101
|
MIR17HG-miR-18a/19a axis, regulated by interferon regulatory factor-1, promotes gastric cancer metastasis via Wnt/β-catenin signalling. Cell Death Dis 2019; 10:454. [PMID: 31186404 PMCID: PMC6560107 DOI: 10.1038/s41419-019-1685-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022]
Abstract
MIR17HG, located on chromosome 13, is a class of Pri-miRNAs that generates six miRNAs: miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1 and miR-92-1. These miRNAs are ubiquitously overexpressed in diverse tumour types and exhibit complex biological links to tumour metastasis. We demonstrated that MIR17HG-derived miR-18a and miR-19a coordinately mediate gastric cancer cell metastasis by directly inhibiting SMAD2 expression and upregulating Wnt/β-catenin signalling. Based on previous studies, we hypothesised that an investigation of MIR17HG inhibition would be beneficial to clinical gastric cancer treatment, and systematically coupled bioinformatics analyses brought interferon regulatory factor-1 (IRF-1) to our attention. We then established stable clones in gastric cancer cells containing a doxycycline-inducible IRF-1 expression system and found that the expression of IRF-1 downregulates the embedded miRNAs of MIR17HG in gastric cancer cells and inhibits gastric cancer cell metastasis by attenuating Wnt/β-catenin signalling. Further rescue assays confirmed the crucial roles of miR-18a and miR-19a in the IRF-1-mediated inhibition of Wnt/β-catenin signalling. We also demonstrated that IRF-1 binds to the transcriptional site in the MIR17HG promoter and inhibits MIR17HG expression. Moreover, IFN-γ induced the IRF-1-mediated downregulation of MIR17HG in gastric cancer cells. Our hypothesis was supported by the results of immunohistochemistry analyses of clinical gastric cancer samples, and we also demonstrated the role of IRF-1 in inhibiting MIR17HG expression and tumour metastasis in vivo. We conclude that IRF-1 inhibits gastric cancer metastasis by downregulating MIR17HG-miR-18a/miR-19a axis expression and attenuating Wnt/β-catenin signalling.
Collapse
|
102
|
Yang Y, Wang X, Yang J, Duan J, Wu Z, Yang F, Zhang X, Xiao S. Loss of ARID1A promotes proliferation, migration and invasion via the Akt signaling pathway in NPC. Cancer Manag Res 2019; 11:4931-4946. [PMID: 31213911 PMCID: PMC6549766 DOI: 10.2147/cmar.s207329] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Abstract
Background: AT-rich interactive domain-containing protein 1A (ARID1A) is a member of the switch/sucrose nonfermentable chromatin remodeling complex, which has been observed to be mutated in various tumors. The loss of ARID1A is reported to be frequently associated with PI3K/Akt pathway activation. Objective: The roles of ARID1A in nasopharyngeal carcinoma (NPC) have not been reported until now. The aim of this research was to explore the clinical significance and potential mechanism of ARID1A in NPC development and progression. Methods: ARID1A expression levels were investigated in human NPC tissues and cell lines. The effects of ARID1A knockdown on nasopharyngeal cancer cell proliferation, migration and invasion were evaluated in vitro using CCK8, wound healing, transwell and flow cytometry assays. The expression of relevant proteins was evaluated by Western blot assays. Results: In this study, ARID1A was significantly downregulated in NPC tissues and cells. Furthermore, low ARID1A expression was significantly associated with aggressive clinicopathological characteristics and poor survival in NPC patients. Depletion of endogenous ARID1A by siRNA promoted proliferation, migration and invasion in CNE1 and HNE1 cells. Additionally, ARID1A knockdown increased the phosphorylation of Akt in NPC cells. High levels of p-Akt were also observed in NPC biopsies and correlated with ARID1A downregulation. These results imply that the loss of ARID1A could activate Akt signaling. In addition, MK-2206 (a highly selective inhibitor of Akt) partially suppressed NPC cell proliferation, migration and invasion, which were induced by ARID1A knockdown. Conclusion: Our findings indicate that ARID1A plays an essential role in modulating the Akt pathway, functions as a tumor suppressor in NPC and may be a potential target for NPC treatment.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pathology, the Second Affiliated Hospital, Guilin Medical University, Guilin 541199, People's Republic of China
| | - Xiaoyu Wang
- Department of Pathology, the Second Affiliated Hospital, Guilin Medical University, Guilin 541199, People's Republic of China
| | - Junjun Yang
- Department of Stomatology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, People's Republic of China
| | - Jingling Duan
- Department of Pathology, the Second Affiliated Hospital, Guilin Medical University, Guilin 541199, People's Republic of China
| | - Zhen Wu
- Xiangya Medical College of South Central University, Changsha 413000, People's Republic of China
| | - Fan Yang
- Department of Pathology, the Second Affiliated Hospital, Guilin Medical University, Guilin 541199, People's Republic of China
| | - Xiaoling Zhang
- Department of Physiology, Faculty of Basic Medical Science, Guilin Medical University, Guilin 541199, People's Republic of China
| | - Shengjun Xiao
- Department of Pathology, the Second Affiliated Hospital, Guilin Medical University, Guilin 541199, People's Republic of China
| |
Collapse
|
103
|
Vilches-Moure JG. Embryonic Chicken ( Gallus gallus domesticus) as a Model of Cardiac Biology and Development. Comp Med 2019; 69:184-203. [PMID: 31182184 PMCID: PMC6591676 DOI: 10.30802/aalas-cm-18-000061] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/06/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease remains one of the top contributors to morbidity and mortality in the United States. Increasing evidence suggests that many processes, pathways, and programs observed during development and organogenesis are recapitulated in adults in the face of disease. Therefore, a heightened understanding of cardiac development and organogenesis will help increase our understanding of developmental defects and cardiovascular diseases in adults. Chicks have long served as a model system in which to study developmental problems. Detailed descriptions of morphogenesis, low cost, accessibility, ease of manipulation, and the optimization of genetic engineering techniques have made chicks a robust model for studying development and make it a powerful platform for cardiovascular research. This review summarizes the cardiac developmental milestones of embryonic chickens, practical considerations when working with chicken embryos, and techniques available for use in chicks (including tissue chimeras, genetic manipulations, and live imaging). In addition, this article highlights examples that accentuate the utility of the embryonic chicken as model system in which to study cardiac development, particularly epicardial development, and that underscore the importance of how studying development informs our understanding of disease.
Collapse
Affiliation(s)
- José G Vilches-Moure
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California,
| |
Collapse
|
104
|
Role of Hedgehog Signaling in Breast Cancer: Pathogenesis and Therapeutics. Cells 2019; 8:cells8040375. [PMID: 31027259 PMCID: PMC6523618 DOI: 10.3390/cells8040375] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related mortality in women, only followed by lung cancer. Given the importance of BC in public health, it is essential to identify biomarkers to predict prognosis, predetermine drug resistance and provide treatment guidelines that include personalized targeted therapies. The Hedgehog (Hh) signaling pathway plays an essential role in embryonic development, tissue regeneration, and stem cell renewal. Several lines of evidence endorse the important role of canonical and non-canonical Hh signaling in BC. In this comprehensive review we discuss the role of Hh signaling in breast development and homeostasis and its contribution to tumorigenesis and progression of different subtypes of BC. We also examine the efficacy of agents targeting different components of the Hh pathway both in preclinical models and in clinical trials. The contribution of the Hh pathway in BC tumorigenesis and progression, its prognostic role, and its value as a therapeutic target vary according to the molecular, clinical, and histopathological characteristics of the BC patients. The evidence presented here highlights the relevance of the Hh signaling in BC, and suggest that this pathway is key for BC progression and metastasis.
Collapse
|
105
|
Jiao JX, Jiao LJ, Yang S, Zhao YJ. Knockdown of aristaless-like homeobox1 inhibits epithelial-mesenchymal transition through Wnt/β-catenin signaling pathway in melanoma cells. Biochem Biophys Res Commun 2019; 511:105-110. [PMID: 30773258 DOI: 10.1016/j.bbrc.2019.02.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 02/09/2019] [Indexed: 11/17/2022]
Abstract
Aristaless-like homeobox1 (ALX1), a member of the ALX family, is capable of mediating survival and development of mesenchyme-derived elements in vertebrates and its mutation will prevent the fusion of frontonasal and maxillary elements. Recently, ALX1 has been reported to be associated with cancer progression. However, the specific roles of ALX1 in melanoma remain unclear. In this study, we investigated the expression pattern and biological functions of ALX1 in melanoma. We found that ALX1 was highly expressed in melanoma tissues and cell lines. Knockdown of ALX1 suppressed the proliferation and invasion of melanoma cells. Furthermore, we showed that ALX1 knockdown reversed the epithelial-mesenchymal transition (EMT) process in melanoma cells, which might be attributed to inactivation of the Wnt/β-catenin pathway. Taken together, this study provided a new insight into the role of ALX1 as a therapeutic target for melanoma treatment.
