151
|
Chandrakesan P, Panneerselvam J, Qu D, Weygant N, May R, Bronze MS, Houchen CW. Regulatory Roles of Dclk1 in Epithelial Mesenchymal Transition and Cancer Stem Cells. ACTA ACUST UNITED AC 2016; 7. [PMID: 27335684 PMCID: PMC4913783 DOI: 10.4172/2157-2518.1000257] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The identification of functionally relevant subpopulations of therapy-resistant cancer cells is a challenge. These cells, intrinsically resistant to conventional therapy, can cause recurrence. Evidence has suggested that therapy-resistant cancer cells are likely epithelial–mesenchymal transition (EMT) cells and/or stem-like cells called cancer stem cells (CSCs). EMT, a normal embryological process that converts epithelial cells into mesenchymal cells, is frequently activated during cancer development and progression. CSCs are a small subpopulation of cancer cells within a tumor mass that have the ability to self-renew and maintain tumor-initiating capacity by giving rise to heterogeneous lineages of cancer cells that comprise the whole tumor. Although the origin of CSCs and EMT cells remains to be fully explored, a growing body of evidence has indicated that the biology of EMT and CSCs is strongly linked. Doublecortin-like kinase 1 (DCLK1), a cancer stem cell marker, is functionally involved in maintaining cancer stemness and the process of EMT important for cancer initiation, cancer metastasis, and secondary tumor formation. Therefore, targeting these cells may provide new strategies to overcome tumor heterogeneity, therapeutic resistance, and cancer relapse. In this review, we will provide a potential mechanistic link between EMT induction and the emergence of CSCs for the origin and progression of cancer. We will highlight the functional activity of DCLK1 in supporting EMT and cancer cell self-renewal, which will lead us to a better understanding of DCLK1 expression in cancer development and progression, and help us to develop targeted therapies for effective cancer treatment.
Collapse
Affiliation(s)
- P Chandrakesan
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Stephenson Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - J Panneerselvam
- Stephenson Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - D Qu
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Stephenson Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - N Weygant
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - R May
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - M S Bronze
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - C W Houchen
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Stephenson Oklahoma Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA; COARE Biotechnology, Oklahoma City, OK, USA
| |
Collapse
|
152
|
Long H, Xiang T, Qi W, Huang J, Chen J, He L, Liang Z, Guo B, Li Y, Xie R, Zhu B. CD133+ ovarian cancer stem-like cells promote non-stem cancer cell metastasis via CCL5 induced epithelial-mesenchymal transition. Oncotarget 2016; 6:5846-59. [PMID: 25788271 PMCID: PMC4467406 DOI: 10.18632/oncotarget.3462] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/20/2015] [Indexed: 01/06/2023] Open
Abstract
Cancer stem cells (CSCs, also called cancer stem-like cells, CSLCs) can function as "seed cells" for tumor recurrence and metastasis. Here, we report that, in the presence of CD133+ ovarian CSLCs, CD133- non-CSLCs can undergo an epithelial-mesenchymal transition (EMT)-like process and display enhanced metastatic capacity in vitro and in vivo. Highly elevated expression of chemokine (C-C motif) ligand 5 (CCL5) and its receptors chemokine (C-C motif) receptor (CCR) 1/3/5 are observed in clinical and murine metastatic tumor tissues from epithelial ovarian carcinomas. Mechanistically, paracrine CCL5 from ovarian CSLCs activates the NF-κB signaling pathway in ovarian non-CSLCs via binding CCR1/3/5, thereby inducing EMT and tumor invasion. Taken together, our results redefine the metastatic potential of non-stem cancer cells and provide evidence that targeting the CCL5:CCR1/3/5-NF-κB pathway could be an effective strategy to prevent ovarian cancer metastasis.
Collapse
Affiliation(s)
- Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Tong Xiang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wei Qi
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jiani Huang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Junying Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Luhang He
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Zhiqing Liang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bo Guo
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yongsheng Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rongkai Xie
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| |
Collapse
|
153
|
Enhancement of tumor initiation and expression of KCNMA1, MORF4L2 and ASPM genes in the adenocarcinoma of lung xenograft after vorinostat treatment. Oncotarget 2016; 6:8663-75. [PMID: 25796627 PMCID: PMC4496174 DOI: 10.18632/oncotarget.3536] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/12/2015] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) are usually tolerant to chemotherapy and radiotherapy and associated with tumor relapse. Suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor (HDACI), is currently being used in clinical trials of lung cancer. However, SAHA facilitates the formation of induced pluripotent stem cells from somatic cells. We hypothesized that SAHA would mediate the CSCs properties and subsequently confer a more malignant phenotype in lung cancer. Transfected H1299 lung cancer cells, which stably expresses a triple fused reporter gene (DsRedm-Fluc-tTKsr39) under the control of CMV promoter was used to establish a xenograft mouse model. After the treatment of SAHA, H1299 cell line and tumor xenografts were sorted by fluorescence-activated cell sorting (FACS) based on aldehyde dehydrogenase (ALDH) activity. We found that SAHA could suppress the growth of xenografted H1299 tumors with decreased proportion of ALDHbr lung cancer cells indicating that SAHA may target CSCs. However, SAHA significantly enhanced the tumor initiating capacity and the expression of malignant genes such as KCNMA1, MORF4L2 and ASPM in the remaining living ALDHbr cells. These findings suggested that SAHA treatment created a more drug-resistant state in residual ALDHbr cells. The in vivo imaging technique may facilitate searching and characterization of CSCs.
Collapse
|
154
|
Choi YJ, Baek GY, Park HR, Jo SK, Jung U. Smad2/3-Regulated Expression of DLX2 Is Associated with Radiation-Induced Epithelial-Mesenchymal Transition and Radioresistance of A549 and MDA-MB-231 Human Cancer Cell Lines. PLoS One 2016; 11:e0147343. [PMID: 26799321 PMCID: PMC4723265 DOI: 10.1371/journal.pone.0147343] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 12/31/2015] [Indexed: 01/27/2023] Open
Abstract
The control of radioresistance and metastatic potential of surviving cancer cells is important for improving cancer eradication by radiotheraphy. The distal-less homeobox2 (DLX2) gene encodes for a homeobox transcription factor involved in morphogenesis and its deregulation was found in human solid tumors and hematologic malignancies. Here we investigated the role of DLX2 in association with radiation-induced epithelial to mesenchymal transition (EMT) and stem cell-like properties and its regulation by Smad2/3 signaling in irradiated A549 and MDA-MB-231 human cancer cell lines. In irradiated A549 and MDA-MB-231 cells, EMT was induced as demonstrated by EMT marker expression, phosphorylation of Smad2/3, and migratory and invasive ability. Also, irradiated A549 and MDA-MB-231 cells showed increased cancer stem cells (CSCs) marker. Interestingly, DLX2 was overexpressed upon irradiation. Therefore, we examined the role of DLX2 in radiation-induced EMT and radioresistance. The overexpression of DLX2 alone induced EMT, migration and invasion, and CSC marker expression. The reduced colony-forming ability in irradiated cells was partially restored by DLX2 overexpression. On the other hand, the depletion of DLX2 using si-RNA abolished radiation-induced EMT, CSC marker expression, and phosphorylation of Smad2/3 in irradiated A549 and MDA-MB-231 cells. Also, depletion of DLX2 increased the radiation sensitivity in both cell lines. Moreover, knockdown of Smad2/3, a key activator of TGF-β1 pathway, abrogated the radiation-induced DLX2 expression, indicating that radiation-induced DLX2 expression is dependent on Smad2/3 signaling. These results demonstrated that DLX2 plays a crucial role in radioresistance, radiation-induced EMT and CSC marker expression, and the expression of DLX2 is regulated by Smad2/3 signaling in A549 and MDA-MB-231 cell lines.
Collapse
Affiliation(s)
- Yeo-Jin Choi
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Ga-Young Baek
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Hae-Ran Park
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sung-Kee Jo
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Uhee Jung
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
155
|
Deng JJ, Zhang W, Xu XM, Zhang F, Tao WP, Ye JJ, Ge W. Twist mediates an aggressive phenotype in human colorectal cancer cells. Int J Oncol 2016; 48:1117-24. [PMID: 26782761 DOI: 10.3892/ijo.2016.3342] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 12/08/2015] [Indexed: 12/13/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial process providing cancer cells with the ability to migrate and metastasize to distant sites. Recently, EMT was shown to be associated with the cancer stem cell (CSC) phenotype and chemoresistance. Twist is a transcription factor that regulates EMT in a various cancer cells, including colorectal cancer (CRC). Our study was done to determine the role of Twist in mediating aggressive phenotype in CRC. Human CRC cell lines were transduced with a retroviral Twist construct or vector control. Migration and invasion abilities were determined in vitro using modified Boyden chamber assays. Mammosphere formation assay was performed to detect CSC characteristics. EMT and CSC markers were detected using western blotting and RT-PCR. Chemosensitivity to oxaliplatin of the transfected cells were determined by the MTT assay. Human CRC specimens were stained for Twist and P-gp expression. Twist overexpression triggered EMT and a CSC-like phenotype in human CRC cells and enhanced cell migration, invasion and mammosphere formation abilities. In addition, Twist-overexpressing CRC cells were more chemo-resistant to oxaliplatin than control cells. Furthermore, Twist over-expression increased P-gp expression in CRC cells, which is a transmembrane glycoprotein conferred multidrug-resistance phenotype to various cancer cells. Importantly, Twist and P-gp were expressed correlatively in human CRC specimens. Thus, Twist is a potential therapeutic target in metastatic CRC.
Collapse
Affiliation(s)
- Jun-Jian Deng
- Department of Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xi-Ming Xu
- Department of Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fan Zhang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei-Ping Tao
- Department of Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jun-Jie Ye
- Department of Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Ge
- Department of Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
156
|
Park SJ, Kim JG, Kim ND, Yang K, Shim JW, Heo K. Estradiol, TGF-β1 and hypoxia promote breast cancer stemness and EMT-mediated breast cancer migration. Oncol Lett 2016; 11:1895-1902. [PMID: 26998096 DOI: 10.3892/ol.2016.4115] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 12/11/2015] [Indexed: 12/25/2022] Open
Abstract
Breast cancer is one of the most common cancer types among women, acting as a distinct cause of mortality, and has a high incidence of recurrence. External stimuli, including 17β-estradiol (E2), transforming growth factor (TGF)-β1 and hypoxia, may be important in breast cancer growth and metastasis. However, the effects of these stimuli on breast cancer stem cell (CSC) regulation have not been fully investigated. In the present study, the proportion of cluster of differentiation (CD)44+/CD24-/low cells increased following treatment with E2, TGF-β1 and hypoxia in MCF-7 cells. The expression of CSC markers, including SOX2, KLF4 and ABCG2, was upregulated continually by E2, TGF-β1 and hypoxia. In addition, the expression levels of epithelial-mesenchymal transition-associated factors increased following treatment with E2, TGF-β1 and hypoxia. Therefore, the migration ability of E2-, TGF-β1- and hypoxia-treated MCF-7 cells was enhanced compared with control cells. In addition, the enhancement of apoptosis by 5-flurouracil or radiation was abolished following treatment with E2, TGF-β1 and hypoxia. These results indicate that E2, TGF-β1 and hypoxia are important for regulating breast CSCs, and that the modulation of the microenvironment in tumors may improve the efficiency of breast cancer therapy.
Collapse
Affiliation(s)
- Seong-Joon Park
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea; Department of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Joong-Gook Kim
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| | - Nam Deuk Kim
- Department of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Kwangmo Yang
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea; Department of Radiation Oncology, Dongnam Institute of Radiological & Medical Sciences, Busan 46033, Republic of Korea; Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Republic of Korea
| | - Jae Woong Shim
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| | - Kyu Heo
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| |
Collapse
|
157
|
Morphine promotes cancer stem cell properties, contributing to chemoresistance in breast cancer. Oncotarget 2016; 6:3963-76. [PMID: 25686831 PMCID: PMC4414166 DOI: 10.18632/oncotarget.2894] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 12/11/2014] [Indexed: 01/24/2023] Open
Abstract
Morphine is an opioid analgesic drug commonly used for pain relief in cancer patients. Here, we report that morphine enhances the mammosphere forming capacity and increases the expression of stemness-related transcription factors Oct4, Sox2 and Nanog. Treatment with morphine leads to enrichment of a side population fraction in MCF-7 cells and the CD44+/CD24−/low population in BT549 cells. Consistently, morphine activates Wnt/β-catenin signaling to induce epithelial to mesenchymal transition and promotes metastasis. Moreover, morphine decreases the sensitivity of traditional anti-cancer drugs in breast cancer cells. Nalmefene, an antagonist of morphine, reverses morphine-induced cancer stem cell properties and chemoresistance in breast cancer. In addition, nalmefene abolishes morphine enhancing tumorigenesis in a NOD/SCID mouse model. In conclusion, our findings demonstrate that morphine contributes to chemoresistance via expanding the population of cancer stem cells and promotes tumor growth, thereby revealing a novel role of morphine and providing some new guides in clinical use of morphine.
