotarget.6066] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/23/2015] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma Multiforme (GBM) is the most common and aggressive human brain tumor, associated with very poor survival despite surgery, radiotherapy and chemotherapy.The epidermal growth factor receptor (EGFR) and the platelet-derived growth factor receptor β (PDGFRβ) are hallmarks in GBM with driving roles in tumor progression. In approximately half of the tumors with amplified EGFR, the EGFRvIII truncated extracellular mutant is detected. EGFRvIII does not bind ligands, is highly oncogenic and its expression confers resistance to EGFR tyrosine kinase inhibitors (TKIs). It has been demonstrated that EGFRvIII-dependent cancers may escape targeted therapy by developing dependence on PDGFRβ signaling, thus providing a strong rationale for combination therapy aimed at blocking both EGFRvIII and PDGFRβsignaling.We have recently generated two nuclease resistant RNA aptamers, CL4 and Gint4.T, as high affinity ligands and inhibitors of the human wild-type EGFR (EGFRwt) and PDGFRβ, respectively.Herein, by different approaches, we demonstrate that CL4 aptamer binds to the EGFRvIII mutant even though it lacks most of the extracellular domain. As a consequence of binding, the aptamer inhibits EGFRvIII autophosphorylation and downstream signaling pathways, thus affecting migration, invasion and proliferation of EGFRvIII-expressing GBM cell lines.Further, we show that targeting EGFRvIII by CL4, as well as by EGFR-TKIs, erlotinib and gefitinib, causes upregulation of PDGFRβ. Importantly, CL4 and gefitinib cooperate with the anti-PDGFRβ Gint4.T aptamer in inhibiting cell proliferation.The proposed aptamer-based strategy could have impact on targeted molecular cancer therapies and may result in progresses against GBMs.
Collapse
|
32
|
Thejass P, Kuttan G. Inhibition of Endothelial Cell Differentiation and Proinflammatory Cytokine Production During Angiogenesis by Allyl Isothiocyanate and Phenyl Isothiocyanate. Integr Cancer Ther 2016; 6:389-99. [DOI: 10.1177/1534735407309084] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is a crucial step in the growth and metastasis of cancers. The activation of endothelial cells and their further behavior are very critical during angiogenesis. The authors analyze the effect of allyl isothiocyanate (AITC) and phenyl isothiocyanate (PITC) on angiogenesis in an in vitro model using human umbilical vein endothelial cells (HUVECs). AITC and PITC significantly inhibited endothelial cell migration, invasion, and tube formation. 3H-thymidine proliferation assay showed that AITC and PITC significantly inhibited the proliferation of HUVECs in vitro. The authors also studied the effect of AITC and PITC on the serum cytokine profiles of angiogenesis-induced animals and found that these compounds are highly potent in the downregulation of vascular endothelial growth factor (VEGF) and proinflammatory cytokines such as interleukin (IL)—1β , IL-6, granulocyte macrophage colony-stimulating factor (GM-CSF), and tumor necrosis factor α (TNF-α). Treatment with these compounds showed an elevation in the levels of IL-2 and tissue inhibitor of metalloproteinases (TIMP)—1, which are antiangiogenic factors. Moreover, studies using B16F-10 melanoma cells showed that both AITC and PITC significantly reduced VEGF mRNA expression. These findings suggest that AITC and PITC act as angiogenesis inhibitors through the downregulation of VEGF and proinflammatory cytokines such as IL-1β, IL-6, GM-CSF, and TNF-α and upregulation of IL-2 and TIMP.