Collapse
Affiliation(s)
- Jian-Xia Jiao
- Department of Dermatology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Lin-Jun Jiao
- Department of Dermatology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Sen Yang
- Department of Dermatology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Yan-Jun Zhao
- Department of Surgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, 014010, China.
| |
Collapse
|
106
|
Albuquerque APB, Balmaña M, Mereiter S, Pinto F, Reis CA, Beltrão EIC. Hypoxia and serum deprivation induces glycan alterations in triple negative breast cancer cells. Biol Chem 2019; 399:661-672. [PMID: 29894296 DOI: 10.1515/hsz-2018-0121] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/27/2018] [Indexed: 12/26/2022]
Abstract
Triple negative breast cancer (TNBC) is a major global public health problem. The lack of targeted therapy and the elevated mortality evidence the need for better knowledge of the tumor biology. Hypoxia and aberrant glycosylation are associated with advanced stages of malignancy, tumor progression and treatment resistance. Importantly, serum deprivation regulates the invasive phenotype and favors TNBC cell survival. However, in TNBC, the role of hypoxia and serum deprivation in the regulation of glycosylation remains largely unknown. The effects of hypoxia and serum deprivation on the expression of glycosyltransferases and glycan profile were evaluated in the MDA-MB-231 cell line. We showed that the overexpression of HIF-1α was accompanied by acquisition of epithelial-mesenchimal transition features. Significant upregulation of fucosyl- and sialyltransferases involved in the synthesis of tumor-associated carbohydrate antigens was observed together with changes in fucosylation and sialylation detected by Aleuria aurantia lectin and Sambucus nigra agglutinin lectin blots. Bioinformatic analysis further indicated a mechanism by which HIF-1α can regulate ST3GAL6 expression and the relationship within the intrinsic characteristics of TNBC tumors. In conclusion, our results showed the involvement of hypoxia and serum deprivation in glycosylation profile regulation of TNBC cells triggering breast cancer aggressive features and suggesting glycosylation as a potential diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Amanda P B Albuquerque
- Biomarkers in Cancer Research Group (BmC) - Federal University of Pernambuco (UFPE), 50670-901 Recife, Pernambuco, Brazil.,Department of Biochemistry, Federal University of Pernambuco (UFPE), 50670-901 Recife, Pernambuco, Brazil
| | - Meritxell Balmaña
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.,Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Stefan Mereiter
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.,Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Filipe Pinto
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.,Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.,Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal.,Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Eduardo I C Beltrão
- Biomarkers in Cancer Research Group (BmC) - Federal University of Pernambuco (UFPE), 50670-901 Recife, Pernambuco, Brazil.,Department of Biochemistry, Federal University of Pernambuco (UFPE), 50670-901 Recife, Pernambuco, Brazil
| |
Collapse
|
107
|
Lucena SR, Zamarrón A, Carrasco E, Marigil MA, Mascaraque M, Fernández-Guarino M, Gilaberte Y, González S, Juarranz A. Characterisation of resistance mechanisms developed by basal cell carcinoma cells in response to repeated cycles of Photodynamic Therapy. Sci Rep 2019; 9:4835. [PMID: 30886381 PMCID: PMC6423284 DOI: 10.1038/s41598-019-41313-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Photodynamic Therapy (PDT) with methyl-aminolevulinate acid (MAL-PDT) is being used for the treatment of Basal cell carcinoma (BCC), but recurrences have been reported. In this work, we have evaluated resistance mechanisms to MAL-PDT developed by three BCC cell lines (ASZ, BSZ and CSZ), derived from mice on a ptch+/- background and with or without p53 expression, subjected to 10 cycles of PDT (10thG). The resistant populations showed mesenchymal-like structure and diminished proliferative capacity and size compared to the parental (P) cells. The resistance was dependent on the production of the endogenous photosensitiser protoporphyrin IX in the CSZ cell line and on its cellular localisation in ASZ and BSZ cells. Moreover, resistant cells expressing the p53 gene presented lower proliferation rate and increased expression levels of N-cadherin and Gsk3β (a component of the Wnt/β-catenin pathway) than P cells. In contrast, 10thG cells lacking the p53 gene showed lower levels of expression of Gsk3β in the cytoplasm and of E-cadherin and β-catenin in the membrane. In addition, resistant cells presented higher tumorigenic ability in immunosuppressed mice. Altogether, these results shed light on resistance mechanisms of BCC to PDT and may help to improve the use of this therapeutic approach.
Collapse
Affiliation(s)
- Silvia Rocio Lucena
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | - Alicia Zamarrón
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | - Elisa Carrasco
- Molecular Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | | | - Marta Mascaraque
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain
| | | | | | - Salvador González
- Medicine and Medical Specialties Department, Alcalá de Henares University, Madrid, Spain
| | - Angeles Juarranz
- Biology Department, Faculty of Sciences, Autonomous University of Madrid, Madrid, Spain.
- Instituto Ramón y Cajal de Investigaciones Sanitarias, IRYCIS, Madrid, Spain.
| |
Collapse
|
108
|
Wang XL, Huang C. Difference of TGF-β/Smads signaling pathway in epithelial-mesenchymal transition of normal colonic epithelial cells induced by tumor-associated fibroblasts and colon cancer cells. Mol Biol Rep 2019; 46:2749-2759. [PMID: 30835040 DOI: 10.1007/s11033-019-04719-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/25/2019] [Indexed: 02/07/2023]
Abstract
Tumor microenvironment (TME) crucially functions in tumor initiation and progression. Stroma-tumor interactions and cellular transdifferentiation are the prerequisite for tumor formation. Transforming growth factor-β (TGF-β), a major cytokine secreted by tumor-associated fibroblasts (TAFs) and cancer cells, is a crucial player involving cell transdifferentiation. Therefore, we hypothesized that these TAFs and cancer cells also affect normal colon epithelium. In our study, we found for the first time that colon cancer cells HCT116 and TAF-like CCD-18Co cells induced epithelial-mesenchymal transition (EMT)-like transdifferentiation in colon epithelial cells HCoEpiCs, with enhanced migratio. Dysfunction of TGF-β/Smads signal was also observed in the EMT-transformed HCoEpiCs. We wondered whether these phenomena were regulated by TGF-β/Smads signaling pathway. A TGFβ receptor kinase I (TβRI) inhibitor LY364947 was used. We found that the EMT induced by the HCT116- and CCD-18Co-derived CM was suppressed by the LY364947. Besides, different expression profiles for the components of TGF-β/Smads pathway were found in the EMT-like HCoEpiCs, but high expression of p-Smad2/3 and Smad4 was the common feature. Our observations suggest that the mechanisms of phenotypic transition of colon epithelial cells are cellular environment-dependent, which maybe a basis of potential therapy targeting TME.
Collapse
Affiliation(s)
- Xiu-Lian Wang
- Community Health Service Center, Shenzhen Bao'an Traditional Chinese Medicine Hospital Group, Shenzhen, China
| | - Chao Huang
- Central Laboratory, Affiliated Bao'an Hospital of Shenzhen, Southern Medical University, Shenzhen, China.
| |
Collapse
|
109
|
Liu Z, Liu J, Dong X, Hu X, Jiang Y, Li L, Du T, Yang L, Wen T, An G, Feng G. Tn antigen promotes human colorectal cancer metastasis via H-Ras mediated epithelial-mesenchymal transition activation. J Cell Mol Med 2019; 23:2083-2092. [PMID: 30637914 PMCID: PMC6378212 DOI: 10.1111/jcmm.14117] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 12/16/2022] Open
Abstract
Tn antigen is a truncated O-glycan, frequently detected in colorectal cancer (CRC), but its precise role in CRC metastasis is not well addressed. Here we investigated the effects of Core 1 β3Gal-T specific molecular chaperone (Cosmc) deletion-mediated Tn antigen exposure on CRC metastasis and its underlying mechanism. We first used CRISPR/Cas9 technology to knockout Cosmc, which is required for normal O-glycosylation, and thereby obtained Tn-positive CRC cells. We then investigated the biological consequences of Tn antigen expression in CRC. The results showed that Tn-positive cells exhibited an enhanced metastatic capability both in vitro and in vivo. A further analysis indicated that Tn antigen expression induced typical activation of epithelial-mesenchymal transition (EMT). Mechanistically, we found that H-Ras, which is known to drive EMT, was markedly up-regulated in Tn-positive cells, whereas knockdown of H-Ras suppressed Tn antigen induced activation of EMT. Furthermore, we confirmed that LS174T cells (Tn-positive) transfected with wild-type Cosmc, thus expressing no Tn antigen, had down-regulation of H-Ras expression and subsequent inhibition of EMT process. In addition, analysis of 438 samples in TCGA cohort demonstrated that Cosmc expression was reversely correlated with H-Ras, underscoring the significance of Tn antigen-H-Ras signalling in CRC patients. These data demonstrated that Cosmc deletion-mediated Tn antigen exposure promotes CRC metastasis, which is possibly mediated by H-Ras-induced EMT activation.