Collapse
|
158
|
Abstract
Hes1 is one mammalian counterpart of the Hairy and Enhancer of split proteins that play a critical role in many physiological processes including cellular differentiation, cell cycle arrest, apoptosis and self-renewal ability. Recent studies have shown that Hes1 functions in the maintenance of cancer stem cells (CSCs), metastasis and antagonizing drug-induced apoptosis. Pathways that are involved in the up-regulation of Hes1 level canonically or non-canonically, such as the Hedgehog, Wnt and hypoxia pathways are frequently aberrant in cancer cells. Here, we summarize the recent data supporting the idea that Hes1 may have an important function in the maintenance of cancer stem cells self-renewal, cancer metastasis, and epithelial-mesenchymal transition (EMT) process induction, as well as chemotherapy resistance, and conclude with the possible mechanisms by which Hes1 functions have their effect, as well as their crosstalk with other carcinogenic signaling pathways.
Collapse
Key Words
- ABC, ATP-binding cassette
- CSCs, cancer stem cells
- CSL, CBF1/ Suppressor of Hairless / Lag1
- EMT, epithelial–mesenchymal transition
- GSI, γ-secretase inhibitor
- HDACs, histone deacetylases
- Hes1
- MAML, Mastermind-like protein family
- MASH-1, Mammalian achaete-scute homolog-1
- NICD, Notch intracellular domain
- Notch signaling pathway
- Runx2, Runt-related protein 2
- TLE, transducin-like Enhancer of split
- bHLH, basic helix-loop-helix
- cancer stem cell
- chemotherapy resistance
- dnMAM, dominant-negative mastermind
- metastasis
- non-canonical Notch
Collapse
|
159
|
Glyceollin I Reverses Epithelial to Mesenchymal Transition in Letrozole Resistant Breast Cancer through ZEB1. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 13:ijerph13010010. [PMID: 26703648 PMCID: PMC4730401 DOI: 10.3390/ijerph13010010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 08/20/2015] [Accepted: 08/24/2015] [Indexed: 01/01/2023]
Abstract
Although aromatase inhibitors are standard endocrine therapy for postmenopausal women with early-stage metastatic estrogen-dependent breast cancer, they are limited by the development of drug resistance. A better understanding of this process is critical towards designing novel strategies for disease management. Previously, we demonstrated a global proteomic signature of letrozole-resistance associated with hormone-independence, enhanced cell motility and implications of epithelial mesenchymal transition (EMT). Letrozole-resistant breast cancer cells (LTLT-Ca) were treated with a novel phytoalexin, glyceollin I, and exhibited morphological characteristics synonymous with an epithelial phenotype and decreased proliferation. Letrozole-resistance increased Zinc Finger E-Box Binding Homeobox 1 (ZEB1) expression (4.51-fold), while glyceollin I treatment caused a -3.39-fold reduction. Immunofluorescence analyses resulted of glyceollin I-induced increase and decrease in E-cadherin and ZEB1, respectively. In vivo studies performed in ovariectomized, female nude mice indicated that glyceollin treated tumors stained weakly for ZEB1 and N-cadherin and strongly for E-cadherin. Compared to letrozole-sensitive cells, LTLT-Ca cells displayed enhanced motility, however in the presence of glyceollin I, exhibited a 68% and 83% decrease in invasion and migration, respectively. These effects of glyceollin I were mediated in part by inhibition of ZEB1, thus indicating therapeutic potential of glyceollin I in targeting EMT in letrozole resistant breast cancer.
Collapse
|
160
|
MiRNAs and Other Epigenetic Changes as Biomarkers in Triple Negative Breast Cancer. Int J Mol Sci 2015; 16:28347-76. [PMID: 26633365 PMCID: PMC4691037 DOI: 10.3390/ijms161226090] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/30/2015] [Accepted: 11/12/2015] [Indexed: 02/06/2023] Open
Abstract
Triple negative breast cancer (TNBC) is characterised by the lack of receptors for estrogen (ER), progesterone (PR), and human epidermal growth factor 2 (HER2). Since it cannot be treated by current endocrine therapies which target these receptors and due to its aggressive nature, it has one of the worst prognoses of all breast cancer subtypes. The only treatments remain chemo- and/or radio-therapy and surgery and because of this, novel biomarkers or treatment targets are urgently required to improve disease outcomes. MicroRNAs represent an attractive candidate for targeted therapies against TNBC, due to their natural ability to act as antisense interactors and regulators of entire gene sets involved in malignancy and their superiority over mRNA profiling to accurately classify disease. Here we review the current knowledge regarding miRNAs as biomarkers in TNBC and their potential use as therapeutic targets in this disease. Further, we review other epigenetic changes and interactions of these changes with microRNAs in this breast cancer subtype, which may lead to the discovery of new treatment targets for TNBC.
Collapse
|
161
|
Chen X, Zhang H, Zhu H, Yang X, Yang Y, Yang Y, Min H, Chen G, Liu J, Lu J, Cheng H, Sun X. Endostatin combined with radiotherapy suppresses vasculogenic mimicry formation through inhibition of epithelial-mesenchymal transition in esophageal cancer. Tumour Biol 2015; 37:4679-88. [PMID: 26511968 DOI: 10.1007/s13277-015-4284-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/19/2015] [Indexed: 02/08/2023] Open
Abstract
The growth of solid tumors requires angiogenesis to provide oxygen and nutrients and to support cell proliferation. The switch from an avascular to a vascular phenotype is typically related to acceleration of tumor growth. Anti-angiogenic therapy is becoming a very promising way for malignant tumors. Meanwhile, malignant tumor cells themselves were able to develop the formation of cell-lined vessels that contribute to tumor neovascularization and supply the nutrients and oxygen, which is called vasculogenic mimicry (VM). However, the molecular mechanism of VM remains unclear. The purpose of this study was to investigate the efficacy of the novel recombinant human endostatin (rh-Endo) protein combined with radiotherapy on human esophageal squamous cell carcinoma (ESCC) cell lines Eca-109 and TE13. Our results showed that rh-Endo combined with radiotherapy significantly inhibited the proliferation, migration, invasion, and VM of human esophageal cancer cells in a dose-dependent manner; however, it has no direct effect on apoptosis of carcinoma cells, which indicated that rh-Endo combined with radiotherapy significantly changed the microenvironment of esophageal carcinoma, and played an important role in preventing distant metastasis. Our findings suggested that rh-Endo inhibited the metastasis of esophageal cancer and the activation of AKT pathway, and the down-regulation of epithelial-mesenchymal transition (EMT) may be associated with such effect of rh-Endo. These results also supported the bright prospect of rh-Endo combined with radiotherapy for clinical applications in the future.
Collapse
Affiliation(s)
- Xiaochen Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hao Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hongcheng Zhu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xi Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yuehua Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yan Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hua Min
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Guangzong Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Jia Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Jing Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hongyan Cheng
- Department of General Internal Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
162
|
Khorrami S, Zavaran Hosseini A, Mowla SJ, Malekzadeh R. Verification of ALDH Activity as a Biomarker in Colon Cancer Stem Cells-Derived HT-29 Cell Line. IRANIAN JOURNAL OF CANCER PREVENTION 2015; 8:e3446. [PMID: 26634106 PMCID: PMC4667234 DOI: 10.17795/ijcp-3446] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/01/2015] [Indexed: 01/16/2023]
Abstract
Background: Recent evidence has suggested that epithelial cancers including colorectal cancer (CRC) have driven by a small population of self-renewing, multi-potent cells termed cancer stem cells (CSCs) which could be responsible for recurrence of cancer. Aldehyde dehydrogenase 1 (ALDH1) activity has used as a functional stem cell biomarker to isolate CSCs in different cancers such as colorectal cancer. Objectives: The main aim of this research was to determine the utility of ALDH1 activity along with CD44 and EPCAM in identifying stem cell-like cells in human HT-29 colonic adenocarcinoma cell line. Materials and Methods: In this experimental study, colon CSCs biomarkers including CD44, EPCAM and ALDH1 in colonospheres and parent cells have analyzed by flow cytometry. The expression levels of stemness genes in spheroid and parental cells have investigated using SYBR Green real-time PCR. In addition, in vivo xenografts assay has performed to determine tumorigenic potential of tumor spheroid cells in nude mice. Results: According to results, over 92% of spheroids were CD44+/EpCAM+, while parent cells only have expressed 38% of CD44/EpCAM biomarkers (P < 0.001). Controversially, ALDH activity was about 2-fold higher in the parent cells than spheroid cells (P < 0.05). In comparison with the parental cells, expression levels of ‘‘stemness’’ genes, like Sox2, Oct4, Nanog, C-myc, and Klf4 have significantly increased in colonosphere cells (P < 0.05). Further, administration of 2500 spheroids could be sufficient to initiate tumor growth in nude mice, while 1x106 of parental cells has needed to form tumor. Conclusions: For the first time, we have shown that colonospheres with low ALDH1 activity has indicated increased tumorigenic potential and stemness properties. So, it hasn’t seemed that ALDH1 could become a useful biomarker to identify CSCs population in HT-29 cell line.
Collapse
Affiliation(s)
- Samaneh Khorrami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
| | - Ahmad Zavaran Hosseini
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences , Tarbiat Modares University, Tehran, IR Iran
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, IR Iran
| |
Collapse
|
163
|
Martínez-Ramírez AS, Garay E, García-Carrancá A, Vázquez-Cuevas FG. The P2RY2 Receptor Induces Carcinoma Cell Migration and EMT Through Cross-Talk With Epidermal Growth Factor Receptor. J Cell Biochem 2015; 117:1016-26. [PMID: 26443721 DOI: 10.1002/jcb.25390] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/05/2015] [Indexed: 11/11/2022]
Abstract
Extracellular nucleotides are signaling elements present in the tumor microenvironment; however, their role in tumor growth is not completely understood. In the present study, we asked whether nucleotides regulate cell migration in ovarian carcinoma-derived cells. We observed that 100 μM UTP induced migration in SKOV-3 cells (1.57 ± 0.08 fold over basal), and RT-PCR showed expression of transcripts for the P2RY2 and P2RY4 receptors. Knockdown of P2RY2 expression in SKOV-3 cells (P2RY2-KD) abolished the UTP-induced migration. The mechanism activated by UTP to induce migration involves transactivation of the epidermal growth factor receptor (EGFR) since we observed that the EGFR kinase inhibitor AG1478 and the PI3K inhibitor Wortmannin inhibit this response (to 0.76 ± 0.23 and 0.46 ± 0.14 relative to the control, respectively). In agreement with these observations, UTP was able to modify the phosphorylation state of the EGFR; likewise, the induction of ERK1/2 phosphorylation promoted by UTP was abolished by a 30-60 min treatment with AG1478. Our data also suggested that the enhanced cell migration involves the epithelium to mesenchymal transition (EMT) process, since a 12 h stimulation of SKOV-3 cells with 100 μM UTP showed an increase in vimentin and SNAIL protein levels (459.8 ± 132.4% over basal for SNAIL). Interestingly, treatment with apyrase (10 U/mL) reduces the migration of control cells and induces a considerable enrichment of E-cadherin in the cell-cell contacts, favoring an epithelial phenotype and strongly suggesting that the nucleotides released by tumor cells and acting through the P2RY2 receptor are potential regulators of invasiveness.