Collapse
Affiliation(s)
- P. Thejass
- Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala State, India
| | - Girija Kuttan
- Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala State, India,
| |
Collapse
|
33
|
Li C, Yang D, Zhao Y, Qiu Y, Cao X, Yu Y, Guo H, Gu X, Yin X. Inhibitory Effects of Isorhamnetin on the Invasion of Human Breast Carcinoma Cells by Downregulating the Expression and Activity of Matrix Metalloproteinase-2/9. Nutr Cancer 2015; 67:1191-200. [DOI: 10.1080/01635581.2015.1073763] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Dan Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Yuanwei Zhao
- The People's Hospital of Pizhou, Xuzhou, People's Republic of China
| | - Yu Qiu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Xin Cao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Hao Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Xuzhou, People's Republic of China
| |
Collapse
|
34
|
Klueh U, Qiao Y, Czajkowski C, Ludzinska I, Antar O, Kreutzer DL. Basement Membrane-Based Glucose Sensor Coatings Enhance Continuous Glucose Monitoring in Vivo. J Diabetes Sci Technol 2015; 9:957-65. [PMID: 26306494 PMCID: PMC4667328 DOI: 10.1177/1932296815598776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Implantable glucose sensors demonstrate a rapid decline in function that is likely due to biofouling of the sensor. Previous efforts directed at overcoming this issue has generally focused on the use of synthetic polymer coatings, with little apparent effect in vivo, clearly a novel approach is required. We believe that the key to extending sensor life span in vivo is the development of biocompatible basement membrane (BM) based bio-hydrogels as coatings for glucose sensors. METHOD BM based bio-hydrogel sensor coatings were developed using purified BM preparations (ie, Cultrex from Trevigen Inc). Modified Abbott sensors were coated with Cultrex BM extracts. Sensor performance was evaluated for the impact of these coatings in vitro and in vivo in a continuous glucose monitoring (CGM) mouse model. In vivo sensor function was assessed over a 28-day time period expressed as mean absolute relative difference (MARD) values. Tissue reactivity of both Cultrex coated and uncoated glucose sensors was evaluated at 7, 14, 21 and 28 days post-sensor implantation with standard histological techniques. RESULTS The data demonstrate that Cultrex-based sensor coatings had no effect on glucose sensor function in vitro. In vivo glucose sensor performance was enhanced following BM coating as determined by MARD analysis, particularly in weeks 2 and 3. In vivo studies also demonstrated that Cultrex coatings significantly decreased sensor-induced tissue reactions at the sensor implantation sites. CONCLUSION Basement-membrane-based sensor coatings enhance glucose sensor function in vivo, by minimizing or preventing sensor-induced tissues reactions.
Collapse
Affiliation(s)
- Ulrike Klueh
- Center for Molecular Tissue Engineering, University of Connecticut, School of Medicine, Farmington, CT, USA Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT, USA
| | - Yi Qiao
- Center for Molecular Tissue Engineering, University of Connecticut, School of Medicine, Farmington, CT, USA Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT, USA
| | - Caroline Czajkowski
- Center for Molecular Tissue Engineering, University of Connecticut, School of Medicine, Farmington, CT, USA Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT, USA
| | - Izabela Ludzinska
- Center for Molecular Tissue Engineering, University of Connecticut, School of Medicine, Farmington, CT, USA Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT, USA
| | - Omar Antar
- Center for Molecular Tissue Engineering, University of Connecticut, School of Medicine, Farmington, CT, USA Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT, USA
| | - Donald L Kreutzer
- Center for Molecular Tissue Engineering, University of Connecticut, School of Medicine, Farmington, CT, USA Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT, USA
| |
Collapse
|
35
|
Wang Y, Wang LY, Feng F, Zhao Y, Huang MY, Shao Q, Chen C, Sheng H, Chen DL, Zeng ZL, Xu RH, Li YH. Effect of Raf kinase inhibitor protein expression on malignant biological behavior and progression of colorectal cancer. Oncol Rep 2015; 34:2106-14. [PMID: 26238523 DOI: 10.3892/or.2015.4157] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/06/2015] [Indexed: 11/05/2022] Open
Abstract
The Raf kinase inhibitor protein (RKIP) is a novel metastasis suppressor. RKIP was previously found to have low expression in a colorectal cancer (CRC) patient cohort by immunohistochemistry. However, the role of RKIP in CRC remains undetermined. In the present study, immunohistochemistry was performed to compare RKIP expression between 129 paired stage II CRC and adjacent non-tumorous tissues. The correlations between clinical parameters, prognosis and RKIP expression were evaluated. To investigate the effect of RKIP on proliferation and metastasis, RKIP was overexpressed and knocked down in colon cancer cell lines. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), Transwell and wound-healing assays were performed. Murine models were established to confirm the influence of RKIP on malignant tumor phenotypes in vivo. Our results showed that RKIP expression was significantly decreased in the CRC tissues compared to the adjacent non‑cancerous tissues (p<0.001) and was correlated with the risk of relapse in stage II CRC (p<0.05). Overexpression of RKIP suppressed HCT116 cell metastasis in vitro and in vivo, whereas knockdown of RKIP expression in SW480 cells and its murine model increased metastatic ability (p<0.05). No effect of RKIP on cell proliferation in CRC was observed. These data suggest that RKIP is an important metastasis-suppressor gene in CRC. The re-expression of RKIP could be a potential therapeutic target for antimetastatic strategies for CRC.
