1
|
Yang X, Zhang C, Yan C, Ma L, Ma J, Meng X. System analysis based on the ER stress-related genes identifies WFS1 as a novel therapy target for colon cancer. Aging (Albany NY) 2022; 14:9243-9263. [PMID: 36445321 PMCID: PMC9740360 DOI: 10.18632/aging.204404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Colon cancer (COAD) is the third-largest common malignant tumor and the fourth major cause of cancer death in the world. Endoplasmic reticulum (ER) stress has a great influence on cell growth, migration, proliferation, invasion, angiogenesis, and chemoresistance of massive tumors. Although ER stress is known to play an important role in various types of cancer, the prognostic model based on ER stress-related genes (ERSRGs) in colon cancer has not been constructed yet. In this study, we established an ERSRGs prognostic risk model to assess the survival of COAD patients. METHODS The COAD gene expression profile and clinical information data of the training set were obtained from the GEO database (GSE40967) and the test set COAD gene expression profile and clinical informative data were downloaded from the TCGA database. The endoplasmic reticulum stress-related genes (ERSRGs) were obtained from Gene Set Enrichment Analysis (GSEA) website. Differentially expressed ERSRGs between normal samples and COAD samples were identified by R "limma" package. Based on the univariate, lasso, and multivariate Cox regression analysis, we developed an ERSRGs prognostic risk model to predict survival in COAD patients. Finally, we verified the function of WFS1 in COAD through in vitro experiments. RESULTS We built a 9-gene prognostic risk model based on the univariate, lasso, and multivariate Cox regression analysis. Kaplan-Meier survival analysis and Receiver operating characteristic (ROC) curve revealed that the prognostic risk model has good predictive performance. Subsequently, we screened 60 compounds with significant differences in the estimated half-maximal inhibitory concentration (IC50) between high-risk and low-risk groups. In addition, we found that the ERSRGs prognostic risk model was related to immune cell infiltration and the expression of immune checkpoint molecules. Finally, we determined that knockdown of the expression of WFS1 inhibits the proliferation of colon cancer cells. CONCLUSIONS The prognostic risk model we built may help clinicians accurately predict the survival of patients with COAD. Our findings provide valuable insights into the role of ERSRGs in COAD and may provide new targets for COAD therapy.
Collapse
Affiliation(s)
- Xianguang Yang
- School of Life Sciences, State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| | - Chaoyang Zhang
- School of Life Sciences, State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, China
| | - Cheng Yan
- School of Pharmacy, Key Laboratory of Nano-carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, Henan 453000, China
| | - Liukai Ma
- School of Pharmacy, Key Laboratory of Nano-carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, Henan 453000, China
| | - Jiahao Ma
- School of Pharmacy, Key Laboratory of Nano-carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, Henan 453000, China
| | - Xiaoke Meng
- School of Pharmacy, Key Laboratory of Nano-carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, Henan 453000, China
| |
Collapse
|
2
|
Takeda T, Tsubaki M, Kato N, Genno S, Ichimura E, Enomoto A, Imano M, Satou T, Nishida S. Sorafenib treatment of metastatic melanoma with c-Kit aberration reduces tumor growth and promotes survival. Oncol Lett 2021; 22:827. [PMID: 34691254 DOI: 10.3892/ol.2021.13089] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
Melanomas are highly malignant tumors that readily metastasize and have poor prognosis. Targeted therapy is a cornerstone of treatment for patients with melanoma. Although c-Kit gene aberration has found in 5-10% of melanoma cases, research on c-Kit inhibitors for melanoma with c-Kit aberration have been disappointing. Sorafenib is a tyrosine kinase inhibitor, whose targets include c-Kit, platelet derived growth factor receptor (PDGFR), VEGFR and RAF. The present study aimed to examine the effect of sorafenib on metastatic melanoma with c-Kit aberration. Cell viability was assessed via trypan blue assay. Migration and invasion were analyzed using cell culture inserts. The anti-metastatic effects and antitumour activity of sorafenib were determined in an in vivo model. Protein expression was detected via western blotting, and the expression of MMP and very late antigen (VLA) was detected via reverse transcription-quantitative PCR. It was identified that sorafenib decreased cell viability, migration and invasion in vitro. Furthermore, sorafenib inhibited metastasis and tumor growth in vivo. Mechanistically, sorafenib inhibited c-Kit, PDGFR, VEGFR, B-Raf and c-Raf phosphorylation both in vitro and in vivo. In addition, sorafenib reduced the expression levels of MMPs and VLA. Importantly, there was a significant effect of sorafenib treatment on overall survival in mice. Collectively, this study suggests that sorafenib may serve as a novel therapeutic option for melanoma with c-Kit dysregulation.
Collapse
Affiliation(s)
- Tomoya Takeda
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Masanobu Tsubaki
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Natsuki Kato
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Shuji Genno
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Eri Ichimura
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Aya Enomoto
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University School of Medicine, Osakasayama, Osaka 589-0014, Japan
| | - Takao Satou
- Department of Pathology, Kindai University School of Medicine, Osakasayama, Osaka 589-0014, Japan
| | - Shozo Nishida
- Department of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| |
Collapse
|
3
|
Gardouh AR, Ewedah TM, Abd-Allah FI, Ghorab MM, Omran MM, El-Sawy HS. Enhanced efficacy, cellular uptake, and antiangiogenic activity of the optimized imatinib mesylate-loaded proniosomal-derived nanovesicles. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
4
|
Imatinib exhibit synergistic pleiotropy in the prevention of colorectal cancer by suppressing proinflammatory, cell survival and angiogenic signaling. Cell Signal 2020; 76:109803. [PMID: 33022360 DOI: 10.1016/j.cellsig.2020.109803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022]
Abstract
Recent global incidences and mortality rates have placed colorectal cancer (CRC) at third and second positions, respectively, among both sexes of all ages. Resistance during chemotherapy is a big problem in the treatment and disease-free survival of CRC patients. Discovery of new anticancer drug(s) is a time taking process and therefore, invites studies for repurposing the known therapeutics. The present study was conceived to analyze the anticancer role of Imatinib in experimental CRC at early stages. Different experimental procedures e.g. tumor incidences or histoarchitectural changes, gene and protein expression analysis, estimations of intracellular calcium, ROS, mitochondrial membrane potential, apoptotic index and molecular docking was performed to support the hypothesis. It was observed that Imatinib could function as an immunomodulator by breaking the feed-back loop between the proinflammatory cytokines (IL-1β and TNF-α) and transcription factors (NF-κB, Jak3/Stat3) knowingly involved in increased cell proliferation during tumorigenesis via activating different intracellular signaling. Also, Imatinib could independently deregulate the other cell survival and proliferation signaling e.g. PI3-K/Akt/mTOR, Wnt/β-catenin and MAPK. Proinflammatory cytokines orchestrated intracellular signaling also involve angiogenic factors to be upregulated during CRC which were also seemed to be independently suppressed by Imatinib. Restoration of physiological apoptosis by increasing the release of intracellular calcium to generate ROS thereby reducing the mitochondrial membrane potential for the release of cytochrome c and activation of caspase-3 was also reported with Imatinib administration. Thus, it may be suggested that Imatinib show synergistic pleiotropy in suppressing the interlinked tumorigenic signaling pathways independently.
Collapse
|
5
|
Lafitte M, Sirvent A, Roche S. Collagen Kinase Receptors as Potential Therapeutic Targets in Metastatic Colon Cancer. Front Oncol 2020; 10:125. [PMID: 32117772 PMCID: PMC7028753 DOI: 10.3389/fonc.2020.00125] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of tumor-related death worldwide. While surgery can cure patients with early stage CRC, the 5-year survival rate is only 10% for patients with metastatic disease. Therefore, new anti-metastatic therapies are needed for this cancer. Metastatic spread defines the dissemination of cancer cells with tumor-initiating capacities from the primary tumor and their colonization of distinct organs, mainly the liver, for secondary tumor formation. Although the underlying mechanisms are not fully understood, components of the tumor microenvironment have gained strong interest. Among the known metastatic-promoting factors, collagens are extracellular matrix components that are deposited within the tumor, the tumor microenvironment, and at metastatic site(s), and are recognized to play essential roles during metastasis development. Here, we review recent findings on the metastatic role of the collagen receptors Discoidin Domain Receptors 1 and 2 (DDR1 and DDR2) in CRC and discuss the therapeutic value of targeting these receptor tyrosine kinases in this cancer.