Collapse
Affiliation(s)
- A S Martínez-Ramírez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla Querétaro, CP 76230, Querétaro, Mexico
| | - E Garay
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla Querétaro, CP 76230, Querétaro, Mexico
| | - A García-Carrancá
- Laboratorio de Virus y Cáncer, Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. San Fernando #22, Colonia Sección XVI, Tlalpan, CP 14080, Mexico.,División de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, México. Av. San Fernando #22, Colonia Sección XVI, Tlalpan, CP 14080, Mexico
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Juriquilla Querétaro, CP 76230, Querétaro, Mexico
| |
Collapse
|
164
|
Hong D, Jang SY, Jang EH, Jung B, Cho IH, Park MJ, Jeong SY, Kim JH. Shikonin as an inhibitor of the LPS-induced epithelial-to-mesenchymal transition in human breast cancer cells. Int J Mol Med 2015; 36:1601-6. [PMID: 26498588 DOI: 10.3892/ijmm.2015.2373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 10/02/2015] [Indexed: 11/05/2022] Open
Abstract
Shikonin (SK), a natural naphthoquinone isolated from the Chinese medicinal herb, has been known to suppress the proliferation of several cancer cells. However, its role in the epithelial mesenchymal transition (EMT) has yet to be demonstrated. The aim of the present study was to examine the effects of SK on EMT. Lipopolysaccharide (LPS) induced EMT-like phenotypic changes, enhancing cell migration and invasion. SK markedly reduced the expression of the LPS-induced EMT markers, including N-cadherin in MDA-MB‑231 cells, and increased the expression of E-cadherin in MCF-7 cells. SK also inhibited cell migration and invasion in vitro. The effects of SK on the LPS-induced EMT were mediated by the inactivation of the NF-κB-Snail signaling pathway. The results provided new evidence that SK suppresses breast cancer cell invasion and migration by inhibiting the EMT. Therefore, SK is a potentially effective anticancer agent for breast tumors, by inhibiting metastasis.
Collapse
Affiliation(s)
- Darong Hong
- Department of Life and Nanopharmaceutical Science, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Soon Young Jang
- Department of Pharmacy, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Eun Hyang Jang
- Department of Pharmacy, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Bom Jung
- Department of Pharmacy, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - In-Hye Cho
- Department of Pharmacy, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Min-Ju Park
- Department of Pharmacy, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Seo Young Jeong
- Department of Life and Nanopharmaceutical Science, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Jong-Ho Kim
- Department of Pharmacy, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| |
Collapse
|
165
|
Guo Q, Qin W. DKK3 blocked translocation of β-catenin/EMT induced by hypoxia and improved gemcitabine therapeutic effect in pancreatic cancer Bxpc-3 cell. J Cell Mol Med 2015; 19:2832-41. [PMID: 26395974 PMCID: PMC4687707 DOI: 10.1111/jcmm.12675] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 08/03/2015] [Indexed: 12/12/2022] Open
Abstract
The Wnt/β-catenin signalling pathway is activated in pancreatic cancer initiation and progression. Dickkopf-related protein 3 (DKK3) is a member of the human Dickkopf family and an antagonist of Wnt ligand activity. However, the function of DKK3 in this pathway in pancreatic cancer is rarely known. We examined the expression of DKK3 in six human pancreatic cancer cell lines, 75 pancreatic cancer and 75 adjacent non-cancerous tissues. Dickkopf-related protein 3 was frequently silenced and methylation in pancreatic cancer cell lines (3/6). The expression of DKK3 was significantly lower in pancreatic cancer tissues than in adjacent normal pancreas tissues. Further, ectopic expression of DKK3 inhibits nuclear translocation of β-catenin induced by hypoxia in pancreatic cancer Bxpc-3 cell. The forced expression of DKK3 markedly suppressed migration and the stem cell-like phenotype of pancreatic cancer Bxpc-3 cell in hypoxic conditions through reversing epithelial-mesenchymal transition (EMT). The stable expression of DKK3 sensitizes pancreatic cancer Bxpc-3 cell to gemcitabine, delays tumour growth and augments gemcitabine therapeutic effect in pancreatic cancer xenotransplantation model. Thus, we conclude from our finding that DKK3 is a tumour suppressor and improved gemcitabine therapeutic effect through inducing apoptosis and regulating β-catenin/EMT signalling in pancreatic cancer Bxpc-3 cell.
Collapse
Affiliation(s)
- Qingqu Guo
- Department of Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenjie Qin
- Department of Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
166
|
Deciphering Seed Sequence Based Off-Target Effects in a Large-Scale RNAi Reporter Screen for E-Cadherin Expression. PLoS One 2015; 10:e0137640. [PMID: 26361354 PMCID: PMC4567318 DOI: 10.1371/journal.pone.0137640] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 08/19/2015] [Indexed: 12/28/2022] Open
Abstract
Functional RNAi based screening is affected by large numbers of false positive and negative hits due to prevalent sequence based off-target effects. We performed a druggable genome targeting siRNA screen intended to identify novel regulators of E-cadherin (CDH1) expression, a known key player in epithelial mesenchymal transition (EMT). Analysis of primary screening results indicated a large number of false-positive hits. To address these crucial difficulties we developed an analysis method, SENSORS, which, similar to published methods, is a seed enrichment strategy for analyzing siRNA off-targets in RNAi screens. Using our approach, we were able to demonstrate that accounting for seed based off-target effects stratifies primary screening results and enables the discovery of additional screening hits. While traditional hit detection methods are prone to false positive results which are undetected, we were able to identify false positive hits robustly. Transcription factor MYBL1 was identified as a putative novel target required for CDH1 expression and verified experimentally. No siRNA pool targeting MYBL1 was present in the used siRNA library. Instead, MYBL1 was identified as a putative CDH1 regulating target solely based on the SENSORS off-target score, i.e. as a gene that is a cause for off-target effects down regulating E-cadherin expression.
Collapse
|
167
|
Zhu F, Li X, Jiang Y, Zhu H, Zhang H, Zhang C, Zhao Y, Luo F. GdCl3 suppresses the malignant potential of hepatocellular carcinoma by inhibiting the expression of CD206 in tumor‑associated macrophages. Oncol Rep 2015; 34:2643-55. [PMID: 26352004 DOI: 10.3892/or.2015.4268] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/06/2015] [Indexed: 11/06/2022] Open
Abstract
In the present study, we aimed to ascertain whether there is a correlation between CD206 expression in tumor associated-macrophages (TAMs) and the prognosis of primary hepatocellular carcinomas (HCC) and we investigated the effect of GdCl3 on HCC. The expression of CD206 in HCC tumor tissues and peri-carcinoma tissues was measured using an array for liver tissues. The effects of GdCl3 on CD206 expression were examined in stimulated RAW264.7 cells. Target gene expression was evaluated by RT-PCR, western blotting and immunohistochemistry. The transwell system was used to assess the invasiveness of HCC cells. Finally, we established a mouse model for HCC using N-nitrosodiethylamine (DEN) to determine the effect of GdCl3 on HCC. Liver tissue array analysis revealed that CD206 was highly expressed in the HCC tissues compared to the level in peri-carcinoma tissue. We found that GdCl3 suppressed the expression of CD206 in the M2 macrophage phenotype of stimulated RAW264.7 cells with an IC10 value of 0.07 µg/µl. In addition, GdCl3 also induced cell apoptosis in the RAW264.7 cells. Addition of GdCl3 into the culture medium of RAW264.7 cells markedly reduced the invasive ability of Hepa1-6 cells compared to the control cells. Accordingly, GdCl3 treatment increased the expression of the epithelial-mesenchymal transition (EMT)-related protein E-cadherin while expression of N-cadherin, TWIST and Snail was reduced in IL-4-stimulated cells. Moreover, GdCl3 treatment inhibited HCC progression in DEN-induced HCC mice, possibly by downregulating CD206. Our findings indicate that CD206 is a potential biomarker for predicting HCC prognosis and that GdCl3 suppresses HCC progression by downregulating the expression of CD206 in TAMs.
Collapse
Affiliation(s)
- Fangyu Zhu
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiangnan Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yong Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Haoran Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Haolong Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chengyao Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yu Zhao
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Fang Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
168
|
CHO INHYE, JANG EUNHYANG, HONG DARONG, JUNG BOM, PARK MINJU, KIM JONGHO. Suppression of LPS-induced epithelial-mesenchymal transition by aqueous extracts of Prunella vulgaris through inhibition of the NF-κB/Snail signaling pathway and regulation of EMT-related protein expression. Oncol Rep 2015; 34:2445-50. [DOI: 10.3892/or.2015.4218] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/17/2015] [Indexed: 11/06/2022] Open
|
169
|
Zhi Y, Mou Z, Chen J, He Y, Dong H, Fu X, Wu Y. B7H1 Expression and Epithelial-To-Mesenchymal Transition Phenotypes on Colorectal Cancer Stem-Like Cells. PLoS One 2015; 10:e0135528. [PMID: 26284927 PMCID: PMC4540313 DOI: 10.1371/journal.pone.0135528] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/22/2015] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) can invade and metastasize by epithelial-to-mesenchymal transition (EMT). However, how they escape immune surveillance is unclear. B7H1 is crucial negative co-stimulatory molecule but little information about whether it works in CSCs. Therefore, we determined the expression of B7H1 and EMT-associated markers in colorectal cancer stem-like cells to investigate a possible immunoevasion way of CSCs. We enriched CD133+ colorectal cancer cells which manifested the CSCs-like properties such as higher levels of other stem cell markers Oct-4 and Sox-2, tumor sphere forming ability and more tumorigenic in NOD/SCID mice. These CD133+ cells possess EMT gene expression profile including higher level of Snail, Twist, vimentin, fibronectin and lower level of E-cadherin. Moreover, CD133+ cells in both cell line and colorectal cancer tissues expressed high level of negative co-stimulate molecule B7H1. Furthermore, some B7H1+ cancer cells also showed the characteristic of EMT, indicating EMT cells could escape immune attack during metastasis. B7H1 expression and EMT phenotypes on CSCs indicates a possible immunoevasion way.
Collapse
Affiliation(s)
- Yidan Zhi
- Institute of Immunology of PLA, Third Military Medical University, Chongqing, China
| | - Zhirong Mou
- Institute of Immunology of PLA, Third Military Medical University, Chongqing, China
- * E-mail: (YW); (ZM)
| | - Jun Chen
- Department of General surgery, Southwest Hospital, Chongqing, China
| | - Yujun He
- Department of General surgery, Daping Hospital, Chongqing, China
| | - Hui Dong
- Institute of Immunology of PLA, Third Military Medical University, Chongqing, China
| | - Xiaolan Fu
- Institute of Immunology of PLA, Third Military Medical University, Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology of PLA, Third Military Medical University, Chongqing, China
- * E-mail: (YW); (ZM)
| |
Collapse
|
170
|
Chung S, Dwabe S, Elshimali Y, Sukhija H, Aroh C, Vadgama JV. Identification of Novel Biomarkers for Metastatic Colorectal Cancer Using Angiogenesis-Antibody Array and Intracellular Signaling Array. PLoS One 2015; 10:e0134948. [PMID: 26258407 PMCID: PMC4530953 DOI: 10.1371/journal.pone.0134948] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/15/2015] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is one of the three leading causes for cancer mortality. CRC kills over 600,000 people annually worldwide. The most common cause of death from CRC is the metastasis to distant organs. However, biomarkers for CRC metastasis remain ill-defined. We compared primary and metastatic CRC cell lines for their angiogenesis-protein profiles and intracellular signaling profiles to identify novel biomarkers for CRC metastasis. To this end, we used primary and metastatic CRC cell lines as a model system and normal human colon cell line as a control. The angiogenesis profiles two isogenic CRC cell lines, SW480 and SW620, and HT-29 and T84 revealed that VEGF was upregulated in both SW620 and T84 whereas coagulation factor III, IGFBP-3, DPP IV, PDGF AA/AB, endothelin I and CXCL16 were downregulated specifically in metastatic cell lines. Furthermore, we found that TIMP-1, amphiregulin, endostatin, angiogenin were upregulated in SW620 whereas downregulated in T84. Angiogenin was downregulated in T84 and GM-CSF was also downregulated in SW620. To induce CRC cell metastasis, we treated cells with pro-inflammatory cytokine IL-6. Upon IL-6 treatment, epithelial-mesenchymal transition was induced in CRC cells. When DLD-1 and HT-29 cells were treated with IL-6; Akt, STAT3, AMPKα and Bad phosphorylation levels were increased. Interestingly, SW620 showed the same signal activation pattern with IL-6 treatment of HT-29 and DLD-1. Our data suggest that Akt, STAT3, AMPKα and Bad activation can be biomarkers for metastatic colorectal cancer. IL-6 treatment specifically reduced phosphorylation levels of EGFR, HER2 receptor, Insulin R and IGF-1R in receptor tyrosine kinase array study with HT-29. Taken together, we have identified novel biomarkers for metastatic CRC through the angiogenesis-antibody array and intracellular signaling array studies. Present study suggests that those novel biomarkers can be used as CRC prognosis biomarkers, and as potential targets for the metastatic CRC therapy.