Collapse
Affiliation(s)
- Yun Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Ling-Yun Wang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Fen Feng
- Department of Oncology, The First People's Hospital of Foshan City, Foshan, Guangdong, P.R. China
| | - Yang Zhao
- Department of Anesthesiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Ma-Yan Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Qiong Shao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Cui Chen
- Department of Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Hui Sheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Dong-Liang Chen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Zhao-Lei Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Yu-Hong Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| |
Collapse
|
36
|
Cyclodextrin mediated delivery of NF-κB and SRF siRNA reduces the invasion potential of prostate cancer cells in vitro. Gene Ther 2015; 22:802-10. [PMID: 26005860 DOI: 10.1038/gt.2015.50] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/08/2015] [Accepted: 05/12/2015] [Indexed: 02/01/2023]
Abstract
Prostate cancer is the most common cancer in men of the western world. To date, no effective treatment exists for metastatic prostate cancer and consequently, there is an urgent need to develop new and improved therapeutics. In recent years, the therapeutic potential of RNA interference (RNAi) has been extensively explored in a wide range of diseases including prostate cancer using numerous gene delivery vectors. The aims of this study were to investigate the ability of a non-viral modified cyclodextrin (CD) vector to deliver siRNA to the highly metastatic PC-3 prostate cancer cell line, to quantify the resulting knockdown of the two target genes (RelA and SRF) and to study the effects of the silencing on metastasis. Data from a Matrigel in vitro invasion assay indicated that the silencing of the target genes achieved by the CD vector resulted in significant reductions (P=0.0001) in the metastatic potential of these cells. As the silencing of these target genes was shown not to have a negative impact on cell viability, we hypothesise that the mechanism of invasion inhibition is due, in part, to the significant reduction observed (P⩽0.0001) in the level of pro-inflammatory cytokine, MMP9, which is known to be implicated in the metastasis of prostate cancer.
Collapse
|
37
|
Chiappetta G, Valentino T, Vitiello M, Pasquinelli R, Monaco M, Palma G, Sepe R, Luciano A, Pallante P, Palmieri D, Aiello C, Rea D, Losito SN, Arra C, Fusco A, Fedele M. PATZ1 acts as a tumor suppressor in thyroid cancer via targeting p53-dependent genes involved in EMT and cell migration. Oncotarget 2015; 6:5310-23. [PMID: 25595894 PMCID: PMC4467151 DOI: 10.18632/oncotarget.2776] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/18/2014] [Indexed: 02/06/2023] Open
Abstract
PATZ1, a POZ-Zinc finger protein, is emerging as an important regulator of development and cancer, but its cancer-related function as oncogene or tumor-suppressor is still debated. Here, we investigated its possible role in thyroid carcinogenesis. We demonstrated PATZ1 is down-regulated in thyroid carcinomas compared to normal thyroid tissues, with an inverse correlation to the degree of cell differentiation. In fact, PATZ1 expression was significantly further down-regulated in poorly differentiated and anaplastic thyroid cancers compared to the papillary histotype, and it resulted increasingly delocalized from the nucleus to the cytoplasm proceeding from differentiated to undifferentiated thyroid carcinomas. Restoration of PATZ1 expression in three thyroid cancer-derived cell lines, all characterized by fully dedifferentiated cells, significantly inhibited their malignant behaviors, including in vitro proliferation, anchorage-independent growth, migration and invasion, as well as in vivo tumor growth. Consistent with recent studies showing a role for PATZ1 in the p53 pathway, we showed that ectopic expression of PATZ1 in thyroid cancer cells activates p53-dependent pathways opposing epithelial-mesenchymal transition and cell migration to prevent invasiveness. These results provide insights into a potential tumor-suppressor role of PATZ1 in thyroid cancer progression, and thus may have potential clinical relevance for the prognosis and therapy of thyroid cancer.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blotting, Western
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/pathology
- Cell Movement
- Cell Proliferation
- Chromatin Immunoprecipitation
- Epithelial-Mesenchymal Transition
- Female
- Genes, Tumor Suppressor
- Humans
- Immunoenzyme Techniques
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice
- Mice, Nude
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Serpins/genetics
- Serpins/metabolism
- Thyroid Carcinoma, Anaplastic/genetics
- Thyroid Carcinoma, Anaplastic/metabolism
- Thyroid Carcinoma, Anaplastic/pathology
- Thyroid Gland/metabolism
- Thyroid Gland/pathology
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Gennaro Chiappetta
- Department of Experimental Oncology, Functional Genomic Unit, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Teresa Valentino
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Counsil (CNR), 80131 Naples, Italy
| | - Michela Vitiello
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Counsil (CNR), 80131 Naples, Italy
| | - Rosa Pasquinelli
- Department of Experimental Oncology, Functional Genomic Unit, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Mario Monaco
- Department of Experimental Oncology, Functional Genomic Unit, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Giuseppe Palma