Collapse
Affiliation(s)
| | | | - Serge Roche
- CRBM, CNRS, Univ. Montpellier, Montpellier, France
| |
Collapse
|
6
|
Imatinib modulates pro-inflammatory microenvironment with angiostatic effects in experimental lung carcinogenesis. Inflammopharmacology 2019; 28:231-252. [PMID: 31676982 DOI: 10.1007/s10787-019-00656-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 10/10/2019] [Indexed: 10/25/2022]
Abstract
Lung cancer has second highest rate of incidence and mortality around the world. Smoking cigarettes is the main stream cause of lung carcinogenesis along with other factors such as spontaneous mutations, inactivation of tumor suppressor genes. The present study was aimed to identify the mechanistic role of Imatinib in the chemoprevention of experimental lung carcinogenesis in rat model. Gross morphological observations for tumor formation, histological examinations, RT-PCR, Western blotting, fluorescence spectroscopy and molecular docking studies were performed to elucidate the chemopreventive effects of Imatinib and support our hypothesis by various experiments. It is evident that immuno-compromised microenvironment inside solid tumors is responsible for tumor progression and drug resistance. Therefore, it is inevitable to modulate the pro-inflammatory signaling inside solid tumors to restrict neoangiogenesis. In the present study, we observed that Imatinib could downregulate the inflammatory signaling and also attributed angiostatic effects. Moreover, Imatinib also altered the biophysical properties of BAL cells such as plasma membrane potential, fluidity and microviscosity to restrict their infiltration and thereby accumulation to mount immuno-compromised environment inside the solid tumors during angiogenesis. Our molecular docking studies suggest that immunomodulatory and angiostatic properties of Imatinib could be either independent of each other or just a case of synergistic pleiotropy. Imatinib was observed to activate the intrinsic or mitochondrial pathway of apoptosis to achieve desired effects in cancer cell killings. Interestingly, binding of Imatinib inside the catalytic domain of PARP-1 also suggests that it has caspase-independent properties in promoting cancer cell deaths.
Collapse
|
7
|
Kotlarz A, Przybyszewska M, Swoboda P, Neska J, Miłoszewska J, Grygorowicz MA, Kutner A, Markowicz S. Imatinib inhibits the regrowth of human colon cancer cells after treatment with 5-FU and cooperates with vitamin D analogue PRI-2191 in the downregulation of expression of stemness-related genes in 5-FU refractory cells. J Steroid Biochem Mol Biol 2019; 189:48-62. [PMID: 30772447 DOI: 10.1016/j.jsbmb.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/31/2018] [Accepted: 02/12/2019] [Indexed: 12/14/2022]
Abstract
Conventional cytotoxic drugs preferentially eliminate differentiated cancer cells but spare relatively more resistant stem-like cancer cells capable to initiate recurrence. Due to cancer cell plasticity, the stem-like phenotype can be also acquired by cancer cells refractory to treatment with cytotoxic drugs. We investigated whether drugs inhibiting receptor tyrosine kinases could be used to target human colon cancer cells initiating cancer regrowth following conventional cytotoxic treatment. The moderately differentiated cell line HT-29 and poorly differentiated cell line HCT-116 were exposed to 5-fluorouracil (5-FU). Cells that resisted the exposure to 5-FU were subsequently treated with imatinib or sunitinib. Both drugs reduced clonogenicity of 5-FU-refractory cells under normoxic and hypoxic culture conditions. The expression of numerous stemness-related genes was upregulated in cancer cells following the exposure to 5-FU, and remained at a high level in 5-FU-refractory cells undergoing renewal under normoxia, but decreased spontaneously under hypoxia. Imatinib downregulated the expression of stemness-related genes in cells undergoing renewal under normoxia. A combination of imatinib with PRI-2191, an analogue of 1,25-dihydroxyvitamin D3, downregulated stemness-related genes in HCT-116/5-FU cells more efficiently than imatinib alone. A synthetic analogue of 1,25-dihydroxyvitamin D2 (PRI-1906) abolished the effect of imatinib on gene expression in HCT-116/5-FU cells undergoing renewal under normoxia. Sunitinib promoted shift of phenotype of HT-29/5-FU cells undergoing renewal toward stem-like one. It suggests that the phenotype shift toward stemness induced by sequential sunitinib treatment following 5-FU treatment could increase a risk of cancer recurrence. In contrast to sunitinib, imatinib could be used both to interfere with cancer regrowth after conventional chemotherapy and to downregulate the expression of stemness-related genes in residual colon cancer cells capable to initiate cancer recurrence. The findings suggest that imatinib could also be combined with vitamin D analogue PRI-2191 to prevent recurrence more efficiently than imatinib alone and to compensate for vitamin D deficiency resulting from imatinib treatment.
Collapse
Affiliation(s)
- Agnieszka Kotlarz
- Department of Immunology, Maria Sklodowska-Curie Institute - Oncology Center, 5 WK Roentgen Str., 02-781 Warszawa, Poland.
| | - Małgorzata Przybyszewska
- Department of Immunology, Maria Sklodowska-Curie Institute - Oncology Center, 5 WK Roentgen Str., 02-781 Warszawa, Poland.
| | - Paweł Swoboda
- Department of Immunology, Maria Sklodowska-Curie Institute - Oncology Center, 5 WK Roentgen Str., 02-781 Warszawa, Poland.
| | - Jacek Neska
- Department of Immunology, Maria Sklodowska-Curie Institute - Oncology Center, 5 WK Roentgen Str., 02-781 Warszawa, Poland.
| | - Joanna Miłoszewska
- Department of Immunology, Maria Sklodowska-Curie Institute - Oncology Center, 5 WK Roentgen Str., 02-781 Warszawa, Poland.
| | - Monika Anna Grygorowicz
- Department of Immunology, Maria Sklodowska-Curie Institute - Oncology Center, 5 WK Roentgen Str., 02-781 Warszawa, Poland.
| | - Andrzej Kutner
- Pharmacology Department, Pharmaceutical Research Institute, 8 Rydygiera, 01-793 Warsaw, Poland.
| | - Sergiusz Markowicz
- Department of Immunology, Maria Sklodowska-Curie Institute - Oncology Center, 5 WK Roentgen Str., 02-781 Warszawa, Poland.
| |
Collapse
|
8
|
Design, synthesis and 3D QSAR based pharmacophore study of novel imatinib analogs as antitumor-apoptotic agents. Future Med Chem 2018; 10:1421-1433. [PMID: 29788766 DOI: 10.4155/fmc-2017-0242] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIM Imatinib possesses various mechanisms for combating cancer, making the development of imatinib analogs an attractive target for cancer research. METHOD Two series of analogs were designed and synthesized, maintaining the essential pharmacophoric features in imatinib structure. The synthesized compounds were subjected to cell-based antiproliferative assays against nonsmall lung (A549) and colon cancer cell lines. In addition, flow cytometry cell cycle and caspase-3 colorimetric assays were performed. RESULTS Most compounds showed potent anticancer activity against both cell lines with IC50 = 0.14-5.07 μM. Three compounds demonstrated ability to reinforce cell cycle arrest at G1 stage in a manner similar to imatinib. In addition, they induced apoptosis via activation of caspase-3.
Collapse
|
9
|
Sargin I, Akyuz L, Kaya M, Tan G, Ceter T, Yildirim K, Ertosun S, Aydin GH, Topal M. Controlled release and anti-proliferative effect of imatinib mesylate loaded sporopollenin microcapsules extracted from pollens of Betula pendula. Int J Biol Macromol 2017; 105:749-756. [PMID: 28716746 DOI: 10.1016/j.ijbiomac.2017.07.093] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/14/2017] [Accepted: 07/14/2017] [Indexed: 01/15/2023]
Abstract
Sporopollenin is a promising material for drug encapsulation due to its excellent properties; uniformity in size, non-toxicity, chemically and thermally resilient nature. Herein, morphologically intact sporopollenin microcapsules were extracted from Betula pendula pollens. Cancer therapeutic agent (imatinib mesylate) was loaded into the microcapsules. The encapsulation efficiency by passive loading technique was found to be 21.46%. Release behaviour of the drug from microcapsules was found to be biphasic, with an initial fast release followed by a slower rate of release. Imatinib mesylate release from the drug itself (control) was faster than from imatinib mesylate-loaded sporopollenin microcapsules. The release profiles for both free and entrapped drug samples were significantly slower and more controlled in PBS buffer (pH 7.4) than in HCl (pH 1.2) buffer. Cumulative drug release from IM-MES-loaded sporopollenin microcapsules was found to be 65% within 24h for PBS, whereas release from the control was completed within 1h. Also, a complete dissolution of control in HCl buffer was observed within first 30min. MTT assay revealed that drug-loaded microcapsules were effective on WiDr human colon carcinoma cell line. B. pendula sporopollenin can be suggested as an effective carrier for oral delivery of imatinib mesylate.