Collapse
Affiliation(s)
- Seyung Chung
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 120 street, Los Angeles, California, 90059, United States of America
| | - Sami Dwabe
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 120 street, Los Angeles, California, 90059, United States of America
| | - Yayha Elshimali
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 120 street, Los Angeles, California, 90059, United States of America
| | - Hemlata Sukhija
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 120 street, Los Angeles, California, 90059, United States of America
| | - Clement Aroh
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 120 street, Los Angeles, California, 90059, United States of America
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 120 street, Los Angeles, California, 90059, United States of America
- David Geffen UCLA School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
171
|
Ha NH, Woo BH, Kim DJ, Ha ES, Choi JI, Kim SJ, Park BS, Lee JH, Park HR. Prolonged and repetitive exposure to Porphyromonas gingivalis increases aggressiveness of oral cancer cells by promoting acquisition of cancer stem cell properties. Tumour Biol 2015; 36:9947-60. [PMID: 26178482 DOI: 10.1007/s13277-015-3764-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/03/2015] [Indexed: 02/06/2023] Open
Abstract
Periodontitis is the most common chronic inflammatory condition occurring in the human oral cavity, but our knowledge on its contribution to oral cancer is rather limited. To define crosstalk between chronic periodontitis and oral cancer, we investigated whether Porphyromonas gingivalis, a major pathogen of chronic periodontitis, plays a role in oral cancer progression. To mimic chronic irritation by P. gingivalis in the oral cavity, oral squamous cell carcinoma (OSCC) cells were infected with P. gingivalis twice a week for 5 weeks. Repeated infection of oral cancer cells by P. gingivalis resulted in morphological changes of host cancer cells into an elongated shape, along with the decreased expression of epithelial cell markers, suggesting acquisition of an epithelial-to-mesenchymal transition (EMT) phenotype. The prolonged exposure to P. gingivalis also promoted migratory and invasive properties of OSCC cells and provided resistance against a chemotherapeutic agent, all of which are described as cellular characteristics undergoing EMT. Importantly, long-term infection by P. gingivalis induced an increase in the expression level of CD44 and CD133, well-known cancer stem cell markers, and promoted the tumorigenic properties of infected cancer cells compared to non-infected controls. Furthermore, increased invasiveness of P. gingivalis-infected OSCC cells was correlated with enhanced production of matrix metalloproteinase (MMP)-1 and MMP-10 that was stimulated by interleukin-8 (IL-8) release. This is the first report demonstrating that P. gingivalis can increase the aggressiveness of oral cancer cells via epithelial-mesenchymal transition-like changes and the acquisition of stemness, implicating P. gingivalis as a potential bacterial risk modifier.
Collapse
Affiliation(s)
- Na Hee Ha
- Department of Oral Pathology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan-Si, Kyeongsangnam-Do, 626-870, South Korea
| | - Bok Hee Woo
- Department of Oral Pathology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan-Si, Kyeongsangnam-Do, 626-870, South Korea
| | - Da Jeong Kim
- Department of Oral Pathology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan-Si, Kyeongsangnam-Do, 626-870, South Korea
| | - Eun Sin Ha
- Department of Oral Pathology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan-Si, Kyeongsangnam-Do, 626-870, South Korea
| | - Jeom Il Choi
- Department of Periodontology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan, 626-870, South Korea
| | - Sung Jo Kim
- Department of Periodontology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan, 626-870, South Korea
| | - Bong Soo Park
- Department of Oral Anatomy, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan, 626-870, South Korea
| | - Ji Hye Lee
- Department of Oral Pathology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan-Si, Kyeongsangnam-Do, 626-870, South Korea.,Institute of Translational Dental Sciences, Pusan National University, 49 Busandaehak-Ro, Yangsan, 626-870, South Korea
| | - Hae Ryoun Park
- Department of Oral Pathology, School of Dentistry, Pusan National University, 49 Busandaehak-Ro, Yangsan-Si, Kyeongsangnam-Do, 626-870, South Korea. .,Institute of Translational Dental Sciences, Pusan National University, 49 Busandaehak-Ro, Yangsan, 626-870, South Korea.
| |
Collapse
|
172
|
Extracellular vesicles from women with breast cancer promote an epithelial-mesenchymal transition-like process in mammary epithelial cells MCF10A. Tumour Biol 2015; 36:9649-59. [PMID: 26150337 DOI: 10.1007/s13277-015-3711-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/23/2015] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) mediate many stages of tumor progression including angiogenesis, escape from immune surveillance, and extracellular matrix degradation. We studied whether EVs from plasma of women with breast cancer are able to induce an epithelial-mesenchymal transition (EMT) process in mammary epithelial cells MCF10A. Our findings demonstrate that EVs from plasma of breast cancer patients induce a downregulation of E-cadherin expression and an increase of vimentin and N-cadherin expression. Moreover, EVs induce migration and invasion, as well as an increase of NFκB-DNA binding activity and MMP-2 and MMP-9 secretions. In summary, our findings demonstrate, for the first time, that EVs from breast cancer patients induce an EMT-like process in human mammary non-tumorigenic epithelial cells MCF10A.
Collapse
|
173
|
Finlay J, Roberts CM, Dong J, Zink JI, Tamanoi F, Glackin CA. Mesoporous silica nanoparticle delivery of chemically modified siRNA against TWIST1 leads to reduced tumor burden. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1657-66. [PMID: 26115637 DOI: 10.1016/j.nano.2015.05.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/18/2015] [Accepted: 05/30/2015] [Indexed: 02/07/2023]
Abstract
UNLABELLED Growth and progression of solid tumors depend on the integration of multiple pro-growth and survival signals, including the induction of angiogenesis. TWIST1 is a transcription factor whose reactivation in tumors leads to epithelial to mesenchymal transition (EMT), including increased cancer cell stemness, survival, and invasiveness. Additionally, TWIST1 drives angiogenesis via activation of IL-8 and CCL2, independent of VEGF signaling. In this work, results suggest that chemically modified siRNA against TWIST1 reverses EMT both in vitro and in vivo. siRNA delivery with a polyethyleneimine-coated mesoporous silica nanoparticle (MSN) led to reduction of TWIST1 target genes and migratory potential in vitro. In mice bearing xenograft tumors, weekly intravenous injections of the siRNA-nanoparticle complexes resulted in decreased tumor burden together with a loss of CCL2 suggesting a possible anti-angiogenic response. Therapeutic use of TWIST1 siRNA delivered via MSNs has the potential to inhibit tumor growth and progression in many solid tumor types. FROM THE CLINICAL EDITOR Tumor progression and metastasis eventually lead to patient mortality in the clinical setting. In other studies, it has been found that TWIST1, a transcription factor, if reactivated in tumors, would lead to downstream events including angiogenesis and result in poor prognosis in cancer patients. In this article, the authors were able to show that when siRNA against TWIST1 was delivered via mesoporous silica nanoparticle, there was tumor reduction in an in-vivo model. The results have opened up a new avenue for further research in this field.
Collapse
Affiliation(s)
- James Finlay
- Division of Comparative Medicine and, Irell & Manella Graduate School of Biological Sciences, City of Hope, Beckman Research Institute, Duarte, CA, USA.
| | - Cai M Roberts
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Beckman Research Institute, Duarte, CA, USA.
| | - Juyao Dong
- Department of Chemistry and Biochemistry, Jonsson Comprehensive Cancer Center, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA.
| | - Jeffrey I Zink
- Department of Chemistry and Biochemistry, Jonsson Comprehensive Cancer Center, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA.
| | - Fuyuhiko Tamanoi
- Department of Microbiology Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center, California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA.
| | - Carlotta A Glackin
- Department of Neurosciences, City of Hope, Beckman Research Institute, Duarte, CA, USA.
| |
Collapse
|
174
|
Zhao M, Kong L, Liu Y, Qu H. dbEMT: an epithelial-mesenchymal transition associated gene resource. Sci Rep 2015; 5:11459. [PMID: 26099468 PMCID: PMC4477208 DOI: 10.1038/srep11459] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 04/28/2015] [Indexed: 12/16/2022] Open
Abstract
As a cellular process that changes epithelial cells to mesenchymal cells, Epithelial-mesenchymal transition (EMT) plays important roles in development and cancer metastasis. Recent studies on cancer metastasis have identified many new susceptibility genes that control this transition. However, there is no comprehensive resource for EMT by integrating various genetic studies and the relationship between EMT and the risk of complex diseases such as cancer are still unclear. To investigate the cellular complexity of EMT, we have constructed dbEMT (http://dbemt.bioinfo-minzhao.org/), the first literature-based gene resource for exploring EMT-related human genes. We manually curated 377 experimentally verified genes from literature. Functional analyses highlighted the prominent role of proteoglycans in tumor metastatic cascades. In addition, the disease enrichment analysis provides a clue for the potential transformation in affected tissues or cells in Alzheimer's disease and Type 2 Diabetes. Moreover, the global mutation pattern of EMT-related genes across multiple cancers may reveal common cancer metastasis mechanisms. Our further reconstruction of the EMT-related protein-protein interaction network uncovered a highly modular structure. These results illustrate the importance of dbEMT to our understanding of cell development and cancer metastasis, and also highlight the utility of dbEMT for elucidating the functions of EMT-related genes.
Collapse
Affiliation(s)
- Min Zhao
- School of Engineering, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore DC, Queensland, 4558, Australia
| | - Lei Kong
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, P.R. China
| | - Yining Liu
- School of Engineering, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore DC, Queensland, 4558, Australia
| | - Hong Qu
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, P.R. China
| |
Collapse
|
175
|
Tan TZ, Miow QH, Miki Y, Noda T, Mori S, Huang RYJ, Thiery JP. Epithelial-mesenchymal transition spectrum quantification and its efficacy in deciphering survival and drug responses of cancer patients. EMBO Mol Med 2015; 6:1279-93. [PMID: 25214461 PMCID: PMC4287932 DOI: 10.15252/emmm.201404208] [Citation(s) in RCA: 534] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a reversible and dynamic process hypothesized to be co-opted by carcinoma during invasion and metastasis. Yet, there is still no quantitative measure to assess the interplay between EMT and cancer progression. Here, we derived a method for universal EMT scoring from cancer-specific transcriptomic EMT signatures of ovarian, breast, bladder, lung, colorectal and gastric cancers. We show that EMT scoring exhibits good correlation with previously published, cancer-specific EMT signatures. This universal and quantitative EMT scoring was used to establish an EMT spectrum across various cancers, with good correlation noted between cell lines and tumours. We show correlations between EMT and poorer disease-free survival in ovarian and colorectal, but not breast, carcinomas, despite previous notions. Importantly, we found distinct responses between epithelial- and mesenchymal-like ovarian cancers to therapeutic regimes administered with or without paclitaxelin vivo and demonstrated that mesenchymal-like tumours do not always show resistance to chemotherapy. EMT scoring is thus a promising, versatile tool for the objective and systematic investigation of EMT roles and dynamics in cancer progression, treatment response and survival.
Collapse
Affiliation(s)
- Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Qing Hao Miow
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Yoshio Miki
- Cancer Institute of Japanese Foundation for Cancer Research, Kyoto, Japan
| | - Tetsuo Noda
- Cancer Institute of Japanese Foundation for Cancer Research, Kyoto, Japan
| | - Seiichi Mori
- Cancer Institute of Japanese Foundation for Cancer Research, Kyoto, Japan
| | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore Department of Obstetrics and Gynaecology, National University Health System, Singapore
| | - Jean Paul Thiery
- Cancer Science Institute of Singapore, National University of Singapore, Singapore Institute of Molecular and Cell Biology, A*STAR, Singapore Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
176
|
He E, Pan F, Li G, Li J. Fractionated Ionizing Radiation Promotes Epithelial-Mesenchymal Transition in Human Esophageal Cancer Cells through PTEN Deficiency-Mediated Akt Activation. PLoS One 2015; 10:e0126149. [PMID: 26000878 PMCID: PMC4441389 DOI: 10.1371/journal.pone.0126149] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/29/2015] [Indexed: 01/10/2023] Open
Abstract
In some esophageal cancer patients, radiotherapy may not prevent distant metastasis thus resulting in poor survival. The underlying mechanism of metastasis in these patients is not well established. In this study, we have demonstrated that ionizing radiation may induce epithelial-mesenchymal transition (EMT) accompanied with increased cell migration and invasion, through downregulation of phosphatase and tensin homolog (PTEN), and activation of Akt/GSK-3β/Snail signaling. We developed a radioresistant (RR) esophageal squamous cancer cell line, KYSE-150/RR, by fractionated ionizing radiation (IR) treatment, and confirmed its radioresistance using a clonogenic survival assay. We found that the KYSE-150/RR cell line displayed typical morphological and molecular characteristics of EMT. In comparison to the parental cells, KYSE-150/RR cells showed an increase in post-IR colony survival, migration, and invasiveness. Furthermore, a decrease in PTEN in KYSE-150/RR cells was observed. We postulated that over-expression of PTEN may induce mesenchymal-epithelial transition in KYSE-150/RR cells and restore IR-induced increase of cell migration. Mechanistically, fractionated IR inhibits expression of PTEN, which leads to activation of Akt/GSK-3β signaling and is associated with the elevated levels of Snail protein, a transcription factor involved in EMT. Correspondingly, treatment with LY294002, a phosphatidylinositol-3-kinase inhibitor, mimicked PTEN overexpression effect in KYSE-150/RR cells, further suggesting a role for the Akt/GSK-3β/Snail signaling in effects mediated through PTEN. Together, these results strongly suggest that fractionated IR-mediated EMT in KYSE-150/RR cells is through PTEN-dependent pathways, highlighting a direct proinvasive effect of radiation treatment on tumor cells.