Collapse
Affiliation(s)
- Zhe Liu
- Department of OncologyBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Jian Liu
- Medical Research CenterBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Xichen Dong
- Medical Research CenterBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Xin Hu
- Department of OncologyBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Yuliang Jiang
- Department of OncologyBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Lina Li
- Department of OncologyBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Tan Du
- Department of OncologyBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Lei Yang
- Medical Research CenterBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Tao Wen
- Medical Research CenterBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Guangyu An
- Department of OncologyBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Guosheng Feng
- Department of OncologyBeijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
110
|
Zhu B, Zhao L, Liu Y, Jin Y, Feng J, Zhao F, Sun J, Geng R, Wei Y. Induction of phosphatase shatterproof 2 by evodiamine suppresses the proliferation and invasion of human cholangiocarcinoma. Int J Biochem Cell Biol 2019; 108:98-110. [DOI: 10.1016/j.biocel.2019.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/23/2018] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
|
111
|
Macharia LW, Wanjiru CM, Mureithi MW, Pereira CM, Ferrer VP, Moura-Neto V. MicroRNAs, Hypoxia and the Stem-Like State as Contributors to Cancer Aggressiveness. Front Genet 2019; 10:125. [PMID: 30842790 PMCID: PMC6391339 DOI: 10.3389/fgene.2019.00125] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/04/2019] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that play key regulatory roles in cancer acting as both oncogenes and tumor suppressors. Due to their potential roles in improving cancer prognostic, predictive, diagnostic and therapeutic approaches, they have become an area of intense research focus in recent years. Several studies have demonstrated an altered expression of several miRNAs under hypoxic condition and even shown that the hypoxic microenvironment drives the selection of a more aggressive cancer cell population through cellular adaptations referred as the cancer stem-like cell. These minor fractions of cells are characterized by their self-renewal abilities and their ability to maintain the tumor mass, suggesting their crucial roles in cancer development. This review aims to highlight the interconnected role between miRNAs, hypoxia and the stem-like state in contributing to the cancer aggressiveness as opposed to their independent contributions, and it is based in four aggressive tumors, namely glioblastoma, cervical, prostate, and breast cancers.
Collapse
Affiliation(s)
- Lucy Wanjiku Macharia
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline Muriithi Wanjiru
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Valéria Pereira Ferrer
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
112
|
NETO2 promotes invasion and metastasis of gastric cancer cells via activation of PI3K/Akt/NF-κB/Snail axis and predicts outcome of the patients. Cell Death Dis 2019; 10:162. [PMID: 30770791 PMCID: PMC6377647 DOI: 10.1038/s41419-019-1388-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 12/28/2022]
Abstract
Aberrant expression of neuropilin and tolloid-like 2 (NETO2) has been observed during the progression of some human carcinomas. However, the expression pattern and clinical relevance of NETO2 in gastric cancer (GC) remain to be elucidated. In this study, we found that NETO2 expression was higher in GC tissues compared with paired non-cancerous tissues. Moreover, the expression of NETO2 was positively correlated with clinical stage, invasion depth, lymph node metastasis, and tumor size, but inversely correlated with overall and disease-free survival rates. Cox regression analysis identified NETO2 as an independent prognostic indicator for GC patients. Overexpression of NETO2 facilitated migration and invasion of GC cells in vitro and metastasis in vivo in association with induction of epithelial-mesenchymal transition. Conversely, knockdown of NETO2 had the opposite effects. Mechanistically, silencing NETO2 reduced the phosphorylation of PI3K, AKT, and NF-κB p65 as well as the expression of Snail, whereas NETO2 overexpression achieved the opposite results. Furthermore, we identified TNFRSF12A as a mediator for NETO2 to activate PI3K/AKT/NF-κB/Snail axis. Collectively, our results demonstrate that NETO2 promotes invasion and metastasis of GC cells and represents a novel prognostic indicator as well as a potential therapeutic target in GC.
Collapse
|
113
|
Gu J, Cui CF, Yang L, Wang L, Jiang XH. Emodin Inhibits Colon Cancer Cell Invasion and Migration by Suppressing Epithelial-Mesenchymal Transition via the Wnt/β-Catenin Pathway. Oncol Res 2019; 27:193-202. [PMID: 29301594 PMCID: PMC7848449 DOI: 10.3727/096504018x15150662230295] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Colon cancer (CC) is the third most common cancer worldwide. Emodin is an anthraquinone-active substance that has the ability to affect tumor progression. Our study aims to explore the effects and the relevant mechanism of emodin on the invasion and migration of CC in vitro and in vivo. In our study, we found that emodin inhibited the invasion and migration abilities of RKO cells and decreased the expression of matrix metalloproteinase-7 (MMP-7), MMP-9, and vascular endothelial growth factor (VEGF) in a dose-dependent manner. Further research suggested that emodin inhibited EMT by increasing the mRNA level of E-cadherin and decreasing the expression of N-cadherin, Snail, and β-catenin. Emodin also significantly inhibited the activation of the Wnt/β-catenin signaling pathway by downregulating the expression of related downstream target genes, including TCF4, cyclin D1, and c-Myc. A Wnt/β-catenin signaling pathway agonist abolished the effect of emodin on EMT and cell mobility, suggesting that emodin exerted its regulating role through the Wnt/β-catenin pathway. The CC xenograft model was established to study the antitumor efficiency of emodin in vivo. The in vivo study further demonstrated that emodin (40 mg/kg) suppressed tumor growth by inhibiting EMT via the Wnt/β-catenin signaling pathway in vivo. Taken together, we suggest that emodin inhibits the invasion and migration of CC cells in vitro and in vivo by blocking EMT, which is related with the inhibition of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Juan Gu
- *Department of Clinical Pharmacy, West China School of Pharmacy, Sichuan University, Sichuan, P.R. China
| | - Chang-fu Cui
- †Department of Neurology, Research Institute of China Weapons Industry, 521 Hospital, Shanxi, P.R. China
| | - Li Yang
- ‡Microbiological Laboratory, Xinyang Vocational and Technical College, Henan, P.R. China
| | - Ling Wang
- *Department of Clinical Pharmacy, West China School of Pharmacy, Sichuan University, Sichuan, P.R. China
| | - Xue-hua Jiang
- *Department of Clinical Pharmacy, West China School of Pharmacy, Sichuan University, Sichuan, P.R. China
| |
Collapse
|
114
|
Chen G, Wu J, Li J, Wang J. Identification and Characterization of Glycine Decarboxylase as a Direct Target of Snail in the Epithelial-Mesenchymal Transition of Cancer Cells. ACTA ACUST UNITED AC 2019; 1:55-62. [PMID: 33869785 PMCID: PMC8049539 DOI: 10.4103/tme.tme_8_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Context/Aims: Metabolic reprogramming and cellular plasticity drive tumorigenesis. However, how these cellular events collectively contribute to the oncogenic process is poorly understood. Epithelial-mesenchymal transition (EMT), a fundamental mechanism of cellular plasticity, is governed by the EMT transcription repressors such as Snail. In the present study, through establishment and characterization of inducible overexpression of Snail in A549 lung cancer cells, we aim to define the metabolic reprogramming in response to Snail in the EMT of lung cancer cells. Methods/Results: Our metabolomic analysis suggests that forced expression of Snail accompanied reduced diversion of glycolytic metabolites to the serine/glycine metabolic shunt, a critical metabolic branch that distributes glucose catabolic intermediates to the major anabolic pathways. Our gene expression profiling and molecular characterization revealed that Snail actively suppressed the expression of glycine decarboxylase (GLDC), a key enzyme on the serine/glycine metabolic shunt, through binding to an evolutionarily conserved E-box motif and thereby inhibiting the promoter of the GLDC gene. Besides, knockdown of GLDC led to a cellular function shift from proliferation to migration. Conclusion: This study has revealed a novel molecular link that integrates the serine/glycine metabolism with the Snail-mediated EMT program in cancer cells.
Collapse
Affiliation(s)
- Guohua Chen
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jianmei Wu
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jian Wang
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, USA.,Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
115
|
Liao Z, Zhang H, Fan P, Huang Q, Dong K, Qi Y, Song J, Chen L, Liang H, Chen X, Zhang Z, Zhang B. High PLK4 expression promotes tumor progression and induces epithelial‑mesenchymal transition by regulating the Wnt/β‑catenin signaling pathway in colorectal cancer. Int J Oncol 2019; 54:479-490. [PMID: 30570110 PMCID: PMC6317648 DOI: 10.3892/ijo.2018.4659] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 10/24/2018] [Indexed: 02/07/2023] Open
Abstract
Polo‑like kinase 4 (PLK4) has been identified as an oncogene, which is overexpressed in various types of human cancer; however, its role in colorectal cancer (CRC) development remains unknown. The present study demonstrated that PLK4 protein expression was upregulated in CRC tissues compared with in normal tissues through western blotting. In addition, immunohistochemical analysis of 39 CRC specimens further demonstrated that PLK4 protein expression was upregulated in 64.1% (25/39) of samples. Increased PLK4 expression was closely associated with enhanced tumor size (P=0.031), lymph node metastasis (P=0.016) and TNM stage (P=0.001). Subsequently, cell viability, wound scratch, migration and invasion assays were conducted in vitro, and nude mice CRC xenograft models were generated. The results demonstrated that knockdown of PLK4 in CRC cells resulted in significant decreases in cell viability and proliferation, and decreased the protein expression levels of N‑cadherin and snail, which are biomarkers of epithelial‑mesenchymal transition. Furthermore, PLK4 knockdown inactivated the Wnt/β‑catenin pathway in CRC cells in vitro and in vivo, and suppressed the growth of xenograft tumors in nude mice. In conclusion, these results suggested that PLK4 may promote the carcinogenesis and metastasis of CRC, thus indicating that PLK4 may be considered a molecular target for CRC treatment.