- Animal Facility, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Romina Sepe
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Counsil (CNR), 80131 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy
| | - Antonio Luciano
- Animal Facility, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Pierlorenzo Pallante
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Counsil (CNR), 80131 Naples, Italy
| | - Dario Palmieri
- Departments of Molecular Virology, Immunology and Human Genetics, Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Concetta Aiello
- Department of Experimental Oncology, Functional Genomic Unit, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Domenica Rea
- Animal Facility, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Simona Nunzia Losito
- Department of Experimental Oncology, Functional Genomic Unit, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Claudio Arra
- Animal Facility, National Cancer Institute “Fondazione Giovanni Pascale”, IRCCS, 80131 Naples, Italy
| | - Alfredo Fusco
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Counsil (CNR), 80131 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Counsil (CNR), 80131 Naples, Italy
| |
Collapse
|
38
|
Li C, Zhao Y, Yang D, Yu Y, Guo H, Zhao Z, Zhang B, Yin X. Inhibitory effects of kaempferol on the invasion of human breast carcinoma cells by downregulating the expression and activity of matrix metalloproteinase-9. Biochem Cell Biol 2015; 93:16-27. [DOI: 10.1139/bcb-2014-0067] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Matrix metalloproteinases (MMPs) have been regarded as major critical molecules assisting tumor cells during metastasis, for excessive ECM (ECM) degradation, and cancer cell invasion. In the present study, in vitro and in vivo assays were employed to examine the inhibitory effects of kaempferol, a natural polyphenol of flavonoid family, on tumor metastasis. Data showed that kaempferol could inhibit adhesion, migration, and invasion of MDA-MB-231 human breast carcinoma cells. Moreover, kaempferol led to the reduced activity and expression of MMP-2 and MMP-9, which were detected by gelatin zymography, real-time PCR, and western blot analysis, respectively. Further elucidation of the mechanism revealed that kaempferol treatment inhibited the activation of transcription factor activator protein-1 (AP-1) and MAPK signaling pathway. Moreover, kaempferol repressed phorbol-12-myristate-13-acetate (PMA)-induced MMP-9 expression and activity through suppressing the translocation of protein kinase Cδ (PKCδ) and MAPK signaling pathway. Our results also indicated that kaempferol could block the lung metastasis of B16F10 murine melanoma cells as well as the expression of MMP-9 in vivo. Taken together, these results demonstrated that kaempferol could inhibit cancer cell invasion through blocking the PKCδ/MAPK/AP-1 cascade and subsequent MMP-9 expression and its activity. Therefore, kaempferol might act as a therapeutic potential candidate for cancer metastasis.
Collapse
Affiliation(s)
- Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Tongshan Road 209#, 221004, Xuzhou, People’s Republic of China
| | - Yuanwei Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Tongshan Road 209#, 221004, Xuzhou, People’s Republic of China
| | - Dan Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Tongshan Road 209#, 221004, Xuzhou, People’s Republic of China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Tongshan Road 209#, 221004, Xuzhou, People’s Republic of China
| | - Hao Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Tongshan Road 209#, 221004, Xuzhou, People’s Republic of China
| | - Ziming Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Tongshan Road 209#, 221004, Xuzhou, People’s Republic of China
| | - Bei Zhang
- Department of Gynecology, Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, People’s Republic of China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical College, Tongshan Road 209#, 221004, Xuzhou, People’s Republic of China
| |
Collapse
|
39
|
Lamberti G, Prabhakarpandian B, Garson C, Smith A, Pant K, Wang B, Kiani MF. Bioinspired microfluidic assay for in vitro modeling of leukocyte-endothelium interactions. Anal Chem 2014; 86:8344-51. [PMID: 25135319 PMCID: PMC4139165 DOI: 10.1021/ac5018716] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/28/2014] [Indexed: 01/08/2023]
Abstract
Current in vitro models of the leukocyte adhesion cascade cannot be used for real-time studies of the entire leukocyte adhesion cascade, including rolling, adhesion, and migration in a single assay. In this study, we have developed and validated a novel bioinspired microfluidic assay (bMFA) and used it to test the hypothesis that blocking of specific steps in the adhesion/migration cascade significantly affects other steps of the cascade. The bMFA consists of an endothelialized microvascular network in communication with a tissue compartment via a 3 μm porous barrier. Human neutrophils in bMFA preferentially adhered to activated human endothelial cells near bifurcations with rolling and adhesion patterns in close agreement with in vivo observations. Treating endothelial cells with monoclonal antibodies to E-selectin or ICAM-1 or treating neutrophils with wortmannin reduced rolling, adhesion, and migration of neutrophils to 60%, 20%, and 18% of their respective control values. Antibody blocking of specific steps in the adhesion/migration cascade (e.g., mAb to E-selectin) significantly downregulated other steps of the cascade (e.g., migration). This novel in vitro assay provides a realistic human cell based model for basic science studies, identification of new treatment targets, selection of pathways to target validation, and rapid screening of candidate agents.