Collapse
Affiliation(s)
- Enhui He
- Nankai University School of Medicine, Tianjin, China
- Chinese PLA General Hospital and Chinese PLA Medical School, Beijing, China
- Beijing Friendship Hospital, affiliated with Capital Medical University, Beijing, China
| | - Fei Pan
- Nankai University School of Medicine, Tianjin, China
| | - Guangchao Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Jingjing Li
- Chinese PLA General Hospital and Chinese PLA Medical School, Beijing, China
- Beijing Friendship Hospital, affiliated with Capital Medical University, Beijing, China
- * E-mail:
| |
Collapse
|
177
|
Yang Y, Otte A, Hass R. Human mesenchymal stroma/stem cells exchange membrane proteins and alter functionality during interaction with different tumor cell lines. Stem Cells Dev 2015; 24:1205-1222. [PMID: 25525832 PMCID: PMC4425222 DOI: 10.1089/scd.2014.0413] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/19/2014] [Indexed: 12/22/2022] Open
Abstract
To analyze effects of cellular interaction between human mesenchymal stroma/stem cells (MSC) and different cancer cells, direct co-cultures were performed and revealed significant growth stimulation of the tumor populations and a variety of protein exchanges. More than 90% of MCF-7 and primary human HBCEC699 breast cancer cells as well as NIH:OVCAR-3 ovarian adenocarcinoma cells acquired CD90 proteins during MSC co-culture, respectively. Furthermore, SK-OV-3 ovarian cancer cells progressively elevated CD105 and CD90 proteins in co-culture with MSC. Primary small cell hypercalcemic ovarian carcinoma cells (SCCOHT-1) demonstrated undetectable levels of CD73 and CD105; however, both proteins were significantly increased in the presence of MSC. This co-culture-mediated protein induction was also observed at transcriptional levels and changed functionality of SCCOHT-1 cells by an acquired capability to metabolize 5'cAMP. Moreover, exchange between tumor cells and MSC worked bidirectional, as undetectable expression of epithelial cell adhesion molecule (EpCAM) in MSC significantly increased after co-culture with SK-OV-3 or NIH:OVCAR-3 cells. In addition, a small population of chimeric/hybrid cells appeared in each MSC/tumor cell co-culture by spontaneous cell fusion. Immune fluorescence demonstrated nanotube structures and exosomes between MSC and tumor cells, whereas cytochalasin-D partially abolished the intercellular protein transfer. More detailed functional analysis of FACS-separated MSC and NIH:OVCAR-3 cells after co-culture revealed the acquisition of epithelial cell-specific properties by MSC, including increased gene expression for cytokeratins and epithelial-like differentiation factors. Vice versa, a variety of transcriptional regulatory genes were down-modulated in NIH:OVCAR-3 cells after co-culture with MSC. Together, these mutual cellular interactions contributed to functional alterations in MSC and tumor cells.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
- Tongji Hospital Affiliated by Tongji University, Shanghai, China
| | - Anna Otte
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
178
|
Ran J, Lin DL, Wu RF, Chen QH, Huang HP, Qiu NX, Quan S. ZEB1 promotes epithelial-mesenchymal transition in cervical cancer metastasis. Fertil Steril 2015; 103:1606-14.e1-2. [PMID: 25963537 DOI: 10.1016/j.fertnstert.2015.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/02/2015] [Accepted: 03/17/2015] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate role of Zinc finger E-box binding homeobox 1 (ZEB1) in cervical cancer tissue (squamous cell carcinoma, SCC). DESIGN Exploratory study. SETTING University hospital. PATIENT(S) Sixty patients with SCC, including stage CINIII (n = 10), IB1 (n = 10), IB2 (n = 10), IIA1 (n = 10), IIA2 (n = 10), and IIB (n = 10) were studied. INTERVENTION(S) Caski cells were transfected with recombinant shZEB1 lentivirus or shCtrl lentivirus to generate stable ZEB1-knockdown Caski cells. MAIN OUTCOME MEASURE(S) ZEB1 expression was analyzed by quantitative real-time polymerase chain reaction and immunohistochemistry in cervical cancer tissues. ZEB1 expression in Caski cells was down-regulated by short-hairpin RNA (shRNA) interference, and changes in ZEB1 expression corresponded with changes in the proliferation and migratory ability of Caski cells. RESULT(S) Quantitative real-time polymerase chain reaction and immunohistochemistry results revealed that ZEB1 expression and the ratio of Vimentin to E-cadherin were high in 27 of 50 SCC patients and correlated with advanced International Federation of Gynecology and Obstetrics stage, tumor size >4 cm, and parametrial invasion. However, the expression of ZEB1 in cervical cancer tissue was independent of age and SCC antigen level. Transfection of ZEB1 shRNA in Caski cells significantly decreased the messenger RNA and protein expression of ZEB1, parallel with increased expression of the epithelial marker E-cadherin and decreased expression of the mesenchymal marker Vimentin. Furthermore, the proliferation and migratory ability of Caski cells were significantly lower in the transfected group than in the nontransfected control group. CONCLUSION(S) Down-regulation of ZEB1 expression may protect the invasive front of the tumors from converting to a mesenchymal phenotype by reducing the proliferation and motility of cervical cancer cells, suggesting that ZEB1 might be a potential therapeutic target for SCC.
Collapse
Affiliation(s)
- Jing Ran
- Reproductive Medicine Center, Nan Fang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Gynecology and Obstetrics, First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Dian-Liang Lin
- Reproductive Medicine Center, Nan Fang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Rong-Feng Wu
- State Key Laboratory of Cellular Stress Biology and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Qiong-Hua Chen
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Hui-Ping Huang
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Na-Xuan Qiu
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Song Quan
- Reproductive Medicine Center, Nan Fang Hospital, Southern Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
179
|
Dandawate P, Padhye S, Ahmad A, Sarkar FH. Novel strategies targeting cancer stem cells through phytochemicals and their analogs. Drug Deliv Transl Res 2015; 3:165-82. [PMID: 24076568 DOI: 10.1007/s13346-012-0079-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer stem cells (CSCs) are cells that exist within a tumor with a capacity of self-renewal and an ability to differentiate, giving rise to heterogeneous populations of cancer cells. These cells are increasingly being implicated in resistance to conventional therapeutics and have also been implicated in tumor recurrence. Several cellular signaling pathways including Notch, Wnt, phosphoinositide-3-kinase-Akt-mammalian target of rapamycin pathways, and known markers such as CD44, CD133, CD166, ALDH, etc. have been associated with CSCs. Here, we have reviewed our current understanding of self-renewal pathways and factors that help in the survival of CSCs with special emphasis on those that have been documented to be modulated by well characterized natural agents such as curcumin, sulforaphane, resveratrol, genistein, and epigallocatechin gallate. With the inclusion of a novel derivative of curcumin, CDF, we showcase how natural agents can be effectively modified to increase their efficacy, particularly against CSCs. We hope that this article will generate interest among researchers for further mechanistic and clinical studies exploiting the cancer preventive and therapeutic role of nutraceuticals by targeted elimination of CSCs.
Collapse
Affiliation(s)
- Prasad Dandawate
- ISTRA, Department of Chemistry, Abeda Inamdar Senior College, University of Pune, Pune 411001, India
| | | | | | | |
Collapse
|
180
|
Orellana R, Kato S, Erices R, Bravo ML, Gonzalez P, Oliva B, Cubillos S, Valdivia A, Ibañez C, Brañes J, Barriga MI, Bravo E, Alonso C, Bustamente E, Castellon E, Hidalgo P, Trigo C, Panes O, Pereira J, Mezzano D, Cuello MA, Owen GI. Platelets enhance tissue factor protein and metastasis initiating cell markers, and act as chemoattractants increasing the migration of ovarian cancer cells. BMC Cancer 2015; 15:290. [PMID: 25886038 PMCID: PMC4410584 DOI: 10.1186/s12885-015-1304-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/31/2015] [Indexed: 12/17/2022] Open
Abstract
Background An increase in circulating platelets, or thrombocytosis, is recognized as an independent risk factor of bad prognosis and metastasis in patients with ovarian cancer; however the complex role of platelets in tumor progression has not been fully elucidated. Platelet activation has been associated with an epithelial to mesenchymal transition (EMT), while Tissue Factor (TF) protein expression by cancer cells has been shown to correlate with hypercoagulable state and metastasis. The aim of this work was to determine the effect of platelet-cancer cell interaction on TF and “Metastasis Initiating Cell (MIC)” marker levels and migration in ovarian cancer cell lines and cancer cells isolated from the ascetic fluid of ovarian cancer patients. Methods With informed patient consent, ascitic fluid isolated ovarian cancer cells, cell lines and ovarian cancer spheres were co-cultivated with human platelets. TF, EMT and stem cell marker levels were determined by Western blotting, flow cytometry and RT-PCR. Cancer cell migration was determined by Boyden chambers and the scratch assay. Results The co-culture of patient-derived ovarian cancer cells with platelets causes: 1) a phenotypic change in cancer cells, 2) chemoattraction and cancer cell migration, 3) induced MIC markers (EMT/stemness), 3) increased sphere formation and 4) increased TF protein levels and activity. Conclusions We present the first evidence that platelets act as chemoattractants to cancer cells. Furthermore, platelets promote the formation of ovarian cancer spheres that express MIC markers and the metastatic protein TF. Our results suggest that platelet-cancer cell interaction plays a role in the formation of metastatic foci. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1304-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Renan Orellana
- Departament of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| | - Sumie Kato
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| | - Rafaela Erices
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| | - María Loreto Bravo
- Departament of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| | - Pamela Gonzalez
- Departament of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| | - Bárbara Oliva
- Departament of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| | - Sofía Cubillos
- Departament of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Andrés Valdivia
- Departament of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Carolina Ibañez
- Division de Hematology & Oncology, Faculty of Medicine, Santiago, Chile. .,Center UC Investigation in Oncology, Santiago, Chile.
| | - Jorge Brañes
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | - Erasmo Bravo
- Hospital Gustavo Fricke, Viña de Mar, Santiago, Chile.
| | | | - Eva Bustamente
- Fundación Arturo López Pérez, Av. Rancagua 878, Providencia, Santiago, Chile.
| | - Enrique Castellon
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile Avda, Independencia 1027, Santiago, Chile.
| | - Patricia Hidalgo
- Division de Hematology & Oncology, Faculty of Medicine, Santiago, Chile.
| | - Cesar Trigo
- Division de Hematology & Oncology, Faculty of Medicine, Santiago, Chile.
| | - Olga Panes
- Division de Hematology & Oncology, Faculty of Medicine, Santiago, Chile.
| | - Jaime Pereira
- Division de Hematology & Oncology, Faculty of Medicine, Santiago, Chile.
| | - Diego Mezzano
- Division de Hematology & Oncology, Faculty of Medicine, Santiago, Chile. .,Center UC Investigation in Oncology, Santiago, Chile.
| | - Mauricio A Cuello
- Department of Obstetrics and Gynecology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Center UC Investigation in Oncology, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| | - Gareth I Owen
- Departament of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Center UC Investigation in Oncology, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile. .,Biomedical Research Consortium of Chile, Alameda 440, Piso 13, Santiago, Chile.
| |
Collapse
|
181
|
Zhu J, Yang DR, Sun Y, Qiu X, Chang HC, Li G, Shan Y, Chang C. TR4 Nuclear Receptor Alters the Prostate Cancer CD133+ Stem/Progenitor Cell Invasion via Modulating the EZH2-Related Metastasis Gene Expression. Mol Cancer Ther 2015; 14:1445-53. [PMID: 25833838 DOI: 10.1158/1535-7163.mct-14-0971] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/23/2015] [Indexed: 11/16/2022]
Abstract
The testicular nuclear receptor 4 (TR4) is a member of the nuclear receptor superfamily that mediates various biologic functions with key impacts on metabolic disorders and tumor progression. Here, we demonstrate that TR4 may play a positive role in prostate cancer CD133(+) stem/progenitor (S/P) cell invasion. Targeting TR4 with lentiviral silencing RNA significantly suppressed prostate cancer CD133(+) S/P cell invasion both in vitro and in vivo. Mechanism dissection found that TR4 transcriptionally regulates the oncogene EZH2 via binding to its 5' promoter region. The consequences of targeting TR4 to suppress EZH2 expression may then suppress the expression of its downstream key metastasis-related genes, including NOTCH1, TGFβ1, SLUG, and MMP9. Rescue approaches via adding the EZH2 reversed the TR4-mediated prostate cancer S/P cell invasion. Together, these results suggest that the TR4→EZH2 signaling may play a critical role in the prostate cancer S/P cell invasion and may allow us to develop a better therapy to battle the prostate cancer metastasis.