Collapse
Affiliation(s)
- Idris Sargin
- Aksaray University, Faculty of Science and Letters, Department of Biotechnology and Molecular Biology, 68100 Aksaray, Turkey.
| | - Lalehan Akyuz
- Aksaray University, Technical Vocational School, Department of Chemistry Technology, 68100, Aksaray, Turkey
| | - Murat Kaya
- Aksaray University, Faculty of Science and Letters, Department of Biotechnology and Molecular Biology, 68100 Aksaray, Turkey
| | - Gamze Tan
- Aksaray University, Faculty of Science and Letters, Department of Biology, 68100 Aksaray, Turkey
| | - Talip Ceter
- Kastamonu University, Faculty of Arts and Sciences, Department of Biology, 37100 Kastamonu, Turkey
| | - Kevser Yildirim
- Aksaray University, Faculty of Science and Letters, Department of Biotechnology and Molecular Biology, 68100 Aksaray, Turkey
| | - Seymanur Ertosun
- Aksaray University, Faculty of Science and Letters, Department of Biotechnology and Molecular Biology, 68100 Aksaray, Turkey
| | - Gozde Hatun Aydin
- Aksaray University, Faculty of Science and Letters, Department of Biotechnology and Molecular Biology, 68100 Aksaray, Turkey
| | - Muge Topal
- Aksaray University, Faculty of Science and Letters, Department of Biotechnology and Molecular Biology, 68100 Aksaray, Turkey
| |
Collapse
|
10
|
Lee J, Park D, Lee Y. Metformin Synergistically Potentiates the Antitumor Effects of Imatinib in Colorectal Cancer Cells. Dev Reprod 2017; 21:139-150. [PMID: 28785735 PMCID: PMC5532306 DOI: 10.12717/dr.2017.21.2.139] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/27/2022]
Abstract
Metformin is the most commonly prescribed anti-diabetic drug with relatively
minor side effect. Substantial evidence has suggested that metformin is
associated with decreased cancer risk and anticancer activity against diverse
cancer cells. The tyrosine kinase inhibitor imatinib has shown powerful activity
for treatment of chronic myeloid leukemia and also induces growth arrest and
apoptosis in colorectal cancer cells. In this study, we tested the combination
of imatinib and metformin against HCT15 colorectal cancer cells for effects on
cell viability, cell cycle and autophagy. Our data show that metformin
synergistically enhances the imatinib cytotoxicity in HCT15 cells as indicated
by combination and drug reduction indices. We also demonstrate that the
combination causes synergistic down-regulation of pERK, cell cycle arrest in S
and G2/M phases via reduction of cyclin B1 level. Moreover, the
combination resulted in autophagy induction as revealed by increased acidic
vesicular organelles and cleaved form of LC3-II. Inhibition of autophagic
process by chloroquine led to decreased cell viability, suggesting that
induction of autophagy seems to play a cell protective role that may act against
anticancer effects. In conclusion, our present data suggest that metformin in
combination with imatinib might be a promising therapeutic option in colorectal
cancer.
Collapse
Affiliation(s)
- Jaeryun Lee
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 690-756, Korea
| | - Deokbae Park
- Dept. of Histology, Jeju National University School of Medicine, Jeju 690-756, Korea
| | - Youngki Lee
- Dept. of Histology, Jeju National University School of Medicine, Jeju 690-756, Korea
| |
Collapse
|
11
|
Abd-El Fattah AA, Darwish HA, Fathy N, Shouman SA. Carbonic anhydrase inhibition boosts the antitumor effects of Imatinib mesylate via potentiating the antiangiogenic and antimetastatic machineries. Toxicol Appl Pharmacol 2017; 316:123-138. [DOI: 10.1016/j.taap.2016.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/14/2016] [Accepted: 12/23/2016] [Indexed: 12/19/2022]
|
12
|
Kassem MA, El-Sawy HS, Abd-Allah FI, Abdelghany TM, El-Say KM. Maximizing the Therapeutic Efficacy of Imatinib Mesylate–Loaded Niosomes on Human Colon Adenocarcinoma Using Box-Behnken Design. J Pharm Sci 2017; 106:111-122. [DOI: 10.1016/j.xphs.2016.07.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/29/2016] [Accepted: 07/12/2016] [Indexed: 01/01/2023]
|
13
|
Atari-Hajipirloo S, Nikanfar S, Heydari A, Kheradmand F. Imatinib and its combination with 2,5-dimethyl-celecoxibinduces apoptosis of human HT-29 colorectal cancer cells. Res Pharm Sci 2017; 12:67-73. [PMID: 28255316 PMCID: PMC5333482 DOI: 10.4103/1735-5362.199049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Mono-targeting by imatinib as a main antitumor agent does not always accomplish complete cancer suppression. 2,5-dimethyl-celecoxib (DMC) is a close structural analog of the selective cyclooxygenase-2 (COX-2) inhibitor, celecoxib, that lacks COX-2 inhibitory function. In this study, we aimed to show the apoptotic effects of imatinib in combination with DMC in human HT-29 colorectal cancer (CRC) cells. HT-29 CRC cells were treated with IC50 dose of imatinib (6.60 μM), DMC (23.45 μM), and their combination (half dose of IC50) for 24 h. The caspase-3 activity was estimated with colorimetric kit. The caspase-3 gene expression was evaluated by real-time PCR method. There was a significant up-regulation in caspase-3 enzyme activity and caspase-3 expression by imatinib and its half dose combination with DMC as compared to control. As a summary, the results of this study strongly suggest that half dose combination of imatinib with DMC induced apoptosis as potent as full dose imatinib in human HT-29 CRC cells, while minimizing undesired side effects related to imatinib mono-therapy. This study also pointed towards possible caspase-dependent actions of imatinib and DMC.
Collapse
Affiliation(s)
- Somayeh Atari-Hajipirloo
- Department of Biochemistry, Student Research Committee, Urmia University of Medical Sciences, Urmia, I.R. Iran
| | - Saba Nikanfar
- Department of Biochemistry, Urmia University of Medical Sciences, Urmia, I.R. Iran
| | - Amir Heydari
- Department of Pharmacology, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, I.R. Iran
| | - Fatemeh Kheradmand
- Department of Clinical Biochemistry, Cellular and Molecular and Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, I.R. Iran
| |
Collapse
|
14
|
Gajski G, Gerić M, Domijan AM, Garaj-Vrhovac V. Combined cyto/genotoxic activity of a selected antineoplastic drug mixture in human circulating blood cells. CHEMOSPHERE 2016; 165:529-538. [PMID: 27681109 DOI: 10.1016/j.chemosphere.2016.09.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/06/2016] [Accepted: 09/14/2016] [Indexed: 06/06/2023]
Abstract
Antineoplastic drugs are highly cytotoxic chemotherapeutic agents that can often interfere directly or indirectly with the cell's genome. In an environmental or medical setting simultaneous exposure may occur. Such multiple exposures may pose a higher risk than it could be assumed from the studies evaluating the effect of a single substance. Therefore, in the present study we tested the combined cyto/genotoxicity of a mixture of selected antineoplastic drugs with different mechanisms of action (5-fluorouracil, etoposide, and imatinib mesylate) towards human lymphocytes in vitro. The results suggest that the selected antineoplastic drug mixture is potentially cyto/genotoxic and that it can induce cell and genome damage even at low concentrations. Moreover, the changes in the measured oxidative stress parameters suggest the participation of reactive oxygen species in the cyto/genotoxicity of the selected mixture. The obtained results indicate not only that such mixtures may pose a risk to cell and genome integrity, but also that single compound toxicity data are not sufficient for the predicting toxicity in a complex environment. Altogether, the results emphasise the need for further toxicological screening of antineoplastic drug mixtures, especially at low environmentally relevant concentrations, as to avoid any possible adverse effects on the environment and human health.
Collapse
Affiliation(s)
- Goran Gajski
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska cesta 2, 10000 Zagreb, Croatia.
| | - Marko Gerić
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska cesta 2, 10000 Zagreb, Croatia.
| | - Ana-Marija Domijan
- University of Zagreb, Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb, Croatia.
| | - Vera Garaj-Vrhovac
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska cesta 2, 10000 Zagreb, Croatia.
| |
Collapse
|
15
|
Mišík M, Filipic M, Nersesyan A, Mišíková K, Knasmueller S, Kundi M. Analyses of combined effects of cytostatic drugs on micronucleus formation in the Tradescantia. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:14762-14770. [PMID: 26620864 DOI: 10.1007/s11356-015-5837-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/18/2015] [Indexed: 06/05/2023]
Abstract
Recent experiments showed that 5-fluorouracil (5FU), cisplatin (CDDP), etoposide (ET), and imatinib mesylate (IM), which are currently among the most widely used anticancer drugs, cause damage of the genetic material in higher plants. The aim of the present study was to determine whether mixtures of these drugs cause synergistic or antagonistic effects which may have an impact on their environmental safety. Therefore, the effects of binary mixtures of these anticancer drugs on the induction of micronuclei (MN) which reflect structural and numerical chromosomal aberrations were assessed in Tradescantia tetrads. Synergistic/antagonistic effects were determined by comparison with single exposures that would be equally effective in a reference model of independent action. This comparison was performed at two distinct effect sizes. We found clear evidence for synergisms in combination experiments with IM and antagonism in a high-dose experiment with ET and 5FU. Our findings indicate that IM increases the genotoxic effects of other anticancer drugs. The maximal effects which we found were in the range between 19 and 38 % in the excess of effect sizes predicted under independent action. These effects may have an impact on the overall genotoxic activities of untreated hospital waste waters but not on the environment in general as the predicted environmental concentrations of the studied drugs are several orders of magnitude lower as the levels which are required to cause induction of MN in higher plants.