Collapse
Affiliation(s)
- Giuseppina Lamberti
- Department
of Mechanical Engineering, Temple University, 1947 N. 12th street, Philadelphia, Pennsylvania 19122, United States
| | | | - Charles Garson
- Biomedical
Technology, CFD Research Corporation, 701 McMillian Way, Huntsville, Alabama 35806, United
States
| | - Ashley Smith
- Biomedical
Technology, CFD Research Corporation, 701 McMillian Way, Huntsville, Alabama 35806, United
States
| | - Kapil Pant
- Biomedical
Technology, CFD Research Corporation, 701 McMillian Way, Huntsville, Alabama 35806, United
States
| | - Bin Wang
- Department
of Mechanical Engineering, Temple University, 1947 N. 12th street, Philadelphia, Pennsylvania 19122, United States
- Department
of Biomedical Engineering, Widener University, One University Place, Chester, Pennsylvania 19013-5792, United States
| | - Mohammad F. Kiani
- Department
of Mechanical Engineering, Temple University, 1947 N. 12th street, Philadelphia, Pennsylvania 19122, United States
- Department
of Radiation Oncology, Temple University
School of Medicine, 3500
N. Broad Street, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
40
|
Justus CR, Leffler N, Ruiz-Echevarria M, Yang LV. In vitro cell migration and invasion assays. J Vis Exp 2014. [PMID: 24962652 DOI: 10.3791/51046] [Citation(s) in RCA: 328] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Migration is a key property of live cells and critical for normal development, immune response, and disease processes such as cancer metastasis and inflammation. Methods to examine cell migration are very useful and important for a wide range of biomedical research such as cancer biology, immunology, vascular biology, cell biology and developmental biology. Here we use tumor cell migration and invasion as an example and describe two related assays to illustrate the commonly used, easily accessible methods to measure these processes. The first method is the cell culture wound closure assay in which a scratch is generated on a confluent cell monolayer. The speed of wound closure and cell migration can be quantified by taking snapshot pictures with a regular inverted microscope at several time intervals. More detailed cell migratory behavior can be documented using the time-lapse microscopy system. The second method described in this paper is the transwell cell migration and invasion assay that measures the capacity of cell motility and invasiveness toward a chemo-attractant gradient. It is our goal to describe these methods in a highly accessible manner so that the procedures can be successfully performed in research laboratories even just with basic cell biology setup.
Collapse
Affiliation(s)
- Calvin R Justus
- Division of Hematology/Oncology, Department of Oncology, East Carolina University
| | - Nancy Leffler
- Division of Hematology/Oncology, Department of Oncology, East Carolina University
| | | | - Li V Yang
- Division of Hematology/Oncology, Department of Oncology, East Carolina University;
| |
Collapse
|
41
|
Mei P, Bai J, Shi M, Liu Q, Li Z, Fan Y, Zheng J. BRMS1 suppresses glioma progression by regulating invasion, migration and adhesion of glioma cells. PLoS One 2014; 9:e98544. [PMID: 24879377 PMCID: PMC4039505 DOI: 10.1371/journal.pone.0098544] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 05/05/2014] [Indexed: 11/21/2022] Open
Abstract
Breast cancer metastasis suppressor 1 (BRMS1) is a metastasis suppressor gene in several solid tumors. However, the expression and function of BRMS1 in glioma have not been reported. In this study, we investigated whether BRMS1 play a role in glioma pathogenesis. Using the tissue microarray technology, we found that BRMS1 expression is significantly decreased in glioma compared with tumor adjacent normal brain tissue (P<0.01, χ2 test) and reduced BRMS1 staining is associated with WHO stages (P<0.05, χ2 test). We also found that BRMS1 was significantly downregulated in glioma cell lines compared to normal human astrocytes (P<0.01, χ2 test). Furthermore, we demonstrated that BRMS1 overexpression inhibited glioma cell invasion by suppressing uPA, NF-κB, MMP-2 expression and MMP-2 enzyme activity. Moreover, our data showed that overexpression of BRMS1 inhibited glioma cell migration and adhesion capacity compared with the control group through the Src-FAK pathway. Taken together, this study suggested that BRMS1 has a role in glioma development and progression by regulating invasion, migration and adhesion activities of cancer cells.