Collapse
Affiliation(s)
- Jin Zhu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China. George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Dong-Rong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China. George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Xiaofu Qiu
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York. Department of Urology, Guangdong No. 2 Provincial People's Hospital, Guangzhou, China
| | - Hong-Chiang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York. Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Gonghui Li
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Yuxi Shan
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China. George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York.
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York. Sex Hormone Research Center, China Medical University/Hospital, Taichung, Taiwan.
| |
Collapse
|
182
|
Neagu M, Constantin C, Dumitrascu GR, Lupu AR, Caruntu C, Boda D, Zurac S. Inflammation markers in cutaneous melanoma - edgy biomarkers for prognosis. Discoveries (Craiova) 2015; 3:e38. [PMID: 32309563 PMCID: PMC6941591 DOI: 10.15190/d.2015.30] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
There is a fine balance between inflammation and tumorigenesis. While environmentally induced inflammatory condition can precede a malignant transformation, in other cases an oncogenic change of unknown origin can induce an inflammatory microenvironment that promotes the development of tumors. Regardless of its origin, maintaining the inflammation milieu has many tumor-promoting effects. As a result, inflammation can aid the proliferation and survival of malignant cells, can promote angiogenesis and metastasis, can down-regulate innate/adaptive immune responses, and can alter responses to hormones and chemotherapeutic agents. There is an abundance of studies unveiling molecular pathways of cancer-related inflammation; this wealth of information brings new insights into biomarkers domain in the diagnosis and treatment improvement pursue.
In cutaneous tissue there is an established link between tissue damage, inflammation, and cancer development. Inflammation is a self-limiting process in normal healthy physiological conditions, while tumorigenesis is a complex mechanism of constitutive pathway activation. Once more, in cutaneous melanoma, there is an unmet need for inflammatory biomarkers that could improve prognostication. Targeting inflammation and coping with the phenotypic plasticity of melanoma cells represent rational strategies to specifically interfere with metastatic progression. We have shown that there is a prototype of intratumor inflammatory infiltrate depicting a good prognosis, infiltrate that is composed of numerous T cells CD3+, Langerhans cells, few/absent B cells CD20+ and few/absent plasma cells. Circulating immune cells characterized by phenotype particularities are delicately linked to the stage melanoma is diagnosed in. Hence circulatory immune sub-populations, with activated or suppressor phenotype would give the physician a more detailed immune status of the patient. A panel of tissue/circulatory immune markers can complete the immune status, can add value to the overall prognostic of the patient and, as a result direct/redirect the therapy choice. The future lies within establishing low-cost, affordable/available, easily reproducible assays that will complete the pre-clinical parameters of the patient.
Collapse
Affiliation(s)
- Monica Neagu
- Immunobiology Laboratory, "Victor Babes" National Institute of Pathology and Biomedical Sciences, Bucharest, Romania.,Faculty of Biochemistry, University of Bucharest, Romania
| | - Carolina Constantin
- Immunobiology Laboratory, "Victor Babes" National Institute of Pathology and Biomedical Sciences, Bucharest, Romania
| | - Georgiana Roxana Dumitrascu
- Immunobiology Laboratory, "Victor Babes" National Institute of Pathology and Biomedical Sciences, Bucharest, Romania
| | - Andreea Roxana Lupu
- Immunobiology Laboratory, "Victor Babes" National Institute of Pathology and Biomedical Sciences, Bucharest, Romania
| | - Constantin Caruntu
- Immunobiology Laboratory, "Victor Babes" National Institute of Pathology and Biomedical Sciences, Bucharest, Romania.,Dermatology Research Laboratory, "Carol Davila" University of Medicine & Pharmacy, Bucharest, Romania
| | - Daniel Boda
- Dermatology Research Laboratory, "Carol Davila" University of Medicine & Pharmacy, Bucharest, Romania
| | - Sabina Zurac
- Department of Pathology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Colentina University Hospital, Bucharest, Romania
| |
Collapse
|
183
|
Caceres S, Peña L, de Andres PJ, Illera MJ, Lopez MS, Woodward WA, Reuben JM, Illera JC. Establishment and characterization of a new cell line of canine inflammatory mammary cancer: IPC-366. PLoS One 2015; 10:e0122277. [PMID: 25807360 PMCID: PMC4373858 DOI: 10.1371/journal.pone.0122277] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 02/17/2015] [Indexed: 11/19/2022] Open
Abstract
Canine inflammatory mammary cancer (IMC) shares epidemiologic, histopathological and clinical characteristics with the disease in humans and has been proposed as a natural model for human inflammatory breast cancer (IBC). The aim of this study was to characterize a new cell line from IMC (IPC-366) for the comparative study of both IMC and IBC. Tumors cells from a female dog with clinical IMC were collected. The cells were grown under adherent conditions. The growth, cytological, ultrastructural and immunohistochemical (IHC) characteristics of IPC-366 were evaluated. Ten female Balb/SCID mice were inoculated with IPC-366 cells to assess their tumorigenicity and metastatic potential. Chromosome aberration test and Karyotype revealed the presence of structural aberration, numerical and neutral rearrangements, demonstrating a chromosomal instability. Microscopic examination of tumor revealed an epithelial morphology with marked anysocytosis. Cytological and histological examination of smears and ultrathin sections by electron microscopy revealed that IPC-366 is formed by highly malignant large round or polygonal cells characterized by marked atypia and prominent nucleoli and frequent multinucleated cells. Some cells had cytoplasmic empty spaces covered by cytoplasmic membrane resembling capillary endothelial cells, a phenomenon that has been related to s vasculogenic mimicry. IHC characterization of IPC-366 was basal-like: epithelial cells (AE1/AE3+, CK14+, vimentin+, actin-, p63-, ER-, PR-, HER-2, E-cadherin, overexpressed COX-2 and high Ki-67 proliferation index (87.15 %). At 2 weeks after inoculating the IPC-366 cells, a tumor mass was found in 100 % of mice. At 4 weeks metastases in lung and lymph nodes were found. Xenograph tumors maintained the original IHC characteristics of the female dog tumor. In summary, the cell line IPC-366 is a fast growing malignant triple negative cell line model of inflammatory mammary carcinoma that can be used for the comparative study of both IMC and IBC.
Collapse
Affiliation(s)
- Sara Caceres
- Department of Animal Physiology, Surgery and Pathology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Spain
| | - Laura Peña
- Department of Animal Medicine, Surgery and Pathology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Spain
| | - Paloma J. de Andres
- Department of Animal Medicine, Surgery and Pathology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Spain
| | - Maria J. Illera
- Department of Animal Physiology, Surgery and Pathology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Spain
| | - Mirtha S. Lopez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Wendy A. Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - James M. Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
- * E-mail:
| | - Juan C. Illera
- Department of Animal Physiology, Surgery and Pathology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Spain
| |
Collapse
|
184
|
PIK3R1 negatively regulates the epithelial-mesenchymal transition and stem-like phenotype of renal cancer cells through the AKT/GSK3β/CTNNB1 signaling pathway. Sci Rep 2015; 5:8997. [PMID: 25757764 PMCID: PMC4355729 DOI: 10.1038/srep08997] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/13/2015] [Indexed: 01/03/2023] Open
Abstract
The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway has been identified as an important pathway in renal cell carcinoma (RCC). We have reported a nonsense mutation in PIK3R1, which encodes the regulatory subunit of PI3K, in a metastatic RCC (mRCC), while the mutation was absent in the corresponding primary RCC (pRCC). To identify the function of PIK3R1 in RCC, we examined its expression in normal kidney, pRCC and mRCC by immunohistochemistry and real-time polymerase chain reaction. The expression of PIK3R1 significantly decreased in pRCC and was further reduced in mRCC compared with normal tissue. Besides, its expression levels were negatively correlated with T-category of tumor stage. Additionally, 786-O and A-704 cells with PIK3R1 depletion introduced by CRISPR/Cas9 system displayed enhanced proliferation, migration and epithelial-mesenchymal transition (EMT), and acquired a stem-like phenotype. Moreover, the PIK3R1 depletion promoted the phosphorylation of AKT in the cells. The knockdown of AKT by shRNA reduced p-GSK3β and CTNNB1 expression in the cells, while the depletion of CTNNB1 impaired stem-like phenotype of the cells. Overall, PIK3R1 down-regulation in RCC promotes propagation, migration, EMT and stem-like phenotype in renal cancer cells through the AKT/GSK3β/CTNNB1 pathway, and may contribute to progression and metastasis of RCC.
Collapse
|
185
|
RNA-based TWIST1 inhibition via dendrimer complex to reduce breast cancer cell metastasis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:382745. [PMID: 25759817 PMCID: PMC4339717 DOI: 10.1155/2015/382745] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 12/31/2014] [Accepted: 12/31/2014] [Indexed: 01/11/2023]
Abstract
Breast cancer is the leading cause of cancer-related deaths among women in the United States, and survival rates are lower for patients with metastases and/or triple-negative breast cancer (TNBC; ER, PR, and Her2 negative). Understanding the mechanisms of cancer metastasis is therefore crucial to identify new therapeutic targets and develop novel treatments to improve patient outcomes. A potential target is the TWIST1 transcription factor, which is often overexpressed in aggressive breast cancers and is a master regulator of cellular migration through epithelial-mesenchymal transition (EMT). Here, we demonstrate an siRNA-based TWIST1 silencing approach with delivery using a modified poly(amidoamine) (PAMAM) dendrimer. Our results demonstrate that SUM1315 TNBC cells efficiently take up PAMAM-siRNA complexes, leading to significant knockdown of TWIST1 and EMT-related target genes. Knockdown lasts up to one week after transfection and leads to a reduction in migration and invasion, as determined by wound healing and transwell assays. Furthermore, we demonstrate that PAMAM dendrimers can deliver siRNA to xenograft orthotopic tumors and siRNA remains in the tumor for at least four hours after treatment. These results suggest that further development of dendrimer-based delivery of siRNA for TWIST1 silencing may lead to a valuable adjunctive therapy for patients with TNBC.
Collapse
|
186
|
Pestana D, Teixeira D, Faria A, Domingues V, Monteiro R, Calhau C. Effects of environmental organochlorine pesticides on human breast cancer: putative involvement on invasive cell ability. ENVIRONMENTAL TOXICOLOGY 2015; 30:168-176. [PMID: 23913582 DOI: 10.1002/tox.21882] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 06/02/2023]
Abstract
Human exposure to persistent organic pollutants (POPs) is a certainty, even to long banned pesticides like o,p'-dichlorodiphenyltrichloroethane (o,p'-DDT), and its metabolites p,p'-dichlorodiphenyldichloroethylene (p,p'-DDE), and p,p'-dichlorodiphenyldichloroethane (p,p'-DDD). POPs are known to be particularly toxic and have been associated with endocrine-disrupting effects in several mammals, including humans even at very low doses. As environmental estrogens, they could play a critical role in carcinogenesis, such as in breast cancer. With the purpose of evaluating their effect on breast cancer biology, o,p'-DDT, p,p'-DDE, and p,p'-DDD (50-1000 nM) were tested on two human breast adenocarcinoma cell lines: MCF-7 expressing estrogen receptor (ER) α and MDA-MB-231 negative for ERα, regarding cell proliferation and viability in addition to their invasive potential. Cell proliferation and viability were not equally affected by these compounds. In MCF-7 cells, the compounds were able to decrease cell proliferation and viability. On the other hand, no evident response was observed in treated MDA-MB-231 cells. Concerning the invasive potential, the less invasive cell line, MCF-7, had its invasion potential significantly induced, while the more invasive cell line MDA-MB-231, had its invasion potential dramatically reduced in the presence of the tested compounds. Altogether, the results showed that these compounds were able to modulate several cancer-related processes, namely in breast cancer cell lines, and underline the relevance of POP exposure to the risk of cancer development and progression, unraveling distinct pathways of action of these compounds on tumor cell biology.