Collapse
Affiliation(s)
- Miroslav Mišík
- Institute for Cancer Research, Department of Internal Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Metka Filipic
- Department for Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Armen Nersesyan
- Institute for Cancer Research, Department of Internal Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Katarína Mišíková
- Department of Botany, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Siegfried Knasmueller
- Institute for Cancer Research, Department of Internal Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria.
| | - Michael Kundi
- Medical University of Vienna, Institute of Environmental Health, Medical University of Vienna, Wien, Austria
| |
Collapse
|
16
|
Li L, Duan T, Wang X, Zhang RH, Zhang M, Wang S, Wang F, Wu Y, Huang H, Kang T. KCTD12 Regulates Colorectal Cancer Cell Stemness through the ERK Pathway. Sci Rep 2016; 6:20460. [PMID: 26847701 PMCID: PMC4742820 DOI: 10.1038/srep20460] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/01/2015] [Indexed: 01/05/2023] Open
Abstract
Targeting cancer stem cells (CSCs) in colorectal cancer (CRC) remains a difficult problem, as the regulation of CSCs in CRC is poorly understood. Here we demonstrated that KCTD12, potassium channel tetramerization domain containing 12, is down-regulated in the CSC-like cells of CRC. The silencing of endogenous KCTD12 and the overexpression of ectopic KCTD12 dramatically enhances and represses CRC cell stemness, respectively, as assessed in vitro and in vivo using a colony formation assay, a spheroid formation assay and a xenograft tumor model. Mechanistically, KCTD12 suppresses CRC cell stemness markers, such as CD44, CD133 and CD29, by inhibiting the ERK pathway, as the ERK1/2 inhibitor U0126 abolishes the increase in expression of CRC cell stemness markers induced by the down-regulation of KCTD12. Indeed, a decreased level of KCTD12 is detected in CRC tissues compared with their adjacent normal tissues and is an independent prognostic factor for poor overall and disease free survival in patients with CRC (p = 0.007). Taken together, this report reveals that KCTD12 is a novel regulator of CRC cell stemness and may serve as a novel prognostic marker and therapeutic target for patients with CRC.
Collapse
Affiliation(s)
- Liping Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Tingmei Duan
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Xin Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Ru-Hua Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Meifang Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Suihai Wang
- School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Fen Wang
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
17
|
Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev 2016; 97:4-27. [PMID: 26562801 DOI: 10.1016/j.addr.2015.11.001] [Citation(s) in RCA: 1543] [Impact Index Per Article: 171.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network composed of collagens, proteoglycans/glycosaminoglycans, elastin, fibronectin, laminins, and several other glycoproteins. Matrix components bind each other as well as cell adhesion receptors forming a complex network into which cells reside in all tissues and organs. Cell surface receptors transduce signals into cells from ECM, which regulate diverse cellular functions, such as survival, growth, migration, and differentiation, and are vital for maintaining normal homeostasis. ECM is a highly dynamic structural network that continuously undergoes remodeling mediated by several matrix-degrading enzymes during normal and pathological conditions. Deregulation of ECM composition and structure is associated with the development and progression of several pathologic conditions. This article emphasizes in the complex ECM structure as to provide a better understanding of its dynamic structural and functional multipotency. Where relevant, the implication of the various families of ECM macromolecules in health and disease is also presented.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Chrysostomi Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece; Division of Medical Protein Chemistry, Department of Translational Medicine Malmö, Lund University, S-20502 Malmö, Sweden
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
18
|
Sarin H. Conserved molecular mechanisms underlying the effects of small molecule xenobiotic chemotherapeutics on cells. Mol Clin Oncol 2015; 4:326-368. [PMID: 26998284 DOI: 10.3892/mco.2015.714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/08/2015] [Indexed: 12/14/2022] Open
Abstract
For proper determination of the apoptotic potential of chemoxenobiotics in synergism, it is important to understand the modes, levels and character of interactions of chemoxenobiotics with cells in the context of predicted conserved biophysical properties. Chemoxenobiotic structures are studied with respect to atom distribution over molecular space, the predicted overall octanol-to-water partition coefficient (Log OWPC; unitless) and molecular size viz a viz van der Waals diameter (vdWD). The Log OWPC-to-vdWD (nm-1 ) parameter is determined, and where applicable, hydrophilic interacting moiety/core-to-vdWD (nm-1 ) and lipophilic incorporating hydrophobic moiety/core-to-vdWD (nm-1 ) parameters of their part-structures are determined. The cellular and sub-cellular level interactions of the spectrum of xenobiotic chemotherapies have been characterized, for which a classification system has been developed based on predicted conserved biophysical properties with respect to the mode of chemotherapeutic effect. The findings of this study are applicable towards improving the effectiveness of existing combination chemotherapy regimens and the predictive accuracy of personalized cancer treatment algorithms as well as towards the selection of appropriate novel xenobiotics with the potential to be potent chemotherapeutics for dendrimer nanoparticle-based effective transvascular delivery.
Collapse
Affiliation(s)
- Hemant Sarin
- Freelance Investigator in Translational Science and Medicine, Charleston, WV 25314, USA
| |
Collapse
|
19
|
Abstract
The Abelson (ABL) tyrosine kinases were identified as drivers of leukemia in mice and humans. Emerging data has shown a role for the ABL family kinases, ABL1 and ABL2, in the progression of several solid tumors. This review will focus on recent reports of the involvement of the ABL kinases in tumor progression using mouse models as well as recent data generated from genomic and proteomic studies linking enhanced expression and hyper-activation of the ABL kinases to some human cancers. Preclinical studies on small molecule inhibitors of the ABL kinases suggest that their use may have beneficial effects for the treatment of selected solid tumors.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710 USA
| | - Ann Marie Pendergast
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710 USA
| |
Collapse
|
20
|
Lack of Association between Membrane-Type 1 Matrix Metalloproteinase Expression and Clinically Relevant Molecular or Morphologic Tumor Characteristics at the Leading Edge of Invasive Colorectal Carcinoma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:185404. [PMID: 26106602 PMCID: PMC4461720 DOI: 10.1155/2015/185404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/03/2015] [Accepted: 03/19/2015] [Indexed: 11/17/2022]
Abstract
Colorectal cancer (CRC) is one of the leading causes of death from cancer in the western world, but tumor biology and clinical course show great interindividual variation. Molecular and morphologic tumor characteristics, such as KRAS/BRAF mutation status, mismatch repair (MMR) protein expression, tumor growth pattern, and tumor cell budding, have been shown to be of key therapeutic and/or prognostic relevance in CRC. Membrane-type 1 matrix metalloproteinase (MT1-MMP) is a membrane-anchored zinc-binding endopeptidase that is expressed at the leading edge of various invasive carcinomas and promotes tumor cell invasion through degradation of the extracellular matrix. The aim of this study was to investigate possible associations between MT1-MMP expression and molecular tumor characteristics as well as morphologic features of tumor aggressiveness in a consecutive series of 79 CRC tissue samples. However, although MT1-MMP was expressed in 41/79 samples (52%), there was no significant association between MT1-MMP expression and KRAS/BRAF mutation status, MMR protein expression, presence of lymphovascular invasion, tumor growth pattern, tumor-infiltrating lymphocytes, or tumor cell budding in our sample cohort (P > 0.05). Thus, we conclude that although MT1-MMP may play a role in CRC invasion, it is not of key relevance to the current models of CRC invasion and aggressiveness.
Collapse
|
21
|
Motawi TMK, Sadik NAH, Fahim SA, Shouman SA. Combination of imatinib and clotrimazole enhances cell growth inhibition in T47D breast cancer cells. Chem Biol Interact 2015; 233:147-56. [PMID: 25863232 DOI: 10.1016/j.cbi.2015.03.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/13/2015] [Accepted: 03/25/2015] [Indexed: 01/19/2023]
Abstract
Imatinib mesylate (IM), a tyrosine kinase inhibitor, is used as targeted cancer therapy. However, mono-targeting by IM does not always achieve full tumor eradication and thus it is recommended to combine IM with other anticancer agents. Clotrimazole (CLT) is an antifungal azole derivative with promising anticancer effects due to inhibiting the activity of glycolytic enzymes. The present study aimed to evaluate the effect of combining CLT with IM on breast cancer cell line in an attempt to establish effective new combination. T47D human breast cancer cell line was treated with different concentrations of IM and/or CLT for 48 h. IM-CLT interaction was determined by isobologram equation and combination index. Cell viability was confirmed by measuring LDH activity. As indicators of glycolysis inhibition, the expression of hexokinase-2 (HK-2) and 6-phosphofructo-1-kinase (PFK-1) plus the activity of intracellular lactate dehydrogenase (LDH) and pyruvate kinase (PK) were determined. In addition, glucose consumption and adenosine triphosphate (ATP) production were measured. Moreover, nitric oxide (NO), vascular endothelial growth factor (VEGF) and hypoxia inducible factor-α (HIF-α) were also determined as they are modulators for glycolysis. This study demonstrated that IM or CLT synergistically inhibited cell growth in T47D as shown by combination and dose reduction indices. The combination of 15 μM IM and 20 μM CLT significantly decreased glucose consumption, activity of both PK and intracellular LDH, while increased leaked LDH, VEGF and NO in the medium compared to each drug alone. Furthermore the combination decreased gene expression of HK-2, PFK-1 and ATP content compared to the control. In conclusion, the synergistic effect of CLT on IM cytotoxicity in T47D cell line maybe mediated through inhibition of glycolysis and increasing both NO and VEGF. Further studies are required to confirm the efficiency and safety of this combination.