Collapse
Affiliation(s)
- Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry Health, Wuhan, Hubei 430030
| | - Hongwei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
| | - Pan Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
| | - Qibo Huang
- Department of Clinical Medicine, Medical College of Wuhan University of Science and Technology, Wuhan, Hubei 430081, P.R. China
| | - Keshuai Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
| | - Yongqiang Qi
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
| | - Jia Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
| | - Lin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry Health, Wuhan, Hubei 430030
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry Health, Wuhan, Hubei 430030
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry Health, Wuhan, Hubei 430030
| |
Collapse
|
116
|
Xin H, Wang C, Liu Z. miR-196a-5p promotes metastasis of colorectal cancer via targeting IκBα. BMC Cancer 2019; 19:30. [PMID: 30621631 PMCID: PMC6325824 DOI: 10.1186/s12885-018-5245-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022] Open
Abstract
Background MicroRNA-196a-5p (miR-196a-5p) has been reported to be involved in the metastatic process of several cancers. In present work, we aimed to investigate the effects of miR-196a-5p and its potential target IκBα on migration, invasion and epithelial-mesenchymal transition (EMT) of colorectal cancer (CRC) cells. Methods CCK-8 assay, wound healing assay and cell invasion assay were performed to evaluate the cell proliferation, migration and invasion. In vivo metastasis models were used to investigate the tumor metastasis ability. Real-time PCR, immunofluorescence staining or western blot were utilized to detect the expression of miR-196a-5p, IκBα, p-IκBα, nuclear p65 and EMT markers including E-cadherin, N-cadherin and fibronectin. Dual luciferase reporter assay was carried out to determine whether there is a direct interaction between miR-196a-5p and IκBα mRNA. Results Using SW480 cell with miR-196-5p over-expressed plus SW620 and HCT116 cells with miR-196a-5p knockdown, we found that miR-196a-5p promoted cell proliferation, migration and invasion in vitro and facilitated liver metastasis in vivo. We also observed that miR-196a-5p knockdown or NF-κB pathway inhibition up-regulated E-cadherin while down-regulated N-cadherin and fibronectin. By contrast, miR-196a-5p over-expression promoted EMT process of CRC. Data of dual luciferase reporter assay indicated that miR-196a-5p targeted the IκBα. Moreover, miR-196a-5p down-regulated IκBα expression while up-regulated nuclear p65 expression. Additionally, over-expression of IκBα in CRC cells attenuated the effects of miR-196a-5p on cell migration, invasion and EMT. Conclusions miR-196a-5p may play a key role in EMT, invasion and metastasis of CRC cells via targeting the IκBα. Electronic supplementary material The online version of this article (10.1186/s12885-018-5245-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- He Xin
- Department of Radiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Chuanzhuo Wang
- Department of Radiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Zhaoyu Liu
- Department of Radiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
117
|
Decreased expression of LKB1 is associated with epithelial-mesenchymal transition and led to an unfavorable prognosis in gastric cancer. Hum Pathol 2019; 83:133-139. [DOI: 10.1016/j.humpath.2018.08.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 12/16/2022]
|
118
|
Gao Y, Chen Z, Wang R, Tan X, Huang C, Chen G, Chen Z. LXRα Promotes the Differentiation of Human Gastric Cancer Cells through Inactivation of Wnt/β-catenin Signaling. J Cancer 2019; 10:156-167. [PMID: 30662536 PMCID: PMC6329868 DOI: 10.7150/jca.28600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/02/2018] [Indexed: 12/18/2022] Open
Abstract
LXRα is a subtype of the liver X receptors (LXRs). There is accumulating evidence to support the involvement of LXRα in a variety of malignancies. However, the function and specific mechanism of LXRα in gastric cancer (GC) remain unclear. In this study, the expression of LXRα was significantly lower in poorly differentiated and undifferentiated GC tissues compared with well- and moderately differentiated GC tissues by immunohistochemistry analysis. The activation of LXRα leads to the decreased expression of β-catenin, CD44, and Cyclin D1, whereas the inhibition of LXRα has opposite effect. The same results were obtained in animal experiments. Furthermore, results showed that CD44 and Cyclin D1 expression significantly decreased when Wnt/β-catenin signaling was blocked in LXRα silent GC cells, whereas it was significantly increased when Wnt/β-catenin signaling was activated in LXRα over-expressed GC cells. CD44 and Cyclin D1, downstream targets of Wnt/β-catenin signaling, are specific markers for cell differentiation. Therefore, we conclude that LXRα may promote the differentiation of human GC cells through inactivation of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yu Gao
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Hunan key laboratory of precise diagnosis and treatment of gastrointestinal tumor, Changsha, Hunan, P.R. China
| | - Zihua Chen
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Hunan key laboratory of precise diagnosis and treatment of gastrointestinal tumor, Changsha, Hunan, P.R. China
| | - Ran Wang
- Department of Colorectal and Anus Surgery, Xiangya Hospital of Central South University, Hunan key laboratory of precise diagnosis and treatment of gastrointestinal tumor, Changsha, Hunan, P.R. China
| | - Xiangzhou Tan
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Hunan key laboratory of precise diagnosis and treatment of gastrointestinal tumor, Changsha, Hunan, P.R. China
| | - Changhao Huang
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Hunan key laboratory of precise diagnosis and treatment of gastrointestinal tumor, Changsha, Hunan, P.R. China
| | - Guanyang Chen
- Department of Gastrointestinal Surgery, Xiangya Hospital of Central South University, Hunan key laboratory of precise diagnosis and treatment of gastrointestinal tumor, Changsha, Hunan, P.R. China
| | - Zhikang Chen
- Department of Colorectal and Anus Surgery, Xiangya Hospital of Central South University, Hunan key laboratory of precise diagnosis and treatment of gastrointestinal tumor, Changsha, Hunan, P.R. China
| |
Collapse
|
119
|
You ZP, Chen SS, Yang ZY, Li SR, Xiong F, Liu T, Fu SH. GEP100/ARF6 regulates VEGFR2 signaling to facilitate high-glucose-induced epithelial-mesenchymal transition and cell permeability in retinal pigment epithelial cells. Am J Physiol Cell Physiol 2018; 316:C782-C791. [PMID: 30540496 DOI: 10.1152/ajpcell.00312.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell permeability and epithelial-mesenchymal transition (EMT) were found to be enhanced in diabetic retinopathy, and the aim of this study was to investigate the underlying mechanism. ARPE-19 cell line or primary retinal pigment epithelial (RPE) cells were cultured under high or normal glucose conditions. Specific shRNAs were employed to knock down ADP-ribosylation factor 6 (ARF6), GEP100, or VEGF receptor 2 (VEGFR2) in ARPE-19 or primary RPE cells. Cell migration ability was measured using Transwell assay. Western blotting was used to measure indicated protein levels. RPE cells treated with high glucose showed increased cell migration, paracellular permeability, EMT, and expression of VEGF. Knockdown of VEGFR2 inhibited the high-glucose-induced effects on RPE cells via inactivation of ARF6 and MAPK pathways. Knockdown ARF6 or GEP100 led to inhibition of high-glucose-induced effects via inactivation of VEGFR2 pathway. Knockdown of ARF6, but not GEP100, decreased high-glucose-induced internalization of VEGFR2. High-glucose enhances EMT and cell permeability of RPE cells through activation of VEGFR2 and ARF6/GEP100 pathways, which form a positive feedback loop to maximize the activation of VEGF/VEGFR2 signaling.