Collapse
Affiliation(s)
- Diogo Pestana
- Department of Biochemistry (U38-FCT), Faculty of Medicine, University of Porto, Centro de Investigação Médica, Rua Dr. Plácido da Costa, Porto, Portugal
| | | | | | | | | | | |
Collapse
|
187
|
Zhi X, Lin L, Yang S, Bhuvaneshwar K, Wang H, Gusev Y, Lee MH, Kallakury B, Shivapurkar N, Cahn K, Tian X, Marshall JL, Byers SW, He AR. βII-Spectrin (SPTBN1) suppresses progression of hepatocellular carcinoma and Wnt signaling by regulation of Wnt inhibitor kallistatin. Hepatology 2015; 61:598-612. [PMID: 25307947 PMCID: PMC4327990 DOI: 10.1002/hep.27558] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 10/07/2014] [Indexed: 12/27/2022]
Abstract
UNLABELLED βII-Spectrin (SPTBN1) is an adapter protein for Smad3/Smad4 complex formation during transforming growth factor beta (TGF-β) signal transduction. Forty percent of SPTBN1(+/-) mice spontaneously develop hepatocellular carcinoma (HCC), and most cases of human HCC have significant reductions in SPTBN1 expression. In this study, we investigated the possible mechanisms by which loss of SPTBN1 may contribute to tumorigenesis. Livers of SPTBN1(+/-) mice, compared to wild-type mouse livers, display a significant increase in epithelial cell adhesion molecule-positive (EpCAM(+)) cells and overall EpCAM expression. Inhibition of SPTBN1 in human HCC cell lines increased the expression of stem cell markers EpCAM, Claudin7, and Oct4, as well as decreased E-cadherin expression and increased expression of vimentin and c-Myc, suggesting reversion of these cells to a less differentiated state. HCC cells with decreased SPTBN1 also demonstrate increased sphere formation, xenograft tumor development, and invasion. Here we investigate possible mechanisms by which SPTBN1 may influence the stem cell traits and aggressive behavior of HCC cell lines. We found that HCC cells with decreased SPTBN1 express much less of the Wnt inhibitor kallistatin and exhibit decreased β-catenin phosphorylation and increased β-catenin nuclear localization, indicating Wnt signaling activation. Restoration of kallistatin expression in these cells reversed the observed Wnt activation. CONCLUSION SPTBN1 expression in human HCC tissues is positively correlated with E-cadherin and kallistatin levels, and decreased SPTBN1 and kallistatin gene expression is associated with decreased relapse-free survival. Our data suggest that loss of SPTBN1 activates Wnt signaling, which promotes acquisition of stem cell-like features, and ultimately contributes to malignant tumor progression.
Collapse
Affiliation(s)
- Xiuling Zhi
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
- Laboratory of Medical Molecular Biology, Training Center of Medical Experiments, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ling Lin
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Shaoxian Yang
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Krithika Bhuvaneshwar
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Hongkun Wang
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Yuriy Gusev
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Mi-Hye Lee
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Bhaskar Kallakury
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Narayan Shivapurkar
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Katherine Cahn
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Xuefei Tian
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - John L. Marshall
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Stephen W. Byers
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Aiwu R. He
- Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
- Corresponding author: Aiwu R. He, M.D. Ph.D., Departments of Medicine and Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, NW, Washington, DC 20007, USA., Phone: 02-444-1259, Fax: 202-444-9429,
| |
Collapse
|
188
|
Kong D, Sethi S, Li Y, Chen W, Sakr WA, Heath E, Sarkar FH. Androgen receptor splice variants contribute to prostate cancer aggressiveness through induction of EMT and expression of stem cell marker genes. Prostate 2015; 75:161-74. [PMID: 25307492 PMCID: PMC4270852 DOI: 10.1002/pros.22901] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/25/2014] [Indexed: 11/07/2022]
Abstract
BACKGROUND The mechanism(s) by which androgen receptor (AR) splice variants contribute to castration-resistant prostate cancer (CRPC) is still lacking. METHODS Expressions of epithelial-to-mesenchymal transition (EMT) and stem cell markers were molecularly tested using prostate cancer (PCa) cells transfected with AR and AR3 (also known as AR-V7) plasmids or siRNA, and also cultured cells under androgen deprivation therapy (ADT) condition. Cell migration, clonogenicity, sphere-forming capacity was assessed using PCa cells under all experimental conditions and 3,3'-diindolylmethane (DIM; BR-DIM) treatment. Human PCa samples from BR-DIM untreated or treated patients were also used for assessing the expression of AR3 and stem cell markers. RESULTS Overexpression of AR led to the induction of EMT phenotype, while overexpression of AR3 not only induced EMT but also led to the expression of stem cell signature genes. More importantly, ADT enhanced the expression of AR and AR3 concomitant with up-regulated expression of EMT and stem cell marker genes. Dihydrotestosterone (DHT) treatment decreased the expression of AR and AR3, and reversed the expression of these EMT and stem cell marker genes. BR-DIM administered to PCa patients prior to radical prostatectomy inhibited the expression of cancer stem cell markers consistent with inhibition of self-renewal of PCa cells after BR-DIM treatment. CONCLUSION AR variants could contribute to PCa progression through induction of EMT and acquisition of stem cell characteristics, which could be attenuated by BR-DIM, suggesting that BR-DIM could become a promising agent for the prevention of CRPC and/or for the treatment of PCa.
Collapse
Affiliation(s)
- Dejuan Kong
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan
| | | | | | | | | | | | | |
Collapse
|
189
|
Tanabe S, Aoyagi K, Yokozaki H, Sasaki H. Regulated genes in mesenchymal stem cells and gastric cancer. World J Stem Cells 2015; 7:208-222. [PMID: 25621121 PMCID: PMC4300932 DOI: 10.4252/wjsc.v7.i1.208] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/18/2014] [Accepted: 11/17/2014] [Indexed: 02/07/2023] Open
Abstract
AIM To investigate the genes regulated in mesenchymal stem cells (MSCs) and diffuse-type gastric cancer (GC), gene expression was analyzed. METHODS Gene expression of MSCs and diffuse-type GC cells were analyzed by microarray. Genes related to stem cells, cancer and the epithelial-mesenchymal transition (EMT) were extracted from human gene lists using Gene Ontology and reference information. Gene panels were generated, and messenger RNA gene expression in MSCs and diffuse-type GC cells was analyzed. Cluster analysis was performed using the NCSS software. RESULTS The gene expression of regulator of G-protein signaling 1 (RGS1) was up-regulated in diffuse-type GC cells compared with MSCs. A panel of stem-cell related genes and genes involved in cancer or the EMT were examined. Stem-cell related genes, such as growth arrest-specific 6, musashi RNA-binding protein 2 and hairy and enhancer of split 1 (Drosophila), NOTCH family genes and Notch ligands, such as delta-like 1 (Drosophila) and Jagged 2, were regulated. CONCLUSION Expression of RGS1 is up-regulated, and genes related to stem cells and NOTCH signaling are altered in diffuse-type GC compared with MSCs.
Collapse
Affiliation(s)
- Shihori Tanabe
- Shihori Tanabe, Division of Safety Information on Drug, Food and Chemicals, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan
| | - Kazuhiko Aoyagi
- Shihori Tanabe, Division of Safety Information on Drug, Food and Chemicals, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan
| | - Hiroshi Yokozaki
- Shihori Tanabe, Division of Safety Information on Drug, Food and Chemicals, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan
| | - Hiroki Sasaki
- Shihori Tanabe, Division of Safety Information on Drug, Food and Chemicals, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan
| |
Collapse
|
190
|
Makki J, Myint O, Wynn AA, Samsudin AT, John DV. Expression distribution of cancer stem cells, epithelial to mesenchymal transition, and telomerase activity in breast cancer and their association with clinicopathologic characteristics. Clin Med Insights Pathol 2015; 8:1-16. [PMID: 25624778 PMCID: PMC4287054 DOI: 10.4137/cpath.s19615] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 10/19/2014] [Accepted: 10/21/2014] [Indexed: 12/12/2022] Open
Abstract
A total of 167 surgically resected primary invasive breast carcinomas and 63 metastatic lymph node lesions were analyzed for immunohistochemical (IHC) localization of the CD44+CD24−low breast cancer stem cell (CSC) markers, epithelial to mesenchymal transition (EMT) markers, and telomerase activity by double-staining IHC technique, in formalin-fixed, paraffin-embedded tissue, the results were validated by double-staining immunofluorescent and flow cytometry techniques. The results showed that CSCs with CD44+CD24−low phenotype were significantly increased in node-positive tumors, high-grade tumors, and ductal carcinoma in situ (DCIS). There was a high incidence of telomerase expression in metastatic lymph node lesion. There were considerably high number of tumor cells with EMT expression in metastatic lymph node lesion, and triple-negative tumor. The occurrence of EMT phenomena was usually accompanied by the co-existence of CSCs of CD44+CD24−low phenotype. There was no association between the existence of CSCs and detection of telomerase activity in tumor cells. Increased numbers of both CSCs of CD44+CD24−low phenotype and cells underwent EMT in DCIS lesion might be an initial step in the stromal invasion and propagation of breast cancer, and occurrence of EMT in the breast tumor associated with high prevalence of CSCs, promoting tumor invasiveness and metastasis.
Collapse
Affiliation(s)
- Jaafar Makki
- Pathology Department, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Ohnmar Myint
- Department of Pathology, Faculty of Medicine and Health Science, University Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Aye Aye Wynn
- Department of Pathology, Faculty of Medicine and Health Science, University Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Ahmad Toha Samsudin
- Pathology Department, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Daisy Vanitha John
- Biotechnology Research Institute Universiti Malaysia Sabah, Sabah, Malaysia
| |
Collapse
|
191
|
Patel N, Baranwal S, Patel BB. A strategic approach to identification of selective inhibitors of cancer stem cells. Methods Mol Biol 2015; 1229:529-541. [PMID: 25325978 DOI: 10.1007/978-1-4939-1714-3_41] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Cancer stem-like cells (CSC) have been implicated in resistance to conventional chemotherapy as well as invasion and metastasis resulting in tumor relapse in majority of epithelial cancers including colorectal cancer. Hence, targeting CSC by small molecules is likely to improve therapeutic outcomes. Glycosaminoglycans (GAGs) are long linear polysaccharide molecules with varying degrees of sulfation that allows specific GAG-protein interaction which plays a key role in regulating cancer hallmarks such as cellular growth, angiogenesis, and immune modulation. However, identifying selective CSC-targeting GAG mimetic has been marred by difficulties associated with isolating and enriching CSC in vitro. Herein, we discuss two distinct methods, spheroid growth and EMT-transformed cells, to enrich CSC and set up medium- and high-throughput screen to identify selective CSC-targeting agents.
Collapse
Affiliation(s)
- Nirmita Patel
- Hunter Holmes McGuire VA Medical Center, 980230, Richmond, VA, 23249, USA
| | | | | |
Collapse
|
192
|
Lin J, Liu X, Ding D. Evidence for epithelial-mesenchymal transition in cancer stem-like cells derived from carcinoma cell lines of the cervix uteri. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:847-855. [PMID: 25755785 PMCID: PMC4348812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/22/2014] [Indexed: 06/04/2023]
Abstract
The cancer stem cell (CSC) paradigm is one possible way to understand the genesis of cancer, and cervical cancer in particular. We quantified and enriched ALDH1(+) cells within cervical cancer cell lines and subsequently characterized their phenotypical and functional properties like invasion capacity and epithelial-mesenchymal transition (EMT). ALDH1 expression in spheroid-derived cells (SDC) and the parental monolayer-derived cell (MDC) line was compared by flow-cytometry. Invasion capability was evaluated by Matrigel assay and expression of EMT-related genes Twist 1, Twist 2, Snail 1, Snail 2, Vimentin and E-cadherin by real-time PCR. ALDH1 expression was significantly higher in SDC. ALDH1(+) cells showed increased colony-formation. SDC expressed lower levels of E-cadherin and elevated levels of Twist 1, Twist 2, Snail 1, Snail 2 and Vimentin compared to MDC. Cervical cancer cell lines harbor potential CSC, characterized by ALDH1 expression as well as properties like invasiveness, colony-forming ability, and EMT. CSC can be enriched by anchorage-independent culture techniques, which may be important for the investigation of their contribution to therapy resistance, tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Jiaying Lin
- Shanghai OB/GYN Hospital, Fudan University Shanghai 200011, China
| | - Xishi Liu
- Shanghai OB/GYN Hospital, Fudan University Shanghai 200011, China
| | - Ding Ding
- Shanghai OB/GYN Hospital, Fudan University Shanghai 200011, China
| |
Collapse
|
193
|
Jung CW, Han KH, Seol H, Park S, Koh JS, Lee SS, Kim MJ, Choi IJ, Myung JK. Expression of cancer stem cell markers and epithelial-mesenchymal transition-related factors in anaplastic thyroid carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:560-568. [PMID: 25755746 PMCID: PMC4348894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/23/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is an undifferentiated tumor of the thyroid that has poor prognosis owing to its aggressive behavior and resistance to current treatments. We hypothesized that the stem cell properties induced by the epithelial-mesenchymal transition (EMT) was one of reasons for the dismal outcome of ATC. MATERIALS AND METHODS Paraffin blocks and slides of 17 ATC cases were retrieved. We also collected 60 cases of papillary thyroid carcinoma (PTC) for comparison. We used immunohistochemistry to examine the expression of multiple markers of cancer stem cells and EMT-activating transcriptional factors. RESULTS Majority of ATC cases showed loss of epithelial (E)-cadherin expression (15/17); however, all PTC cases (60/60) retained E-cadherin expression. EMT-activating transcription factors, such as snail and slug, were more frequently expressed in ATC than PTC cases (35.3% versus 6.7%, 76.5% versus 5%, respectively). Cancer stem cell markers such as CD133 and nestin were more highly expressed in ATC than PTC (52.9% versus 5%, 52.9% versus 0%, respectively). CONCLUSION We found that the expression of EMT-related factors and stem cell markers was higher in ATC than PTC. We therefore conclude that stemness induced by EMT plays an important role in the pathogenesis of ATC.