Collapse
Affiliation(s)
- Tarek M K Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nermin A H Sadik
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sally A Fahim
- Biochemistry Department, Faculty of Pharmacy, Ahram Canadian University, Cairo, Egypt
| | - Samia A Shouman
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
22
|
Ellina MI, Bouris P, Aletras AJ, Theocharis AD, Kletsas D, Karamanos NK. EGFR and HER2 exert distinct roles on colon cancer cell functional properties and expression of matrix macromolecules. Biochim Biophys Acta Gen Subj 2014; 1840:2651-61. [DOI: 10.1016/j.bbagen.2014.04.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/24/2014] [Accepted: 04/25/2014] [Indexed: 01/08/2023]
|
23
|
Lim SL, Ricciardelli C, Oehler MK, De Arao Tan IMD, Russell D, Grützner F. Overexpression of piRNA pathway genes in epithelial ovarian cancer. PLoS One 2014; 9:e99687. [PMID: 24932571 PMCID: PMC4059699 DOI: 10.1371/journal.pone.0099687] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 05/19/2014] [Indexed: 11/18/2022] Open
Abstract
The importance of the Piwi-interacting RNA (piRNA) pathway for germ cell maintenance, genome integrity, DNA methylation and retrotransposon control raises possible roles of this pathway in cancer. Indeed aberrant expression of human PIWI orthologs and Maelstrom has been observed in various cancers. In this study we explored the expression and function of piRNA pathway genes in human ovarian cancer, based on our recent work, which showed widespread expression of piRNA pathway genes in the mammalian. Our work shows that PIWIL1 and MAEL expression is significantly increased in malignant EOC (n = 25) compared to benign tumor tissues (n = 19) and normal ovarian tissue (n = 8). The expression of PIWIL3 is lower in malignant and benign tissues when compared to normal ovary. Sequencing of PIWIL1 transcript revealed that in many tumors deletion of exon 17 leads to the introduction of a premature stop codon in the PIWI domain, likely due to a splicing error. In situ hybridization on tumor sections revealed that L1, PIWIL1, 2 and MAEL are specifically expressed in epithelial cells (cancerous cells) of EOC. Furthermore, PIWIL2 and MAEL are co-expressed in the stromal cells adjacent to tumor cells. Since PIWIL1 and MAEL are up regulated in malignant EOC and expressed in the epithelial cells, we investigated if these two genes affect invasiveness of ovarian cancer cell lines that do not normally express these genes. PIWIL1 and MAEL were transiently over expressed in the ovarian cancer cell line SKOV3, followed by real-time measurements of cell invasiveness. Surprisingly both PIWIL1 and MAEL over expression decreased the invasiveness of SKOV3 cells. Our findings support a growing body of evidence that shows that genes in this pathway are upregulated in cancer. In ovarian cancer we show for the first time that Piwil1 transcript may often be abnormal result in non functional product. In contrast to what has been observed in other cell types, we found that PIWIL1 and MAEL have a repressive effect on cell invasiveness.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Argonaute Proteins/genetics
- Argonaute Proteins/metabolism
- Base Sequence
- Carcinoma, Ovarian Epithelial
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- DNA-Binding Proteins
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- In Situ Hybridization
- Middle Aged
- Molecular Sequence Data
- Neoplasm Invasiveness
- Neoplasm Staging
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/pathology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Ovary/metabolism
- Ovary/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction/genetics
- Transcription Factors
- Transfection
Collapse
Affiliation(s)
- Shu Ly Lim
- Robinson Research Institute, School of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Martin K. Oehler
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Izza M. D. De Arao Tan
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Darryl Russell
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Frank Grützner
- Robinson Research Institute, School of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
- * E-mail:
| |
Collapse
|
24
|
UMBREIT CLAUDIA, ADERHOLD CHRISTOPH, FABER ANNE, SAUTER ALEXANDER, HOFHEINZ RALFDIETER, STERN-STRAETER JENS, HOERMANN KARL, SCHULTZ JOHANNESDAVID. Imatinib-associated matrix metalloproteinase suppression in p16-positive squamous cell carcinoma compared to HPV-negative HNSCC cells in vitro. Oncol Rep 2014; 32:668-76. [DOI: 10.3892/or.2014.3225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/13/2014] [Indexed: 11/06/2022] Open
|
25
|
Gialeli C, Viola M, Barbouri D, Kletsas D, Passi A, Karamanos NK. Dynamic interplay between breast cancer cells and normal endothelium mediates the expression of matrix macromolecules, proteasome activity and functional properties of endothelial cells. Biochim Biophys Acta Gen Subj 2014; 1840:2549-59. [PMID: 24582970 DOI: 10.1016/j.bbagen.2014.02.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 02/06/2014] [Accepted: 02/14/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Breast cancer-endothelium interactions provide regulatory signals facilitating tumor progression. The endothelial cells have so far been mainly viewed in the context of tumor perfusion and relatively little is known regarding the effects of such paracrine interactions on the expression of extracellular matrix (ECM), proteasome activity and properties of endothelial cells. METHODS To address the effects of breast cancer cell (BCC) lines MDA-MB-231 and MCF-7 on the endothelial cells, two cell culture models were utilized; one involves endothelial cell culture in the presence of BCCs-derived conditioned media (CM) and the other co-culture of both cell populations in a Transwell system. Real-time PCR was utilized to evaluate gene expression, an immunofluorescence assay for proteasome activity, and functional assays (migration, adhesion and invasion) and immunofluorescence microscopy for cell integrity and properties. RESULTS BCC-CM decreases the cell migration of HUVEC. Adhesion and invasion of BCCs are favored by HUVEC and HUVEC-CM. HA levels and the expression of CD44 and HA synthase-2 by HUVEC are substantially upregulated in both cell culture approaches. Adhesion molecules, ICAM-1 and VCAM-1, are also highly upregulated, whereas MT1-MMP and MMP-2 expressions are significantly downregulated in both culture systems. Notably, the expression and activity of the proteasome β5 subunit are increased, especially by the action of MDA-MB-231-CM on HUVEC. CONCLUSIONS AND GENERAL SIGNIFICANCE BCCs significantly alter the expression of matrix macromolecules, proteasome activity and functional properties of endothelial cells. Deep understanding of such paracrine interactions will help to design novel drugs targeting breast cancer at the ECM level. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Ch Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), 26500 Patras, Greece
| | - M Viola
- Department of Surgery and Morphological Sciences, University of Insubria, Varese, Italy
| | - D Barbouri
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), 26500 Patras, Greece
| | - D Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biology, National Center of Scientific Research "Demokritos", Athens, Greece
| | - A Passi
- Department of Surgery and Morphological Sciences, University of Insubria, Varese, Italy
| | - N K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), 26500 Patras, Greece.
| |
Collapse
|
26
|
Vihinen P, Ala-Aho R, Kähäri VM. Diagnostic and prognostic role of matrix metalloproteases in cancer. ACTA ACUST UNITED AC 2013; 2:1025-39. [PMID: 23495924 DOI: 10.1517/17530059.2.9.1025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Matrix metalloproteases (MMPs) are key players in the progression and metastasis of cancer. MMPs cleave extracellular matrix components and in this way promote tumor growth, invasion and vascularization. MMPs also affect tumor progression by regulating availability and activity of growth factors, inflammatory cytokines and chemokines. Accordingly, several MMPs have been found to serve as prognostic indicators in solid tumors. Usually the increased levels of MMPs in patients' tumor tissue or serum/plasma are associated with poor outcome. Interestingly, recent results show that certain MMPs also serve as tumor suppressors. OBJECTIVE This review discusses the latest view on MMPs as diagnostic and prognostic indicators in cancer patients. METHODS Studies with clinical samples of 70 or more patients are included in particular. In addition, the possible roles of MMPs in future molecular diagnostics and in the evaluation of therapeutic responses are discussed. CONCLUSION MMP-9 in particular has shown prognostic value in various types of tumor, and its measurement in circulation, urine or tumor tissue might help in clinical surveillance of otherwise problematic patient cases. There is upcoming new knowledge on MMPs in therapy response evaluation, in which MMPs might be useful together with CT scans and other clinically more established prognostic factors. Certain MMPs have a dual role in terms of cancer-modulating properties and thus it is essential to evaluate their expression and function in tumor cells and host environment to select validated therapy targets but spare MMP antitargets.
Collapse
Affiliation(s)
- Pia Vihinen
- Turku University Hospital, Department of Oncology and Radiotherapy, POB 52, FIN-20521 Turku, Finland +358 2 313 0804 ; +358 2 313 2809 ;
| | | | | |
Collapse
|
27
|
Tang XP, Tang GD, Fang CY, Liang ZH, Zhang LY. Effects of ginsenoside Rh2 on growth and migration of pancreatic cancer cells. World J Gastroenterol 2013; 19:1582-1592. [PMID: 23538603 PMCID: PMC3602475 DOI: 10.3748/wjg.v19.i10.1582] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/17/2012] [Accepted: 12/17/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of ginsenoside Rh2 on the human pancreatic cancer cell line Bxpc-3.
METHODS: The human pancreatic cancer cell line Bxpc-3 was cultured in vitro and treated with or without ginsenoside Rh2. Growth rates for Bxpc-3 cells were assessed by methyl thiazolyl tetrazolium (MTT) and colony formation assays. Cell cycle changes were analyzed by flow cytometry. Apoptosis was measured by flow cytometry and Hoechst 33258 fluorescence staining. A scratch assay and a Matrigel invasion assay were used to detect cell migration and invasion. Expression of Bax, Bcl-2, survivin, cyclin D1, matrix metalloproteinase (MMP)-2, MMP-9, cleaved caspase-3, caspase-8, and caspase-9 mRNA were determined by reverse transcriptase-polymerase chain reaction (RT-PCR). Bax, Bcl-2, survivin, cyclin D1, cleaved caspase-3, caspase-8 and caspase-9 protein levels were examined by western blotting. Expression of MMP-2 and MMP-9 proteins in culture supernatants were determined by enzyme-linked immunosorbent assay (ELISA).