Collapse
Affiliation(s)
- Zhi-Peng You
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Shan-Shan Chen
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Zhong-Yi Yang
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Shu-Rong Li
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Fan Xiong
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Ting Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| | - Shu-Hua Fu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University , Nanchang , People's Republic of China
| |
Collapse
|
120
|
Huang C, Tao L, Wang X, Pang Z. Berberine reversed the epithelial‐mesenchymal transition of normal colonic epithelial cells induced by SW480 cells through regulating the important components in the TGF‐β pathway. J Cell Physiol 2018; 234:11679-11691. [PMID: 30536375 DOI: 10.1002/jcp.27835] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/06/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Chao Huang
- Institute of Pharmacology, Sun Yat‐Sen Zhongshan Medical College, Sun Yat‐Sen University Guangzhou China
| | - Liang Tao
- Institute of Pharmacology, Sun Yat‐Sen Zhongshan Medical College, Sun Yat‐Sen University Guangzhou China
| | - Xiu‐lian Wang
- Department of Traditional Chinese Medicine Affiliated Bao’an Hospital of Traditional Chinese Medicine of Shenzhen, Traditional Chinese Medicine University Of Guangzhou Shenzhen China
| | - Zuoliang Pang
- Department of Oncology Affiliated Bao’an Hospital of Shenzhen, Southern Medical University Shenzhen China
| |
Collapse
|
121
|
Gimba E, Brum M, Nestal De Moraes G. Full-length osteopontin and its splice variants as modulators of chemoresistance and radioresistance (Review). Int J Oncol 2018; 54:420-430. [DOI: 10.3892/ijo.2018.4656] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/25/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Etel Gimba
- Program of Cellular and Molecular Oncobiology, National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | - Mariana Brum
- Program of Cellular and Molecular Oncobiology, National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | - Gabriela Nestal De Moraes
- Cellular and Molecular Hemato-Oncology Laboratory, Molecular Hemato-Oncology Program, National Cancer Institute, Rio de Janeiro 20230-130, Brazil
| |
Collapse
|
122
|
Peng L, Jiang B, Yuan X, Qiu Y, Peng J, Huang Y, Zhang C, Zhang Y, Lin Z, Li J, Yao W, Deng W, Zhang Y, Meng M, Pan X, Li C, Yin D, Bi X, Li G, Lin DC. Super-Enhancer-Associated Long Noncoding RNA HCCL5 Is Activated by ZEB1 and Promotes the Malignancy of Hepatocellular Carcinoma. Cancer Res 2018; 79:572-584. [PMID: 30482773 DOI: 10.1158/0008-5472.can-18-0367] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 08/01/2018] [Accepted: 11/21/2018] [Indexed: 01/18/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most dominant causes of neoplasm-related deaths worldwide. In this study, we identify and characterize HCCL5, a novel cytoplasmic long noncoding RNA (lncRNA), as a crucial oncogene in HCC. HCCL5 promoted cell growth, G1-S transition, invasion, and metastasis while inhibiting apoptosis of HCC cells both in vitro and in vivo. Moreover, HCCL5 was upregulated in TGF-β1-induced classical epithelial-to-mesenchymal transition (EMT) models, and this lncRNA in turn accelerated the EMT phenotype by upregulating the expression of transcription factors Snail, Slug, ZEB1, and Twist1. HCCL5 was transcriptionally driven by ZEB1 via a super-enhancer and was significantly and frequently overexpressed in human HCC tissues, correlating with worse overall survival of patients with HCC. Together, this study characterizes HCCL5 as a super-enhancer-driven lncRNA promoting HCC cell viability, migration, and EMT. Our data also suggest that HCCL5 may serve as a novel prognostic biomarker and therapeutic target in HCC. SIGNIFICANCE: These findings identify the lncRNA HCCL5 as a super-enhancer-driven oncogenic factor that promotes the malignancy of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Li Peng
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Binyuan Jiang
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Xiaoqing Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuntan Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiangyun Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yongsheng Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaoyang Zhang
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Yin Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhaoyu Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Oral & Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinsong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Oral & Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weicheng Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Weixi Deng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yaqin Zhang
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Meng Meng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xi Pan
- Department of Oncology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xinyu Bi
- Department of Hepato-Biliary Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guancheng Li
- Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China. .,Cancer Research Institute, Central South University, Changsha, China
| | - De-Chen Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
123
|
McNair K, Forrest CM, Vincenten MCJ, Darlington LG, Stone TW. Serine protease modulation of Dependence Receptors and EMT protein expression. Cancer Biol Ther 2018; 20:349-367. [PMID: 30403907 PMCID: PMC6370372 DOI: 10.1080/15384047.2018.1529109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/03/2018] [Accepted: 09/22/2018] [Indexed: 12/11/2022] Open
Abstract
Expression of the tumour suppressor Deleted in Colorectal Cancer (DCC) and the related protein neogenin is reduced by the mammalian serine protease chymotrypsin or the bacterial serine protease subtilisin, with increased cell migration. The present work examines whether these actions are associated with changes in the expression of cadherins, β-catenin and vimentin, established markers of the Epithelial-Mesenchymal Transition (EMT) which has been linked with cell migration and tumour metastasis. The results confirm the depletion of DCC and neogenin and show that chymotrypsin and subtilisin also reduce expression of β-catenin in acutely prepared tissue sections but not in human mammary adenocarcinoma MCF-7 or MDA-MB-231 cells cultured in normal media, or primary normal human breast cells. A loss of β-catenin was also seen in low serum media but transfecting cells with a dcc-containing plasmid induced resistance. E-cadherin was not consistently affected but vimentin was induced by low serum-containing media and was increased by serine proteases in MCF-7 and MDA-MB-231 cells in parallel with increased wound closure. Vimentin might contribute to the promotion of cell migration. The results suggest that changes in EMT proteins depend on the cells or tissues concerned and do not parallel the expression of DCC and neogenin. The increased cell migration induced by serine proteases is not consistently associated with the expression of the EMT proteins implying either that the increased migration may be independent of EMT or supporting the view that EMT is not itself consistently related to migration. (241).
Collapse
Affiliation(s)
- Kara McNair
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Caroline M. Forrest
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Maria C. J. Vincenten
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Trevor W. Stone
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- The Kennedy Institute, University of Oxford, Oxford UK
| |
Collapse
|
124
|
Obesity and gastrointestinal cancer: the interrelationship of adipose and tumour microenvironments. Nat Rev Gastroenterol Hepatol 2018; 15:699-714. [PMID: 30323319 DOI: 10.1038/s41575-018-0069-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing recognition of an association between obesity and many cancer types exists, but how the myriad of local and systemic effects of obesity affect key cellular and non-cellular processes within the tumour microenvironment (TME) relevant to carcinogenesis, tumour progression and response to therapies remains poorly understood. The TME is a complex cellular environment in which the tumour exists along with blood vessels, immune cells, fibroblasts, bone marrow-derived inflammatory cells, signalling molecules and the extracellular matrix. Obesity, in particular visceral obesity, might fuel the dysregulation of key pathways relevant to both the adipose microenvironment and the TME, which interact to promote carcinogenesis in at-risk epithelium. The tumour-promoting effects of obesity can occur at the local level as well as systemically via circulating inflammatory, growth factor and metabolic mediators associated with adipose tissue inflammation, as well as paracrine and autocrine effects. This Review explores key pathways linking visceral obesity and gastrointestinal cancer, including inflammation, hypoxia, altered stromal and immune cell function, energy metabolism and angiogenesis.
Collapse
|
125
|
Li Y, Xu Z, Li J, Ban S, Duan C, Liu W. Interleukin-18 expression in oral squamous cell carcinoma: its role in tumor cell migration and invasion, and growth of tumor cell xenografts. FEBS Open Bio 2018; 8:1953-1963. [PMID: 30524946 PMCID: PMC6275252 DOI: 10.1002/2211-5463.12532] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/16/2018] [Accepted: 09/20/2018] [Indexed: 12/30/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common head and neck malignancies. Advanced stages of the disease are associated with poor survival, highlighting a need for new treatment modalities. We previously showed that the proinflammatory cytokine interleukin‐18 (IL‐18) has a tumor suppressive role in OSCC. Here, we investigated the effects of IL‐18 on proliferation, migration, and invasion of OSCC cells ex vivo and in vitro, and in nude mouse xenografts. We report that expression of tankyrase 2 (TNKS2), β‐catenin, and N‐cadherin was higher in tumor cells than in normal mucosae, whereas the expression of IL‐18 and E‐cadherin was higher in normal than in tumor tissues. Elevated expression of IL‐18 (P < 0.01) and E‐cadherin (P = 0.034) was associated with tumor differentiation, whereas expression of TNKS2 (P < 0.01), β‐catenin (P = 0.012), and N‐cadherin (P < 0.01) was associated with tumor de‐differentiation. Furthermore, compared with the vector control, IL‐18 overexpression promoted tumor cell migration and invasion (P < 0.01), but inhibited growth of tumor cell xenografts (P < 0.05). At the protein level, expression levels of IL‐18 (P < 0.01), TNKS2 (P = 0.045), β‐catenin (P = 0.028), and N‐cadherin (P = 0.068) were upregulated in tumor cells after IL‐18 overexpression compared with those of the vector control mice, whereas expression levels of E‐cadherin (P = 0.045) were decreased. In conclusion, our data suggest that IL‐18 overexpression induces oral SCC cell invasion and metastasis by promoting the tumor cell epithelial–mesenchymal transition via the Wnt/β‐catenin signaling pathway.