Collapse
Affiliation(s)
- Chang Won Jung
- Department of Pathology, Korea Cancer Center Hospital Seoul, Korea
| | - Kang Hee Han
- Department of Pathology, Korea Cancer Center Hospital Seoul, Korea
| | - Hyesil Seol
- Department of Pathology, Korea Cancer Center Hospital Seoul, Korea
| | - Sunhoo Park
- Department of Pathology, Korea Cancer Center Hospital Seoul, Korea ; Laboratory of Radiation Pathology, Korea Cancer Center Hospital Seoul, Korea
| | - Jae Soo Koh
- Department of Pathology, Korea Cancer Center Hospital Seoul, Korea
| | - Seung-Sook Lee
- Department of Pathology, Korea Cancer Center Hospital Seoul, Korea ; Laboratory of Radiation Pathology, Korea Cancer Center Hospital Seoul, Korea
| | - Min Joo Kim
- Internal Medicine, Korea Cancer Center Hospital Seoul, Korea
| | - Ik Joon Choi
- Otolaryngology-Head and Neck Surgery, Korea Cancer Center Hospital Seoul, Korea
| | - Jae Kyung Myung
- Department of Pathology, Korea Cancer Center Hospital Seoul, Korea ; Laboratory of Radiation Pathology, Korea Cancer Center Hospital Seoul, Korea
| |
Collapse
|
194
|
Abstract
The paper gives general information about the epithelial-mesenchymal transition (EMT), its morphological manifestations, altered expression of a number of proteins, types of EMT, and its role in embryogenesis and human diseases, including that about EMT as a mechanism by which the tumor cell acquires prometastatic potential. EMT is a process that is essential in health for gastrulation and the formation of neural crest cells; however, it is also important for the development of abnormalities, among other processes, organ fibrosis and tumor metastases. An understanding of the role of EMT in cancer spread has led to active studies of the process in the past decades. Despite the fact that there are sufficiently many publications on different aspects of EMT, the exact mechanisms regulating the process and the possibility for its therapeutic exposure remain unclear. There is also evidence on the possible association of EMT with the generation of cancer stem cells in tumors.
Collapse
Affiliation(s)
- M V Puchinskaya
- Belarusian State Medical University, Minsk, Republic of Belarus
| |
Collapse
|
195
|
Zheng Y, Li X, Hu H. Comprehensive discovery of DNA motifs in 349 human cells and tissues reveals new features of motifs. Nucleic Acids Res 2015; 43:74-83. [PMID: 25505144 PMCID: PMC4288161 DOI: 10.1093/nar/gku1261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/13/2014] [Accepted: 11/17/2014] [Indexed: 01/15/2023] Open
Abstract
Comprehensive motif discovery under experimental conditions is critical for the global understanding of gene regulation. To generate a nearly complete list of human DNA motifs under given conditions, we employed a novel approach to de novo discover significant co-occurring DNA motifs in 349 human DNase I hypersensitive site datasets. We predicted 845 to 1325 motifs in each dataset, for a total of 2684 non-redundant motifs. These 2684 motifs contained 54.02 to 75.95% of the known motifs in seven large collections including TRANSFAC. In each dataset, we also discovered 43 663 to 2 013 288 motif modules, groups of motifs with their binding sites co-occurring in a significant number of short DNA regions. Compared with known interacting transcription factors in eight resources, the predicted motif modules on average included 84.23% of known interacting motifs. We further showed new features of the predicted motifs, such as motifs enriched in proximal regions rarely overlapped with motifs enriched in distal regions, motifs enriched in 5' distal regions were often enriched in 3' distal regions, etc. Finally, we observed that the 2684 predicted motifs classified the cell or tissue types of the datasets with an accuracy of 81.29%. The resources generated in this study are available at http://server.cs.ucf.edu/predrem/.
Collapse
Affiliation(s)
- Yiyu Zheng
- Department of Electrical Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA
| | - Xiaoman Li
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Haiyan Hu
- Department of Electrical Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
196
|
Acquisition of epithelial-mesenchymal transition and cancer stem-like phenotypes within chitosan-hyaluronan membrane-derived 3D tumor spheroids. Biomaterials 2014; 35:10070-9. [PMID: 25282622 DOI: 10.1016/j.biomaterials.2014.09.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/11/2014] [Indexed: 12/22/2022]
Abstract
Cancer drug development has to go through rigorous testing and evaluation processes during pre-clinical in vitro studies. However, the conventional two-dimensional (2D) in vitro culture is often discounted by the insufficiency to present a more typical tumor microenvironment. The multicellular tumor spheroids have been a valuable model to provide more comprehensive assessment of tumor in response to therapeutic strategies. Here, we applied chitosan-hyaluronan (HA) membranes as a platform to promote three-dimensional (3D) tumor spheroid formation. The biological features of tumor spheroids of human non-small cell lung cancer (NSCLC) cells on chitosan-HA membranes were compared to those of 2D cultured cells in vitro. The cells in tumor spheroids cultured on chitosan-HA membranes showed higher levels of stem-like properties and epithelial-mesenchymal transition (EMT) markers, such as NANOG, SOX2, CD44, CD133, N-cadherin, and vimentin, than 2D cultured cells. Moreover, they exhibited enhanced invasive activities and multidrug resistance by the upregulation of MMP2, MMP9, BCRC5, BCL2, MDR1, and ABCG2 as compared with 2D cultured cells. The grafting densities of HA affected the tumor sphere size and mRNA levels of genes on the substrates. These evidences suggest that chitosan-HA membranes may offer a simple and valuable biomaterial platform for rapid generation of tumor spheroids in vitro as well as for further applications in cancer stem cell research and cancer drug screening.
Collapse
|
197
|
Li D, Masiero M, Banham AH, Harris AL. The notch ligand JAGGED1 as a target for anti-tumor therapy. Front Oncol 2014; 4:254. [PMID: 25309874 PMCID: PMC4174884 DOI: 10.3389/fonc.2014.00254] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/04/2014] [Indexed: 12/26/2022] Open
Abstract
The Notch pathway is increasingly attracting attention as a source of therapeutic targets for cancer. Ligand-induced Notch signaling has been implicated in various aspects of cancer biology; as a consequence, pan-Notch inhibitors and therapeutic antibodies targeting one or more of the Notch receptors have been investigated for cancer therapy. Alternatively, Notch ligands provide attractive options for therapy in cancer treatment due to their more restricted expression and better-defined functions, as well as their low rate of mutations in cancer. One of the Notch ligands, Jagged1 (JAG1), is overexpressed in many cancer types, and plays an important role in several aspects of tumor biology. In fact, JAG1-stimulated Notch activation is directly implicated in tumor growth through maintaining cancer stem cell populations, promoting cell survival, inhibiting apoptosis, and driving cell proliferation and metastasis. In addition, JAG1 can indirectly affect cancer by influencing tumor microenvironment components such as tumor vasculature and immune cell infiltration. This article gives an overview of JAG1 and its role in tumor biology, and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Demin Li
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| | - Massimo Masiero
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| | - Alison H Banham
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| | - Adrian L Harris
- Cancer Research UK Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford , UK
| |
Collapse
|
198
|
van der Horst G, Bos L, van der Mark M, Cheung H, Heckmann B, Clément-Lacroix P, Lorenzon G, Pelger RCM, Bevers RFM, van der Pluijm G. Targeting of alpha-v integrins reduces malignancy of bladder carcinoma. PLoS One 2014; 9:e108464. [PMID: 25247809 PMCID: PMC4172769 DOI: 10.1371/journal.pone.0108464] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 08/29/2014] [Indexed: 12/15/2022] Open
Abstract
Low survival rates of metastatic cancers emphasize the need for a drug that can prevent and/or treat metastatic cancer. αv integrins are involved in essential processes for tumor growth and metastasis and targeting of αv integrins has been shown to decrease angiogenesis, tumor growth and metastasis. In this study, the role of αv integrin and its potential as a drug target in bladder cancer was investigated. Treatment with an αv integrin antagonist as well as knockdown of αv integrin in the bladder carcinoma cell lines, resulted in reduced malignancy in vitro, as illustrated by decreased proliferative, migratory and clonogenic capacity. The CDH1/CDH2 ratio increased, indicating a shift towards a more epithelial phenotype. This shift appeared to be associated with downregulation of EMT-inducing transcription factors including SNAI2. The expression levels of the self-renewal genes NANOG and BMI1 decreased as well as the number of cells with high Aldehyde Dehydrogenase activity. In addition, self-renewal ability decreased as measured with the urosphere assay. In line with these observations, knockdown or treatment of αv integrins resulted in decreased metastatic growth in preclinical in vivo models as assessed by bioluminescence imaging. In conclusion, we show that αv integrins are involved in migration, EMT and maintenance of Aldehyde Dehydrogenase activity in bladder cancer cells. Targeting of αv integrins might be a promising approach for treatment and/or prevention of metastatic bladder cancer.
Collapse
Affiliation(s)
- Geertje van der Horst
- Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
- * E-mail:
| | - Lieke Bos
- Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maaike van der Mark
- Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Henry Cheung
- Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | - Rob C. M. Pelger
- Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Rob F. M. Bevers
- Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gabri van der Pluijm
- Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
199
|
Glackin CA. Targeting the Twist and Wnt signaling pathways in metastatic breast cancer. Maturitas 2014; 79:48-51. [DOI: 10.1016/j.maturitas.2014.06.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 06/21/2014] [Indexed: 01/31/2023]
|
200
|
Zhou XM, Zhang H, Han X. Role of epithelial to mesenchymal transition proteins in gynecological cancers: pathological and therapeutic perspectives. Tumour Biol 2014; 35:9523-30. [PMID: 25168372 DOI: 10.1007/s13277-014-2537-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/20/2014] [Indexed: 12/31/2022] Open
Abstract
Gynecorelogic cancers like ovarian, cervical, and endometrial cancers are among the major threats to modern life, especially to female health. Like some other types of cancers, all of these gynecological cancers have found to be associated with the developmental stage epithelial to mesenchymal transition (EMT). More specifically, the aberrant expression of major EMT markers, such as lower expressions of E-cadherin and alpha-catenin, and overexpressions of N-cadherin, beta-catenin, vimentin, and matrix metalloproteinases, have been reported in ovarian, cervical, and endometrial cancers. The transcription factors, such as Twist, Snail, Slug, and Zeb, which regulate these EMT mediators, are also reported to be overexpressed in gynecological cancers. In addition to the over/lower expression, the promoter methylation of some of these genes has been identified too. In the era of target-specific cancer therapeutics, some promising studies showed that targeting EMT markers might be an interesting and successful tool in future cancer therapy. In this study, we have reviewed the recent development in the research on the association of EMT markers with three major gynecological cancers in the perspectives of carcinogenesis and therapeutics.
Collapse
Affiliation(s)
- Xiao-Mei Zhou
- Department of Gynaecolgy and Obstetrics, Shenzhen FuTian District Traditional Chinese Medicine Hospital, No. 6001 Beihuan Blvd., Futian District, Shenzhen, 518000, China,
| | | | | |
Collapse
|