RESULTS: Rh2 significantly inhibited Bxpc-3 cell proliferation in a dose- and time-dependent manner, as evaluated by the MTT (P < 0.05) and colony formation assays (P < 0.05). Compared to the control group, Rh2 significantly increased the percentage of Bxpc-3 cells in the G0/G1 phase from 43.32% ± 2.17% to 71.32% ± 1.16%, which was accompanied by a decrease in S phase (from 50.86% ± 1.29% to 28.48% ± 1.18%) and G2/M phase (from 5.81% ± 1.19% to 0.20% ± 0.05%) in a dose-dependent manner (P < 0.05), suggesting that Rh2 arrested cell cycle progression at the G0/G1 phase, as measured by flow cytometry. Compared to the control group, cells treated with Rh2 showed significantly higher apoptosis ratios in a dose-dependent manner (percentage of early apoptotic cells: from 5.29% ± 2.28% to 38.90% ± 3.42% (F = 56.20, P < 0.05); percentage of late apoptotic cells: from 4.58% ± 1.42% to 36.32% ± 2.73% (F = 86.70, P < 0.05). Rh2 inhibited Bxpc-3 cell migration and invasion, as detected by scratch wound healing assay and Matrigel invasion assay [percentages of scratch wound healing for 12 h, 24 h and 48 h (control vs experimental group): 37.3% ± 4.8% vs 18.30% ± 1.65%, 58.7% ± 3.5% vs 38.00% ± 4.09% and 93.83% ± 4.65% vs 65.50% ± 4.09%, respectively; t = 6.489, t = 6.656 and t = 7.926, respectively, P < 0.05; the number of cells invading at various concentrations (0 μmol/L, 35 μmol/L, 45 μmol/L and 55 μmol/L): 81.10 ± 9.55, 46.40 ± 6.95, 24.70 ± 6.88 and 8.70 ± 3.34, respectively (F = 502.713, P < 0.05)]. RT-PCR, western blotting or ELISA showed that mRNA and protein expression of Bax, cleaved caspase-3 and caspase-9 were upregulated (P < 0.05), while mRNA and protein expression of Bcl-2, survivin, cyclin D1, MMP-2 and MMP-9 were downregulated (P < 0.05).
CONCLUSION: Ginsenoside Rh2 inhibits proliferation, migration and invasion and induces apoptosis of the human pancreatic cancer cell line Bxpc-3.
Collapse
|
28
|
Malavaki CJ, Roussidis AE, Gialeli C, Kletsas D, Tsegenidis T, Theocharis AD, Tzanakakis GN, Karamanos NK. Imatinib as a key inhibitor of the platelet-derived growth factor receptor mediated expression of cell surface heparan sulfate proteoglycans and functional properties of breast cancer cells. FEBS J 2013; 280:2477-89. [DOI: 10.1111/febs.12163] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 01/25/2013] [Accepted: 01/29/2013] [Indexed: 12/16/2022]
Affiliation(s)
| | - Andreas E. Roussidis
- Laboratory of Biochemistry; Department of Chemistry; University of Patras; Greece
| | | | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing; Institute of Biosciences and Applications; National Centre for Scientific Research‘Demokritos’; Athens; Greece
| | - Theodore Tsegenidis
- Laboratory of Biochemistry; Department of Chemistry; University of Patras; Greece
| | | | | | | |
Collapse
|
29
|
Expression of matrix macromolecules and functional properties of EGF-responsive colon cancer cells are inhibited by panitumumab. Invest New Drugs 2012; 31:516-24. [DOI: 10.1007/s10637-012-9875-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 08/23/2012] [Indexed: 10/27/2022]
|
30
|
Lu G, Zheng M, Zhu Y, Sha M, Wu Y, Han X. Selection of peptide inhibitor to matrix metalloproteinase-2 using phage display and its effects on pancreatic cancer cell lines PANC-1 and CFPAC-1. Int J Biol Sci 2012; 8:650-62. [PMID: 22606046 PMCID: PMC3354623 DOI: 10.7150/ijbs.3897] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 04/22/2012] [Indexed: 11/23/2022] Open
Abstract
Despite tremendous advances in cancer treatment and survival rates, pancreatic cancer remains one of the most deadly afflictions and the fourth leading cause of cancer deaths in the world. Matrix Metalloproteinases (MMPs) are thought to be involved in cancer progression. Matrix metalloproteinase (MMP)-2 is known to play a pivotal role in tumor invasion, metastasis and angiogenesis, and validated to be the anticancer target. Inhibition of MMP-2 activity is able to reduce the cancer cell invasion and suppress tumor growth in vivo. Two novel peptides, M204C4 and M205C4, which could specially inhibit MMP-2 activity, were identified by a phage display library screening. We showed that M204C4 and M205C4 inhibited the activity of MMP-2 in a dose dependent manner in vitro. Two peptides reduced MMP-2 mediated invasion of the pancreatic cancer cell lines PANC-1 and CFPAC-1, but not affected the expression and release of MMP-2. Furthermore, these two peptides could suppress tumor growth in vivo. Our results indicated that two peptides selected by phase display technology may be used as anticancer drugs in the future.
Collapse
Affiliation(s)
- Gao Lu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Clinical Diabetes Centre of Jiangsu Province, the Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
31
|
Huang DY, Chao Y, Tai MH, Yu YH, Lin WW. STI571 reduces TRAIL-induced apoptosis in colon cancer cells: c-Abl activation by the death receptor leads to stress kinase-dependent cell death. J Biomed Sci 2012; 19:35. [PMID: 22462553 PMCID: PMC3348077 DOI: 10.1186/1423-0127-19-35] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 03/30/2012] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In an effort to achieve better cancer therapies, we elucidated the combination cancer therapy of STI571 (an inhibitor of Bcr-Abl and clinically used for chronic myelogenous leukemia) and TNF-related apoptosis-inducing ligand (TRAIL, a developing antitumor agent) in leukemia, colon, and prostate cancer cells. METHODS Colon cancer (HCT116, SW480), prostate cancer (PC3, LNCaP) and leukemia (K562) cells were treated with STI571 and TRAIL. Cell viability was determined by MTT assay and sub-G1 appearance. Protein expression and kinase phosphorylation were determined by Western blotting. c-Abl and p73 activities were inhibited by target-specific small interfering (si)RNA. In vitro kinase assay of c-Abl was conducted using CRK as a substrate. RESULTS We found that STI571 exerts opposite effects on the antitumor activity of TRAIL. It enhanced cytotoxicity in TRAIL-treated K562 leukemia cells and reduced TRAIL-induced apoptosis in HCT116 and SW480 colon cancer cells, while having no effect on PC3 and LNCaP cells. In colon and prostate cancer cells, TRAIL caused c-Abl cleavage to the active form via a caspase pathway. Interestingly, JNK and p38 MAPK inhibitors effectively blocked TRAIL-induced toxicity in the colon, but not in prostate cancer cells. Next, we found that STI571 could attenuate TRAIL-induced c-Abl, JNK and p38 activation in HCT116 cells. In addition, siRNA targeting knockdown of c-Abl and p73 also reduced TRAIL-induced cytotoxicity, rendering HCT116 cells less responsive to stress kinase activation, and masking the cytoprotective effect of STI571. CONCLUSIONS All together we demonstrate a novel mediator role of p73 in activating the stress kinases p38 and JNK in the classical apoptotic pathway of TRAIL. TRAIL via caspase-dependent action can sequentially activate c-Abl, p73, and stress kinases, which contribute to apoptosis in colon cancer cells. Through the inhibition of c-Abl-mediated apoptotic p73 signaling, STI571 reduces the antitumor activity of TRAIL in colon cancer cells. Our results raise additional concerns when developing combination cancer therapy with TRAIL and STI571 in the future.
Collapse
Affiliation(s)
- Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
32
|
de Paula CAA, Coulson-Thomas VJ, Ferreira JG, Maza PK, Suzuki E, Nakahata AM, Nader HB, Sampaio MU, Oliva MLV. Enterolobium contortisiliquum trypsin inhibitor (EcTI), a plant proteinase inhibitor, decreases in vitro cell adhesion and invasion by inhibition of Src protein-focal adhesion kinase (FAK) signaling pathways. J Biol Chem 2011; 287:170-182. [PMID: 22039045 DOI: 10.1074/jbc.m111.263996] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tumor cell invasion is vital for cancer progression and metastasis. Adhesion, migration, and degradation of the extracellular matrix are important events involved in the establishment of cancer cells at a new site, and therefore molecular targets are sought to inhibit such processes. The effect of a plant proteinase inhibitor, Enterolobium contortisiliquum trypsin inhibitor (EcTI), on the adhesion, migration, and invasion of gastric cancer cells was the focus of this study. EcTI showed no effect on the proliferation of gastric cancer cells or fibroblasts but inhibited the adhesion, migration, and cell invasion of gastric cancer cells; however, EcTI had no effect upon the adhesion of fibroblasts. EcTI was shown to decrease the expression and disrupt the cellular organization of molecules involved in the formation and maturation of invadopodia, such as integrin β1, cortactin, neuronal Wiskott-Aldrich syndrome protein, membrane type 1 metalloprotease, and metalloproteinase-2. Moreover, gastric cancer cells treated with EcTI presented a significant decrease in intracellular phosphorylated Src and focal adhesion kinase, integrin-dependent cell signaling components. Together, these results indicate that EcTI inhibits the invasion of gastric cancer cells through alterations in integrin-dependent cell signaling pathways.