Collapse
Affiliation(s)
- Pengjin Mei
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jin Bai
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Meilin Shi
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Qinghua Liu
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zhonglin Li
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yuechao Fan
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
- * E-mail: (JZ); (YF)
| | - Junnian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
- Department of Medical Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
- * E-mail: (JZ); (YF)
| |
Collapse
|
42
|
O’Sullivan D, Henry M, Joyce H, Walsh N, Auley EM, Dowling P, Swan N, Moriarty M, Barnham P, Clynes M, Larkin A. 7B7: a novel antibody directed against the Ku70/Ku80 heterodimer blocks invasion in pancreatic and lung cancer cells. Tumour Biol 2014; 35:6983-97. [DOI: 10.1007/s13277-014-1857-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/17/2014] [Indexed: 02/06/2023] Open
|
43
|
Li C, Li F, Zhao K, Yao J, Cheng Y, Zhao L, Li Z, Lu N, Guo Q. LFG-500 inhibits the invasion of cancer cells via down-regulation of PI3K/AKT/NF-κB signaling pathway. PLoS One 2014; 9:e91332. [PMID: 24618693 PMCID: PMC3950212 DOI: 10.1371/journal.pone.0091332] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 02/09/2014] [Indexed: 01/08/2023] Open
Abstract
Cancer cell invasion, one of the crucial events in local growth and metastatic spread of tumors, possess a broad spectrum of mechanisms, especially altered expression of matrix metalloproteinases. LFG-500 is a novel synthesized flavonoid with strong anti-cancer activity, whose exact molecular mechanism remains incompletely understood. This current study was designed to examine the effects of LFG-500 on tumor metastasis using in vitro and in vivo assays. LFG-500 could inhibit adhesion, migration and invasion of MDA-MB-231 human breast carcinoma cells. Meanwhile, it reduced the activities and expression of MMP-2 and MMP-9 via suppressing the transcriptional activation of NF-κB rather than AP-1 or STAT3. Moreover, LFG-500 repressed TNF-α induced cell invasion through inhibiting NF-κB and subsequent MMP-9 activity. Further elucidation of the mechanism revealed that PI3K/AKT but not MAPK signaling pathway was involved in the inhibitory effect of LFG-500 on NF-κB activation. LFG-500 could also suppress lung metastasis of B16F10 murine melanoma cells in vivo. Taken together, these results demonstrated that LFG-500 could block cancer cell invasion via down-regulation of PI3K/AKT/NF-κB signaling pathway, which provides new evidence for the anti-cancer activity of LFG-500.
Collapse
Affiliation(s)
- Chenglin Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fanni Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Kai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jing Yao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yao Cheng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Li Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
- * E-mail: (QG); (NL)
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, People's Republic of China
- * E-mail: (QG); (NL)
| |
Collapse
|
44
|
Characterization of WZ4003 and HTH-01-015 as selective inhibitors of the LKB1-tumour-suppressor-activated NUAK kinases. Biochem J 2014; 457:215-25. [PMID: 24171924 PMCID: PMC3969223 DOI: 10.1042/bj20131152] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The related NUAK1 and NUAK2 are members of the AMPK (AMP-activated protein kinase) family of protein kinases that are activated by the LKB1 (liver kinase B1) tumour suppressor kinase. Recent work suggests they play important roles in regulating key biological processes including Myc-driven tumorigenesis, senescence, cell adhesion and neuronal polarity. In the present paper we describe the first highly specific protein kinase inhibitors of NUAK kinases namely WZ4003 and HTH-01-015. WZ4003 inhibits both NUAK isoforms (IC50 for NUAK1 is 20 nM and for NUAK2 is 100 nM), whereas HTH-01-015 inhibits only NUAK1 (IC50 is 100 nM). These compounds display extreme selectivity and do not significantly inhibit the activity of 139 other kinases that were tested including ten AMPK family members. In all cell lines tested, WZ4003 and HTH-01-015 inhibit the phosphorylation of the only well-characterized substrate, MYPT1 (myosin phosphate-targeting subunit 1) that is phosphorylated by NUAK1 at Ser(445). We also identify a mutation (A195T) that does not affect basal NUAK1 activity, but renders it ~50-fold resistant to both WZ4003 and HTH-01-015. Consistent with NUAK1 mediating the phosphorylation of MYPT1 we find that in cells overexpressing drug-resistant NUAK1[A195T], but not wild-type NUAK1, phosphorylation of MYPT1 at Ser(445) is no longer suppressed by WZ4003 or HTH-01-015. We also demonstrate that administration of WZ4003 and HTH-01-015 to MEFs (mouse embryonic fibroblasts) significantly inhibits migration in a wound-healing assay to a similar extent as NUAK1-knockout. WZ4003 and HTH-01-015 also inhibit proliferation of MEFs to the same extent as NUAK1 knockout and U2OS cells to the same extent as NUAK1 shRNA knockdown. We find that WZ4003 and HTH-01-015 impaired the invasive potential of U2OS cells in a 3D cell invasion assay to the same extent as NUAK1 knockdown. The results of the present study indicate that WZ4003 and HTH-01-015 will serve as useful chemical probes to delineate the biological roles of the NUAK kinases.