Collapse
Affiliation(s)
- Yuyang Li
- Department of Dental Implantology School and Hospital of Stomatology Jilin University Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Changchun Jilin China
| | - Zhiming Xu
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Jilin University Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Changchun Jilin China
| | - Jia Li
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Jilin University Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Changchun Jilin China
| | - Shuofeng Ban
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Jilin University Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Changchun Jilin China
| | - Congcong Duan
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Jilin University Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Changchun Jilin China
| | - Weiwei Liu
- Department of Oral and Maxillofacial Surgery School and Hospital of Stomatology Jilin University Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling Changchun Jilin China
| |
Collapse
|
126
|
A review of radiation genomics: integrating patient radiation response with genomics for personalised and targeted radiation therapy. JOURNAL OF RADIOTHERAPY IN PRACTICE 2018. [DOI: 10.1017/s1460396918000547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AbstractBackgroundThe success of radiation therapy for cancer patients is dependent on the ability to deliver a total tumouricidal radiation dose capable of eradicating all cancer cells within the clinical target volume, however, the radiation dose tolerance of the surrounding healthy tissues becomes the main dose-limiting factor. The normal tissue adverse effects following radiotherapy are common and significantly impact the quality of life of patients. The likelihood of developing these adverse effects following radiotherapy cannot be predicted based only on the radiation treatment parameters. However, there is evidence to suggest that some common genetic variants are associated with radiotherapy response and the risk of developing adverse effects. Radiation genomics is a field that has evolved in recent years investigating the association between patient genomic data and the response to radiation therapy. This field aims to identify genetic markers that are linked to individual radiosensitivity with the potential to predict the risk of developing adverse effects due to radiotherapy using patient genomic information. It also aims to determine the relative radioresponse of patients using their genetic information for the potential prediction of patient radiation treatment response.Methods and materialsThis paper reports on a review of recent studies in the field of radiation genomics investigating the association between genomic data and patients response to radiation therapy, including the investigation of the role of genetic variants on an individual’s predisposition to enhanced radiotherapy radiosensitivity or radioresponse.ConclusionThe potential for early prediction of treatment response and patient outcome is critical in cancer patients to make decisions regarding continuation, escalation, discontinuation, and/or change in treatment options to maximise patient survival while minimising adverse effects and maintaining patients’ quality of life.
Collapse
|
127
|
Yu XH, Ren XH, Liang XH, Tang YL. Roles of fatty acid metabolism in tumourigenesis: Beyond providing nutrition (Review). Mol Med Rep 2018; 18:5307-5316. [PMID: 30365095 DOI: 10.3892/mmr.2018.9577] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 09/19/2018] [Indexed: 02/05/2023] Open
Abstract
Fatty acid (FA) metabolism, including the uptake, de novo synthesis and oxidation of FAs, is critical for the survival, proliferation, differentiation and metastasis of cancer cells. Several bodies of evidence have confirmed the metabolic reprogramming of FAs that occurs during cancer development. The present review aimed to evaluate FAs in terms of how the hallmarks of cancer are gradually established in tumourigenesis and tumour progression, and consider the auxo‑action and exact mechanisms of FA metabolism in these processes. In addition, this interaction in the tumour microenvironment was also discussed. Based on the role of FA metabolism in tumour development, targeting FA metabolism may effectively target cancer, affecting a number of important characteristics of cancer progression and survival.
Collapse
Affiliation(s)
- Xiang-Hua Yu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, Sichuan 610041, P.R. China
| | - Xiao-Hua Ren
- Department of Stomatology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, P.R. China
| | - Xin-Hua Liang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, Sichuan 610041, P.R. China
| | - Ya-Ling Tang
- Department of Oral Pathology, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
128
|
Miranda-Gonçalves V, Lameirinhas A, Henrique R, Jerónimo C. Metabolism and Epigenetic Interplay in Cancer: Regulation and Putative Therapeutic Targets. Front Genet 2018; 9:427. [PMID: 30356832 PMCID: PMC6190739 DOI: 10.3389/fgene.2018.00427] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
Alterations in the epigenome and metabolism affect molecular rewiring of cancer cells facilitating cancer development and progression. Modulation of histone and DNA modification enzymes occurs owing to metabolic reprogramming driven by oncogenes and expression of metabolism-associated genes is, in turn, epigenetically regulated, promoting the well-known metabolic reprogramming of cancer cells and, consequently, altering the metabolome. Thus, several malignant traits are supported by the interplay between metabolomics and epigenetics, promoting neoplastic transformation. In this review we emphasize the importance of tumour metabolites in the activity of most chromatin-modifying enzymes and implication in neoplastic transformation. Furthermore, candidate targets deriving from metabolism of cancer cells and altered epigenetic factors is emphasized, focusing on compounds that counteract the epigenomic-metabolic interplay in cancer.
Collapse
Affiliation(s)
- Vera Miranda-Gonçalves
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Ana Lameirinhas
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal.,Master in Oncology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
129
|
Chen W, Jin Y, Yang H, Wei L, Lin J. Hedyotis diffusa Willd reduces migration and invasion through inhibition of TGF-β-induced EMT in colorectal cancer cells. Eur J Integr Med 2018. [DOI: 10.1016/j.eujim.2018.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
130
|
Jolly MK, Somarelli JA, Sheth M, Biddle A, Tripathi SC, Armstrong AJ, Hanash SM, Bapat SA, Rangarajan A, Levine H. Hybrid epithelial/mesenchymal phenotypes promote metastasis and therapy resistance across carcinomas. Pharmacol Ther 2018; 194:161-184. [PMID: 30268772 DOI: 10.1016/j.pharmthera.2018.09.007] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer metastasis and therapy resistance are the major unsolved clinical challenges, and account for nearly all cancer-related deaths. Both metastasis and therapy resistance are fueled by epithelial plasticity, the reversible phenotypic transitions between epithelial and mesenchymal phenotypes, including epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET). EMT and MET have been largely considered as binary processes, where cells detach from the primary tumor as individual units with many, if not all, traits of a mesenchymal cell (EMT) and then convert back to being epithelial (MET). However, recent studies have demonstrated that cells can metastasize in ways alternative to traditional EMT paradigm; for example, they can detach as clusters, and/or occupy one or more stable hybrid epithelial/mesenchymal (E/M) phenotypes that can be the end point of a transition. Such hybrid E/M cells can integrate various epithelial and mesenchymal traits and markers, facilitating collective cell migration. Furthermore, these hybrid E/M cells may possess higher tumor-initiation and metastatic potential as compared to cells on either end of the EMT spectrum. Here, we review in silico, in vitro, in vivo and clinical evidence for the existence of one or more hybrid E/M phenotype(s) in multiple carcinomas, and discuss their implications in tumor-initiation, tumor relapse, therapy resistance, and metastasis. Together, these studies drive the emerging notion that cells in a hybrid E/M phenotype may occupy 'metastatic sweet spot' in multiple subtypes of carcinomas, and pathways linked to this (these) hybrid E/M state(s) may be relevant as prognostic biomarkers as well as a promising therapeutic targets.
Collapse
Affiliation(s)
- Mohit Kumar Jolly
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| | - Jason A Somarelli
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Maya Sheth
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Adrian Biddle
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Satyendra C Tripathi
- Department of Clinical Cancer Prevention, UT MD Anderson Cancer Center, Houston, USA
| | - Andrew J Armstrong
- Duke Cancer Institute and Department of Medicine, Duke University Medical Center, Durham, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, UT MD Anderson Cancer Center, Houston, USA
| | - Sharmila A Bapat
- National Center for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development & Genetics, Indian Institute of Science, Bangalore, India
| | - Herbert Levine
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| |
Collapse
|
131
|
MTP18 overexpression contributes to tumor growth and metastasis and associates with poor survival in hepatocellular carcinoma. Cell Death Dis 2018; 9:956. [PMID: 30237394 PMCID: PMC6148245 DOI: 10.1038/s41419-018-0987-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/27/2018] [Accepted: 08/20/2018] [Indexed: 12/18/2022]
Abstract
Background Human MTP18 (mitochondrial protein 18 kDa) is a novel nuclear-encoded mitochondrial membrane protein that is involved in controlling mitochondrial fission. Our bioinformatic analysis of TCGA data revealed an aberrant overexpression of MTP18 in hepatocellular carcinoma (HCC). We analyzed its biological effects and prognostic significance in this malignancy. Methods MTP18 expression was evaluated by qRT-PCR and western blot analysis in 20 paired tumor and peritumor tissues. Clinical impact of MTP18 overexpression was assessed in 156 patients with HCC. The effects of MTP18 knockdown or overexpression on cell growth and metastasis were determined by cell proliferation, colony formation, cell cycle, apoptosis, migration, and invasion assays. Furthermore, the underlying molecular mechanisms by which MTP18 overexpression promoted HCC cell growth and metastasis were explored. Results MTP18 was commonly overexpressed in HCC tissues mainly due to the downregulation of miR-125b, which significantly contributed to poor prognosis of HCC patients. Functional experiments revealed that MTP18 promoted both the growth and metastasis of HCC cells by inducing the progression of cell cycle, epithelial to mesenchymal transition (EMT) and production of MMP–9, and suppressing cell apoptosis. Mechanistically, increased mitochondrial fission and subsequent ROS production was found to be involved in the promotion of growth and metastasis by MTP18 in HCC cells. Conclusions MTP18 plays a pivotal oncogenic role in hepatocellular carcinogenesis; its overexpression may serve as a novel prognostic factor and a therapeutic target in HCC.