Collapse
Affiliation(s)
- Cláudia Alessandra Andrade de Paula
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Vivien Jane Coulson-Thomas
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Joana Gasperazzo Ferreira
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Paloma Korehisa Maza
- Department of Microbiology, Immunology, and Parasitology, Universidade Federal de São Paulo-Escola Paulista de Medicina, 04044-020 São Paulo, Brazil
| | - Erika Suzuki
- Department of Microbiology, Immunology, and Parasitology, Universidade Federal de São Paulo-Escola Paulista de Medicina, 04044-020 São Paulo, Brazil
| | - Adriana Miti Nakahata
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Helena Bonciani Nader
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Misako Uemura Sampaio
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil
| | - Maria Luiza V Oliva
- Department of Biochemistry and Molecular Biology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, 04044-020, Brazil.
| |
Collapse
|
33
|
Xia Z, Xing Y, Jeon J, Kim YP, Gall J, Dragulescu-Andrasi A, Gambhir SS, Rao J. Immobilizing reporters for molecular imaging of the extracellular microenvironment in living animals. ACS Chem Biol 2011; 6:1117-26. [PMID: 21830814 PMCID: PMC3199358 DOI: 10.1021/cb200135e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We report here an immobilization strategy using a collagen binding protein to deliver and confine synthetic reporters to the extracellular microenvironment in vivo for noninvasively imaging the activity of targets in the microenvironment. We show that the immobilization of reporters on collagens in the local microenvironment is highly efficient and physiologically stable for repetitive, long-term imaging. By using this strategy we successfully developed an immobilized bioluminescent activatable reporter and a dual-modality reporter to map and quantitatively image the activity of extracellular matrix metalloproteinases (MMP) in tumor-bearing mice. The inhibition of MMP activity by chemical inhibitor was also demonstrated in living subjects. We further demonstrated the general applicability of this immobilization strategy by imaging MMP activity at the inflammation site in a mouse model. Our results show that the in vivo immobilization of reporters can be used as a general strategy for probing the local extracellular microenvironment.
Collapse
Affiliation(s)
- Zuyong Xia
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| | - Yun Xing
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| | - Jongho Jeon
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| | - Young-Pil Kim
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| | - Jessica Gall
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| | - Anca Dragulescu-Andrasi
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| | - Sanjiv S. Gambhir
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
- Department of Bioengineering, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
- Department of Materials Science and Engineering, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology & Bio-X Program, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
- Department of Chemistry, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
- Biophysics and Cancer Biology Programs, Stanford University, 1201 Welch Road, Stanford, CA 94035-5484, USA
| |
Collapse
|
34
|
Tammali R, Saxena A, Srivastava SK, Ramana KV. Aldose reductase inhibition prevents hypoxia-induced increase in hypoxia-inducible factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF) by regulating 26 S proteasome-mediated protein degradation in human colon cancer cells. J Biol Chem 2011; 286:24089-100. [PMID: 21576240 DOI: 10.1074/jbc.m111.219733] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The development of intratumoral hypoxia, a hallmark of rapidly progressing solid tumors, renders tumor cells resistant to chemotherapy and radiation therapy. We have recently shown that inhibition of aldose reductase (AR), an enzyme that catalyzes the reduction of lipid aldehydes and their glutathione conjugates, prevents human colon cancer cell growth in culture as well as in nude mouse xenografts by inhibiting the NF-κB-dependent activation of oxidative stress-mediated inflammatory and carcinogenic markers. However, the role of AR in mediating hypoxic stress signals is not known. We therefore investigated the molecular mechanisms by which AR inhibition prevents the hypoxia-induced human colon cancer cells growth and invasion. Our results indicate that AR inhibition by the pharmacological inhibitor fidarestat or ablation by AR-specific siRNA prevents hypoxia-induced proliferation of HT29, SW480, and Caco-2 colon cancer cells. Furthermore, hypoxia-induced increase in the level of HIF-1α in colon cancer cells was significantly decreased by AR inhibition. During hypoxic conditions, treatment of HT29 cells with the AR inhibitor fidarestat significantly decreased the expression of vascular endothelial growth factor, a down target of HIF-1α, at both mRNA and protein levels and also prevented the activation of PI3K/AKT, GSK3β, Snail, and lysyl oxidase. Furthermore, inhibition of hypoxia-induced HIF-1α protein accumulation by AR inhibition was abolished in the presence of MG132, a potent inhibitor of the 26 S proteasome. In addition, AR inhibition also prevented the hypoxia-induced inflammatory molecules such as Cox-2 and PGE2 and expression of extracellular matrix proteins such as MMP2, vimentin, uPAR, and lysyl oxidase 2. In conclusion, our results indicate that AR mediates hypoxic signals, leading to tumor progression and invasion.
Collapse
Affiliation(s)
- Ravinder Tammali
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | |
Collapse
|
35
|
Turktekin M, Konac E, Onen HI, Alp E, Yilmaz A, Menevse S. Evaluation of the effects of the flavonoid apigenin on apoptotic pathway gene expression on the colon cancer cell line (HT29). J Med Food 2011; 14:1107-17. [PMID: 21548803 DOI: 10.1089/jmf.2010.0208] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Apigenin (4',5,7-trihydroxyflavone) is one of the leading components supporting targeted treatment options. We explored the cytotoxic and apoptotic effects of various doses of apigenin administered alone and together with 5-fluorouracil (5-FU)-a chemotherapeutic agent with high cytotoxicity-for different incubation periods, on morphologic, DNA, RNA (messenger RNA [mRNA]), and protein levels on the p53 mutant HT29 human colon adenocarcinoma cell line. Treatment with apigenin alone for a 72-hour incubation at 90-μM dose resulted in an apoptotic percentage of 24.92% (P=.001). A higher percentage (29.13%) was observed after treatment with the same dose of apigenin plus 5-FU for the same incubation period (P=.001). These results were confirmed as mRNA and protein expression levels of caspase-3 increased 2.567-fold and mRNA expression levels of caspase-8 increased 3.689-fold compared with the control group. On the other hand, mRNA expression levels of mammalian target of rapamycin (mTOR) and cyclin D1 (CCND1) decreased by 0.423-fold and 0.231-fold, respectively. To our knowledge this is the first study showing that treatment with apigenin alone results in cell cycle arrest through activation of caspase cascade and stimulation of apoptosis in HT29 cells. It also shows that use of apigenin plus 5-FU further increases this effect. This study draws attention to the probable clinical effectiveness of apigenin plus a chemotherapeutic agent with high cytotoxicity. It also highlights the induction of desirable apoptotic effects by targeting the caspase cascade pathway through administration of reduced doses for shorter incubation periods.
Collapse
Affiliation(s)
- Mehmet Turktekin
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
36
|
Hadler-Olsen E, Wetting HL, Ravuri C, Omair A, Rikardsen O, Svineng G, Kanapathippillai P, Winberg JO, Uhlin-Hansen L. Organ specific regulation of tumour invasiveness and gelatinolytic activity at the invasive front. Eur J Cancer 2011; 47:305-15. [DOI: 10.1016/j.ejca.2010.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 08/19/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
|
37
|
Kawada M, Inoue H, Ohba SI, Masuda T, Momose I, Ikeda D. Leucinostatin A inhibits prostate cancer growth through reduction of insulin-like growth factor-I expression in prostate stromal cells. Int J Cancer 2010; 126:810-8. [PMID: 19795463 DOI: 10.1002/ijc.24915] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Targeting stroma in tumor tissues is an attractive new strategy for cancer treatment. We developed in vitro coculture system, in which the growth of human prostate cancer DU-145 cells is stimulated by prostate stromal cells (PrSC) through insulin-like growth factor I (IGF-I). Using this system, we have been searching for small molecules that inhibit tumor growth through modulation of tumor-stromal cell interactions. As a result, we have found that leucinostatins and atpenins, natural antifungal antibiotics, inhibit the growth of DU-145 cells cocultured with PrSC more strongly than that of DU-145 cells alone. In this study we examined the antitumor effects of these small molecules in vitro and in vivo. When DU-145 cells were coinoculated with PrSC subcutaneously in nude mice, leucinostatin A was found to significantly suppress the tumor growth more than atpenin B. The antitumor effect of leucinostatin A in vivo was not obtained against the tumors of DU-145 cells alone. RT-PCR experiments revealed that leucinostatin A specifically inhibited IGF-I expression in PrSC without effect on expressions of other IGF axis molecules. Leucinostatins and atpenins are known to abrogate mitochondrial functions. However, when we used mitochondrial DNA-depleted, pseudo-rho(0) cells, we found that one of leucinostain A actions certainly depended on mitochondrial function, but it actually inhibited the growth of DU-145 cells more strongly in coculture with pseudo-rho(0) PrSC and reduced IGF-I expression in pseudo-rho(0) PrSC. Taken together, our results suggested that leucinostatin A inhibited prostate cancer cell growth through reduction of IGF-I expression in PrSC.