Collapse
|
45
|
Toda M, Kawamoto T, Ueha T, Kishimoto K, Hara H, Fukase N, Onishi Y, Harada R, Minoda M, Kurosaka M, Akisue T. 'Decoy' and 'non-decoy' functions of DcR3 promote malignant potential in human malignant fibrous histiocytoma cells. Int J Oncol 2013; 43:703-12. [PMID: 23817777 PMCID: PMC3787885 DOI: 10.3892/ijo.2013.1999] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/07/2013] [Indexed: 12/12/2022] Open
Abstract
Decoy receptor 3 (DcR3) is a soluble secreted protein that belongs to the tumor necrosis factor receptor (TNFR) superfamily. DcR3 inhibits the Fas ligand (FasL)/Fas apoptotic pathway by binding to FasL, competitively with Fas receptor. Previous studies have reported that overexpression of DcR3 has been detected in various human malignancies and that DcR3 functions as a ‘decoy’ for FasL to inhibit FasL-induced apoptosis. In addition, recent studies have revealed that DcR3 has ‘non-decoy’ functions to promote tumor cell migration and invasion, suggesting that DcR3 may play important roles in tumor progression by decoy and non-decoy functions. We have previously reported that overexpression of DcR3 was observed in human malignant fibrous histiocytoma (MFH), however, the roles of DcR3 in MFH have not been studied. In the present study, to elucidate the roles of DcR3 in tumor progression of MFH, we examined the effects of DcR3 inhibition on cell apoptosis, migration and invasion in human MFH cells. siRNA knockdown of DcR3 enhanced the FasL-induced apoptotic activity and significantly decreased cell migration and invasion with a decrease in the activation of phosphatidylinositol 3 kinase (PI3K)/Akt and matrix metalloproteinase (MMP)-2. The findings in this study strongly suggest that DcR3 plays important roles in tumor progression of human MFH by decoy as well as non-decoy functions and that DcR3 may serve as a potent therapeutic target for human MFH.
Collapse
Affiliation(s)
- Mitsunori Toda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tight junction proteins claudin-3 and claudin-4 control tumor growth and metastases. Neoplasia 2013; 14:974-85. [PMID: 23097631 DOI: 10.1593/neo.12942] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/05/2012] [Accepted: 09/07/2012] [Indexed: 12/30/2022] Open
Abstract
The extent of tight junction (TJ) formation is one of many factors that regulate motility, invasion, and metastasis. Claudins are required for the formation and maintenance of TJs. Claudin-3 (CLDN3) and claudin-4 (CLDN4) are highly expressed in the majority of ovarian cancers. We report here that CLDN3 and CLDN4 each serve to constrain the growth of human 2008 cancer xenografts and limit metastatic potential. Knockdown of CLDN3 increased in vivo growth rate by 2.3-fold and knockdown of CLDN4 by 3.7-fold in the absence of significant change in in vitro growth rate. Both types of tumors exhibited increase in birth rate as measured by Ki67 staining and decrease in death rate as reflected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Knockdown of either claudin did not alter expression of other TJ protein but did reduce TJ formation as measured by transepithelial resistance and paracellular flux of dextran, enhance migration and invasion in in vitro assays, and increase lung colonization following intravenous injection. Knockdown of CLDN3 and CLDN4 increased total lung metastatic burden by 1.7-fold and 2.4-fold, respectively. Loss of either CLDN3 or CLDN4 resulted in down-regulation of E-cadherin mRNA and protein, increased inhibitory phosphorylation of glycogen synthase kinase-3β (GSK-3β), and activation of β-catenin pathway signaling as evidenced by increases in nuclear β-catenin, the dephosphorylated form of the protein, and transcriptional activity of β-catenin/T-cell factor (TCF). We conclude that both CLDN3 and CLDN4 mediate interactions with other cells in vivo that restrain growth and metastatic potential by sustaining expression of E-cadherin and limiting β-catenin signaling.