Collapse
|
132
|
Li J, Sun H, Liu T, Kong J. MicroRNA-423 promotes proliferation, migration and invasion and induces chemoresistance of endometrial cancer cells. Exp Ther Med 2018; 16:4213-4224. [PMID: 30344696 DOI: 10.3892/etm.2018.6710] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
Altered microRNA expression serves essential roles in tumorigenesis and progression in endometrial cancer. In the present study the effect of miR-423 on proliferation, chemosensitivity, migration and invasion of endometrial cancer cells was examined. A WST-1 assay was used to examine the proliferation of HEC-1B and Ishikawa endometrial cancer cells with either upregulation or downregulation of miR-423, with or without cisplatin treatment. The migration and invasion of HEC-1B and Ishikawa endometrial cancer cells were examined via Transwell migration and Matrigel invasion assays. Protein expression levels, including B cell lymphoma-2 (Bcl-2), Bcl-2 associated X protein, E- and N-cadherin, snail, vimentin, phosphatase and tensin homolog (PTEN) and protein kinase B (AKT) were examined by western blotting. A caspase-Glo3/7 assay was carried out to evaluate the effect of miR-423 on cisplatin-induced apoptosis in HEC-1B and Ishikawa endometrial cancer cells. Overexpression of miR-423 enhanced the proliferation, and increased migration and invasion in endometrial cancer cells. miR-423 also decreased the sensitivity of endometrial cancer cells following cisplatin treatment. miR-423 inhibited cisplatin-induced apoptosis in endometrial cancer cells by regulation of caspase 3/7 and Bcl-2 expression. Furthermore, the E-cadherin expression was significantly decreased, and the expression of N-cadherin, snail and Vimentin were increased in both HEC-1B cells and Ishikawa cells following overexpression of miR-423. Conversely, downregulation of miR-423 increased the expression of E-cadherin and decreased the expression of N-cadherin, snail and Vimentin. Further experiments demonstrated that the expression levels of PTEN and phosphorylated-AKT in HEC-1B and Ishikawa endometrial cancer cells was decreased and increased, respectively, following aberrant expression of miR-423. miR-423 displayed an important role in tumorigenesis and progression in endometrial cancer cells, and may therefore be used as a potential biomarker to predict chemotherapy response and prognosis in endometrial cancer.
Collapse
Affiliation(s)
- Jie Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Huijie Sun
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ting Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jian Kong
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
133
|
Sha M, Jeong S, Qiu BJ, Tong Y, Xia L, Xu N, Zhang JJ, Xia Q. Isolation of cancer-associated fibroblasts and its promotion to the progression of intrahepatic cholangiocarcinoma. Cancer Med 2018; 7:4665-4677. [PMID: 30062820 PMCID: PMC6144256 DOI: 10.1002/cam4.1704] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/22/2018] [Accepted: 07/05/2018] [Indexed: 12/15/2022] Open
Abstract
Intrahepatic cholangiocarcinoma is a highly fatal tumor characterized by an abundant stromal environment. Cancer‐associated fibroblasts play key roles in tumor growth and invasiveness and have been regarded as a potential therapeutic target. This study was designed to isolate human primary cancer‐associated fibroblasts of intrahepatic cholangiocarcinoma to study tumor‐stroma interactions and to analyze the clinical relevance of alpha‐smooth muscle actin ‐positive cancer‐associated fibroblasts in patients with intrahepatic cholangiocarcinoma. The isolated cancer‐associated fibroblasts were positive for alpha‐smooth actin, fibroblast‐specific protein‐1, fibroblast activation protein, and PDGFR‐β. In addition, cancer‐associated fibroblasts were found to increase proliferation, migration, and invasion of cholangiocarcinoma cells in vitro and promote tumor growth of mice in vivo. Moreover, alpha‐smooth muscle actin‐positive expression of cancer‐associated fibroblasts predicted unfavorable prognosis in patients with intrahepatic cholangiocarcinoma and showed correlation with presence of lymph node metastasis. This study may provide a useful tool to investigate further effect of cancer‐associated fibroblasts on the molecular mechanism of cholangiocarcinoma cells as well as contribution of cancer‐associated fibroblasts in lymphangiogenesis and lymph node metastasis.
Collapse
Affiliation(s)
- Meng Sha
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Seogsong Jeong
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bi-Jun Qiu
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Tong
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Xia
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ning Xu
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Jun Zhang
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
134
|
Zhang J, Zheng G, Zhou L, Li P, Yun M, Shi Q, Wang T, Wu X. Notch signalling induces epithelial‑mesenchymal transition to promote metastasis in oral squamous cell carcinoma. Int J Mol Med 2018; 42:2276-2284. [PMID: 30015856 DOI: 10.3892/ijmm.2018.3769] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 06/26/2018] [Indexed: 11/06/2022] Open
Abstract
The activation of Notch signalling induces epithelial‑mesenchymal transition (EMT), but this signalling pathway and its association with EMT in the context of cell motility in oral squamous cell carcinoma (OSCC) remains unclear. The present study aimed to investigate the role of the Notch signalling pathway and EMT in the metastatic potential of OSCC using 2 cell lines, Tca8113 and CAL27. The data demonstrated that zinc finger domain SNAI1 (Snail) knockdown by small interfering RNA decreased the expression of vimentin and increased the expression of epithelial cadherin (E‑cadherin). In addition, silencing Snail also significantly inhibited cell migration in the 2 OSCC cell lines. It was also identified that blocking Notch signalling with the g‑secretase inhibitor DAPT decreased the expression of the EMT markers Snail and vimentin and increased E‑cadherin expression, accompanied by a significant inhibition of cell migration in the 2 OSCC cell lines. These data clearly indicate that Notch signalling mediates EMT to promote metastasis in OSCC cells. Therefore, targeting Notch signalling and its association with EMT may provide novel insights into the mechanism of invasion and metastasis in OSCC and potential therapeutic interventions.
Collapse
Affiliation(s)
- Jianping Zhang
- School of Clinical Medicine, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Genjian Zheng
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Lan Zhou
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Pengcheng Li
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Man Yun
- Department of Stomatology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Qi Shi
- School of Clinical Medicine, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Tingli Wang
- School of Clinical Medicine, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Xiaotong Wu
- School of Clinical Medicine, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| |
Collapse
|
135
|
Melnik S, Dvornikov D, Müller-Decker K, Depner S, Stannek P, Meister M, Warth A, Thomas M, Muley T, Risch A, Plass C, Klingmüller U, Niehrs C, Glinka A. Cancer cell specific inhibition of Wnt/β-catenin signaling by forced intracellular acidification. Cell Discov 2018; 4:37. [PMID: 29977599 PMCID: PMC6028397 DOI: 10.1038/s41421-018-0033-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 01/02/2023] Open
Abstract
Use of the diabetes type II drug Metformin is associated with a moderately lowered risk of cancer incidence in numerous tumor entities. Studying the molecular changes associated with the tumor-suppressive action of Metformin we found that the oncogene SOX4, which is upregulated in solid tumors and associated with poor prognosis, was induced by Wnt/β-catenin signaling and blocked by Metformin. Wnt signaling inhibition by Metformin was surprisingly specific for cancer cells. Unraveling the underlying specificity, we identified Metformin and other Mitochondrial Complex I (MCI) inhibitors as inducers of intracellular acidification in cancer cells. We demonstrated that acidification triggers the unfolded protein response to induce the global transcriptional repressor DDIT3, known to block Wnt signaling. Moreover, our results suggest that intracellular acidification universally inhibits Wnt signaling. Based on these findings, we combined MCI inhibitors with H+ ionophores, to escalate cancer cells into intracellular hyper-acidification and ATP depletion. This treatment lowered intracellular pH both in vitro and in a mouse xenograft tumor model, depleted cellular ATP, blocked Wnt signaling, downregulated SOX4, and strongly decreased stemness and viability of cancer cells. Importantly, the inhibition of Wnt signaling occurred downstream of β-catenin, encouraging applications in treatment of cancers caused by APC and β-catenin mutations.