Collapse
Affiliation(s)
- Manabu Kawada
- Numazu Bio-Medical Research Institute, Microbial Chemistry Research Center, 18-24 Miyamoto, Numazu-shi, Shizuoka 410-0301, Japan.
| | | | | | | | | | | |
Collapse
|
38
|
Epidermal growth factor receptor protein expression and gene amplification in the chemorefractory metastatic embryonal carcinoma. Mod Pathol 2009; 22:7-12. [PMID: 18660793 DOI: 10.1038/modpathol.2008.133] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Testicular cancer is the most common cancer in young male patients. The combination of cisplatin-based chemotherapy and surgery has resulted in high cure rates. However, a small percentage of patients have cancers that are refractory to chemotherapy; treatment options for these patients are limited, and their prognosis is generally poor. Further studies are needed to explore the molecular pathogenesis pathways and potential targets of therapy for this group of highly aggressive tumors. We analyzed 21 patients who presented with metastatic embryonal carcinoma, were treated with chemotherapy, and subsequently underwent retroperitoneal lymph node dissection. Immunostaining for epidermal growth factor receptor (EGFR) was performed on paraffin-embedded tissue sections containing tumor from these specimens, using the avidin-biotin-peroxidase method. EGFR gene amplification was performed by interphase fluorescence in situ hybridization (FISH). Immunohistochemically, 9 of 21 cases (43%) demonstrated positive EGFR staining; 12 of 21 cases (57%) had absent or very limited EGFR expression. FISH revealed EGFR amplification in 1 case (5%), polysomy in 15 of 21 cases (71%), and normal disomy pattern in 5 of 21 cases (24%). A significant correlation between EGFR protein expression level and its chromosome polysomy/amplification was established (P=0.02). Our study showed that EGFR protein is frequently expressed in a subset of patients with chemorefractory metastatic embryonal carcinoma. EGFR chromosomal polysomy/amplification may be one of the mechanisms that cause increased EGFR protein expression, and could potentially be used as indication for anti-EGFR therapy.
Collapse
|
39
|
Kawada M, Inoue H, Usami I, Ikeda D. Phthoxazolin A inhibits prostate cancer growth by modulating tumor-stromal cell interactions. Cancer Sci 2009; 100:150-7. [PMID: 19018764 PMCID: PMC11158197 DOI: 10.1111/j.1349-7006.2008.00996.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 09/02/2008] [Accepted: 09/06/2008] [Indexed: 11/28/2022] Open
Abstract
Because stroma in tumor tissues can promote prostate cancer development, modulation of tumor-stromal cell interactions may represent an attractive new strategy for cancer treatment. Here, we report that phthoxazolin A and its analog inthomycin B inhibit the growth of human prostate cancer DU-145 cells by modulating tumor-stromal cell interactions. Using an in vitro coculture system, in which prostate cancer cell growth is upregulated by prostate stromal cells (PrSC), we found that phthoxazolin A and inthomycin B strongly inhibited the growth of DU-145 cells when in coculture with PrSC compared to DU-145 cells cultured alone. Although PrSC consist of both fibroblasts and myofibroblasts, phthoxazolin A and inthomycin B inhibited the expression of smooth muscle alpha-actin, a myofibroblast marker, without affecting vimentin and beta-actin expression. Because myofibroblasts secrete various factors that can promote tumor cell growth, we examined whether the inhibitory compounds affected the secretion of such factors from PrSC. Proteomic analysis and reverse transcription-polymerase chain reaction revealed that phthoxazolin A and inthomycin B inhibited the expression of several insulin-like growth factor binding proteins and insulin-like growth factor (IGF)-I by PrSC. Transforming growth factor-beta1 increased myofibroblast numbers and IGF-I levels in PrSC. Phthoxazolin A inhibited transforming growth factor-beta1 activity without altering phosphorylation of the downstream molecule smad2. Furthermore, conditioned medium from phthoxazolin A-treated PrSC failed to increase the phosphorylation of IGF-IR and Akt in DU-145 cells. Taken together, our results suggested that phthoxazolin A acts as a small-molecule modulator of tumor-stromal cell interactions that can indirectly suppress prostate cancer cell growth through inhibition of IGF-I production by PrSC.
Collapse
Affiliation(s)
- Manabu Kawada
- Drug Development Unit, Numazu Bio-Medical Research Institute, Microbial Chemistry Research Center, 18-24 Miyamoto, Numazu-shi, Shizuoka 410-0301, Japan.
| | | | | | | |
Collapse
|
40
|
Xia Z, Xing Y, So MK, Koh AL, Sinclair R, Rao J. Multiplex detection of protease activity with quantum dot nanosensors prepared by intein-mediated specific bioconjugation. Anal Chem 2008; 80:8649-55. [PMID: 18922019 PMCID: PMC2677517 DOI: 10.1021/ac801562f] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report here a protease sensing nanoplatform based on semiconductor nanocrystals or quantum dots (QDs) and bioluminescence resonance energy transfer (QD-BRET) to detect the protease activity in complex biological samples. These nanosensors consist of bioluminescent proteins as the BRET donor, quantum dots as the BRET acceptor, and protease substrates sandwiched between the two as a sensing group. An intein-mediated conjugation strategy was developed for site-specific conjugation of proteins to QDs in preparing these QD nanosensors. In this traceless ligation, the intein itself is spliced out and excluded from the final conjugation product. With this method, we have synthesized a series of QD nanosensors for highly sensitive detection of an important class of protease matrix metalloproteinase (MMP) activity. We demonstrated that these nanosensors can detect the MMP activity in buffers and in mouse serum with the sensitivity to a few nanograms per milliliter and secreted proteases by tumor cells. The suitability of these nanosensors for a multiplex protease assay has also been shown.
Collapse
Affiliation(s)
- Zuyong Xia
- Biophysics, Cancer Biology, and Molecular Imaging Programs, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484
| | - Yun Xing
- Biophysics, Cancer Biology, and Molecular Imaging Programs, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484
| | - Min-Kyung So
- Biophysics, Cancer Biology, and Molecular Imaging Programs, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484
| | - Ai Leen Koh
- Stanford Nanocharacterization Lab (SNL), Department of Material Science & Engineering, Stanford University, Stanford, CA 94305
| | - Robert Sinclair
- Stanford Nanocharacterization Lab (SNL), Department of Material Science & Engineering, Stanford University, Stanford, CA 94305
| | - Jianghong Rao
- Biophysics, Cancer Biology, and Molecular Imaging Programs, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California 94305-5484
| |
Collapse
|
41
|
Kousidou OC, Berdiaki A, Kletsas D, Zafiropoulos A, Theocharis AD, Tzanakakis GN, Karamanos NK. Estradiol-estrogen receptor: a key interplay of the expression of syndecan-2 and metalloproteinase-9 in breast cancer cells. Mol Oncol 2008; 2:223-32. [PMID: 19383343 DOI: 10.1016/j.molonc.2008.06.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 06/05/2008] [Accepted: 06/10/2008] [Indexed: 12/16/2022] Open
Abstract
Estrogens are related with the growth and development of target tissues and play a critical role in breast cancer progression. The effects of estrogens are mediated by the estrogen receptors ERalpha and ERbeta, which are members of the nuclear steroid receptor superfamily. To date, it is not known how these hormones elicit many of their effects on extracellular matrix molecules and how these effects can be connected with ER expression. For this purpose, the effect of estradiol on ER expression as well as on proteoglycan and metalloproteinase expression was studied. The effect of E2 on extracellular matrix molecule expression has been studied using ERalpha suppression in breast cancer cells. Our studies using ERalpha-positive MCF-7 cells show that estradiol affects the expression of syndecan-2, but not of syndecan-4, through ERalpha. Furthermore, the ability of estradiol to affect MMP-9 and TIMP-1 expression is connected with ERalpha status. Together, these data demonstrate the significant role of ERalpha on mediating the effect of estradiol on extracellular matrix molecules.
Collapse
Affiliation(s)
- Olga Ch Kousidou
- Laboratory of Biochemistry, Section of Organic Chemistry, Biochemistry and Natural Products, Department of Chemistry, University of Patras, 261 10 Patras, Greece
| | | | | | | | | | | | | |
Collapse
|
42
|
Stahtea XN, Kousidou OC, Roussidis AE, Tzanakakis GN, Karamanos NK. Small tyrosine kinase inhibitors as key molecules in the expression of metalloproteinases by solid tumors. Connect Tissue Res 2008; 49:211-4. [PMID: 18661345 DOI: 10.1080/03008200802143307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Critical steps for cancer cell growth, migration, invasion, and metastasis are the interactions of extracellular matrix (ECM) molecules with cells, the disconnection of intercellular adhesion, and the degradation of ECM. The latter is mediated mainly by metalloproteinases (MMPs), the expression and activation of which is related to various tyrosine kinase receptors (RTKs). The aberrant RTK activity is associated with the development and progress of various human cancers. Tyrosine kinase inhibitors (TKIs) are small molecules which compete with ATP for binding to the kinase domain of the RTKs and have been used for the treatment of solid tumors. In this review, the recent advances of the effects of TKIs on MMPs expressed by solid tumors are presented.
Collapse
Affiliation(s)
- Xanthi N Stahtea
- Department of Chemistry, Laboratory of Biochemistry, University of Patras, Patras, Greece
| | | | | | | | | |
Collapse
|