Collapse
|
47
|
T’Joen V, Somers P, Declercq H, Cornelissen M. Evaluation of the behavior of murine and human embryonic stem cells in in vitro migration and invasion assays. Tissue Cell 2013; 45:115-25. [DOI: 10.1016/j.tice.2012.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 09/19/2012] [Accepted: 09/23/2012] [Indexed: 11/28/2022]
|
48
|
Armacki M, Joodi G, Nimmagadda SC, de Kimpe L, Pusapati GV, Vandoninck S, Van Lint J, Illing A, Seufferlein T. A novel splice variant of calcium and integrin-binding protein 1 mediates protein kinase D2-stimulated tumour growth by regulating angiogenesis. Oncogene 2013; 33:1167-80. [DOI: 10.1038/onc.2013.43] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 01/08/2013] [Accepted: 01/11/2013] [Indexed: 12/25/2022]
|
49
|
Rao KVK, Samikkannu T, Dakshayani KB, Zhang X, Sathaye SS, Indap MA, Nair MPN. Chemopreventive potential of an ethyl acetate fraction from Curcuma longa is associated with upregulation of p57(kip2) and Rad9 in the PC-3M prostate cancer cell line. Asian Pac J Cancer Prev 2012; 13:1031-8. [PMID: 22631633 DOI: 10.7314/apjcp.2012.13.3.1031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Turmeric (Curcuma longa) has been shown to possess anti-inflammatory, antioxidant and antitumor properties. However, despite the progress in research with C. longa, there is still a big lacuna in the information on the active principles and their molecular targets. More particularly very little is known about the role of cell cycle genes p57(kip2) and Rad9 during chemoprevention by turmeric and its derivatives especially in prostate cancer cell lines. METHODS Accordingly, in this study, we have examined the antitumor effect of several extracts of C. longa rhizomes by successive fractionation in clonogenic assays using highly metastatic PC-3M prostate cancer cell line. RESULTS A mixture of isopropyl alcohol: acetone: water: chloroform: and methanol extract of C. longa showed significant bioactivity. Further partition of this extract showed that bioactivity resides in the dichloromethane soluble fraction. Column chromatography of this fraction showed presence of biological activity only in ethyl acetate eluted fraction. HPLC, UV-Vis and Mass spectra studies showed presence three curcuminoids in this fraction besides few unidentified components. CONCLUSIONS From these observations it was concluded that the ethyl acetate fraction showed not only inhibition of colony forming ability of PC-3M cells but also up-regulated cell cycle genes p57(kip2) and Rad9 and further reduced the migration and invasive ability of prostate cancer cells.
Collapse
Affiliation(s)
- K V K Rao
- Department of Immunology, College of Medicine, Florida International University, Miami, Florida, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Bhuvaneswari R, Yuen GY, Chee SK, Olivo M. Antiangiogenesis agents avastin and erbitux enhance the efficacy of photodynamic therapy in a murine bladder tumor model. Lasers Surg Med 2012; 43:651-62. [PMID: 22057493 DOI: 10.1002/lsm.21109] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND OBJECTIVE Photodynamic therapy (PDT) has been established as an alternative therapy for the treatment of various types of malignant disorders, including oesophageal, lung, and bladder cancer. However, one of the limitations of PDT is treatment-induced hypoxia that triggers angiogenesis. The objective of this study was to evaluate the effects of combination therapy with PDT and an antiangiogenic protocol using monoclonal antibodies against both vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR). MATERIALS AND METHODS In vitro angiogenesis assays and in vivo matrigel assay were performed to understand the inhibitory effects of the antiangiogenic agents. Tumor bearing mice were assigned to six different categories: Control, PDT only, Avastin + Erbitux, PDT + Avastin, PDT + Erbitux, and PDT + Avastin and Erbitux. Treated and control tumors were monitored for recurrence for up to 90 days. RESULTS In vitro results provided valuable insight into the dynamics of endothelial cells in response to angiogenic stimulants and inhibitors to assess the angiogenesis processes. Addition of VEGF increased the migration of bladder cancer cells and addition of Avastin and Erbitux decreased cell migration significantly. Both inhibitors were also able to suppress invasion and tube formation in human umbilical vein endothelial cells (HUVEC). The in vivo tumor response for PDT with single inhibitor (Avastin or Erbitux) and double inhibitor (Avastin + Erbitux) was comparable; however, targeting both VEGF and EGFR pathways along with PDT resulted in more rapid response. Downregulation of VEGF and EGFR were observed in tumors treated with PDT in combination with Avastin and Erbitux respectively. CONCLUSION Our results show that blocking the VEGF or EGFR pathway along with PDT can effectively suppress tumor growth and the combination of both VEGF and EGFR inhibitors along with PDT could be used to treat more aggressive tumors to achieve rapid response.
Collapse
|