Collapse
Affiliation(s)
- Svitlana Melnik
- 1Division of Epigenetics and Cancer Risks Factors, German Cancer Research Center, Heidelberg, D-69120 Germany.,2DNA vectors, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Dmytro Dvornikov
- 3Division of Systems Biology and Signal Transduction, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Karin Müller-Decker
- 5Tumor Models Unit, Center for Preclinical Research, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Sofia Depner
- 3Division of Systems Biology and Signal Transduction, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Peter Stannek
- Division of Molecular Embryology, DKFZ-ZMBH Allianz, German Cancer Research Center, Heidelberg, D-69120 Germany
| | - Michael Meister
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,7Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, D-69126 Germany
| | - Arne Warth
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,8Institute of Pathology, Heidelberg University Hospital, Heidelberg, 69120 Germany
| | - Michael Thomas
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,7Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, D-69126 Germany
| | - Tomas Muley
- 4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,7Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, D-69126 Germany
| | - Angela Risch
- 1Division of Epigenetics and Cancer Risks Factors, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,9Department of Molecular Biology, University of Salzburg, Salzburg, 5020 Austria.,Cancer Cluster Salzburg, Salzburg, 5020 Austria
| | - Christoph Plass
- 1Division of Epigenetics and Cancer Risks Factors, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Ursula Klingmüller
- 3Division of Systems Biology and Signal Transduction, German Cancer Research Center, Heidelberg, D-69120 Germany.,4Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Allianz, German Cancer Research Center, Heidelberg, D-69120 Germany.,11Institute of Molecular Biology (IMB), Mainz, 55128 Germany
| | - Andrey Glinka
- Division of Molecular Embryology, DKFZ-ZMBH Allianz, German Cancer Research Center, Heidelberg, D-69120 Germany
| |
Collapse
|
136
|
Scotti FM, Mitt VC, Vieira DS, Biz MT, Castro RG, Modolo F. Expression of stem cell markers Nanog and Nestin in lip squamous cell carcinoma and actinic cheilitis. Oral Dis 2018; 24:1209-1216. [PMID: 29761881 DOI: 10.1111/odi.12891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 12/19/2022]
Abstract
Cancer stem cell (CSC) proteins have been observed in several lesions and are associated with tumor beginning, evolution, and resistance to treatment. OBJECTIVES To investigate the presence of NANOG, NESTIN, and β-tubulin in lip squamous cell carcinoma (LSCC), actinic cheilitis (AC), and normal epithelium (NE). MATERIALS AND METHODS Thirty cases of LSCC, thirty cases of AC (both analyzed according to the WHO classification and AC according to the binary classification), and twenty cases of NE were submitted to an immunohistochemical study. RESULTS NANOG was more expressed in the nuclei of AC compared to NE (p = 0.007), as well as in high-risk AC cases (p = 0.017) and well-differentiated LSCCs (no significance). There was an accumulation of nuclear NANOG from mild to moderate and severe ACs. NESTIN was significantly less present in NE compared to AC (p = 0.001) and LSCC (p = 0.003). There was a higher expression in severe dysplasia or high-risk AC and well-differentiated LSCC. These results indicate an upregulation of NANOG and NESTIN in the early stages of carcinogenesis. β-tubulin was intensely present in all lesions. CONCLUSION The results suggest an upregulation of NANOG and NESTIN in the biological behavior these diseases, mainly in the transformation from AC to LSCC.
Collapse
Affiliation(s)
- Fernanda M Scotti
- Dentistry Graduate Program, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Verônica C Mitt
- Multidisciplinary Residence Program, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Daniella Sc Vieira
- Pathology Department, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Michelle T Biz
- Morphology Sciences Department, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Renata G Castro
- Dentistry Department, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Filipe Modolo
- Dentistry Graduate Program, Federal University of Santa Catarina, Florianopolis, Brazil.,Pathology Department, Federal University of Santa Catarina, Florianopolis, Brazil
| |
Collapse
|
137
|
Toth L, Nagy B, Mehes G, Laszlo E, Molnar PP, Poka R, Hernadi Z. Cell adhesion molecule profiles, proliferation activity and p53 expression in advanced epithelial ovarian cancer induced malignant ascites-Correlation of tissue microarray and cytology microarray. Pathol Res Pract 2018; 214:978-985. [PMID: 29801775 DOI: 10.1016/j.prp.2018.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/03/2018] [Accepted: 05/15/2018] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Peritoneal dissemination accompanied by ascites formation is common in epithelial ovarian cancer (EOC). Adhesion molecules are crucial in metastatic spread and the latter involves epithelial-mesenchymal transition (EMT). This study aimed at: (1) clarifying whether E-cadherin and β-catenin expression and proliferative activity in metastatic ovarian cancer are inter-related; (2) Identifying possible correlations between cell adhesion molecular expression profiles, the proliferative activity and p53 expression of tumor cells and tumor grade and stage; (3) testing the cytology microarray (CMA) technique in analyzing metastasis formation. MATERIAL AND METHODS Both tumorous and ascitic samples from 27 EOC patients were examined by using tissue microarray (TMA) and cytology microarray (CMA), respectively. CMA blocks were constructed using cores from each cell block of the ascitic specimens. Expression of E-cadherin, β-catenin, Ki-67 and p53 was immunohistochemically detected both in TMA and CMA blocks. RESULTS E-cadherin expression was higher in ascitic cells than in primary tumor cells (p = .294). β-catenin expression was significantly lower in ascitic cells than in primary tumor cells (p = .006). Expression of Ki-67 was lower and expression of p53 was higher in primary tumors than in ascitic cells, for p53 the difference was significant (p = .001). Both Ki-67 and p53 expression elevated significantly in high-grade primary tumor cells and in ascites cells (p = .039, and p = .004, respectively). CONCLUSION Epithelial-mesenchymal transition- mesenchymal-epithelial transition is suggested as the best descriptive term for our IHC observations which accompany increased proliferative activity of ascitic cells. The CMA method is an adequate and reliable method for the analysis of ascitic tumor cells disseminating from ovarian malignancies.
Collapse
Affiliation(s)
- Laszlo Toth
- Department of Pathology, Clinical Centre, University of Debrecen, Hungary.
| | - Bence Nagy
- Department of Pathology, University of Szeged, Hungary
| | - Gabor Mehes
- Department of Pathology, Clinical Centre, University of Debrecen, Hungary
| | - Eszter Laszlo
- Department of Medical Imaging, University of Debrecen, Hungary
| | - Peter Pal Molnar
- Department of Pathology, Clinical Centre, University of Debrecen, Hungary
| | - Robert Poka
- Department of Obstetrics and Gynecology, Clinical Centre, University of Debrecen, Hungary
| | - Zoltan Hernadi
- Department of Obstetrics and Gynecology, Clinical Centre, University of Debrecen, Hungary
| |
Collapse
|
138
|
Zhang Y, Feng Y, Ji D, Wang Q, Qian W, Wang S, Zhang Z, Ji B, Zhang C, Sun Y, Fu Z. TRIM27 functions as an oncogene by activating epithelial-mesenchymal transition and p-AKT in colorectal cancer. Int J Oncol 2018; 53:620-632. [PMID: 29767249 PMCID: PMC6017157 DOI: 10.3892/ijo.2018.4408] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023] Open
Abstract
Tripartite motif-containing 27 (TRIM27) belongs to the tripartite motif (TRIM) protein family and is involved in various malignant tumor processes. However, the function and mechanism of TRIM27 in colorectal cancer (CRC) remains to be elucidated. In the present study, the expression of TRIM27 was analyzed in CRC tissues and adjacent normal tissues by reverse transcription-quantitative polymerase chain reaction and immunohistochemistry. LoVo and HCT116 cell lines were then selected to further investigate the function of TRIM27 in the proliferation, invasion and metastasis of CRC in vitro and in vivo. Finally, the potential mechanism underlying the effects of TRIM27 in CRC was examined by western blotting. The results showed that TRIM27 was upregulated in CRC tissues, and the expression level of TRIM27 was significantly associated with tumor invasion, metastasis and prognosis. Following TRIM27 inhibition and overexpression in CRC cells, it was found that TRIM27 promoted cell proliferation, possibly via the inhibition of apoptosis and cell cycle regulation. TRIM27 also facilitated invasion and metastasis. Finally, it was observed that TRIM27 promoted epithelial-mesenchymal transition and activated phosphorylated AKT serine/threonine kinase in CRC cells. These results suggested that TRIM27 is an oncogenic protein in the progression of CRC, and may represent a novel target for CRC detection and therapy.
Collapse
Affiliation(s)
- Yue Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yifei Feng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Dongjian Ji
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qingyuan Wang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wenwei Qian
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shijia Wang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhiyuan Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Bing Ji
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chuan Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zan Fu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
139
|
Tang L, Wei F, Wu Y, He Y, Shi L, Xiong F, Gong Z, Guo C, Li X, Deng H, Cao K, Zhou M, Xiang B, Li X, Li Y, Li G, Xiong W, Zeng Z. Role of metabolism in cancer cell radioresistance and radiosensitization methods. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:87. [PMID: 29688867 PMCID: PMC5914062 DOI: 10.1186/s13046-018-0758-7] [Citation(s) in RCA: 294] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Radioresistance is a major factor leading to the failure of radiotherapy and poor prognosis in tumor patients. Following the application of radiotherapy, the activity of various metabolic pathways considerably changes, which may result in the development of resistance to radiation. MAIN BODY Here, we discussed the relationships between radioresistance and mitochondrial and glucose metabolic pathways, aiming to elucidate the interplay between the tumor cell metabolism and radiotherapy resistance. In this review, we additionally summarized the potential therapeutic targets in the metabolic pathways. SHORT CONCLUSION The aim of this review was to provide a theoretical basis and relevant references, which may lead to the improvement of the sensitivity of radiotherapy and prolong the survival of cancer patients.
Collapse
Affiliation(s)
- Le Tang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Wei
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yingfen Wu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yi He
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lei Shi
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ke Cao
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Xiang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
140
|
Epithelial to mesenchymal transition induces stem cell like phenotype in renal cell carcinoma cells. Cancer Cell Int 2018; 18:57. [PMID: 29681769 PMCID: PMC5896088 DOI: 10.1186/s12935-018-0555-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/05/2018] [Indexed: 12/31/2022] Open
Abstract
Background Metastatic dissemination of solid tumors is often initiated by reactivation of an embryonic development program, epithelial-to-mesenchymal-transition (EMT). EMT has been associated with acquiring invasiveness and resistance to conventional therapies. However, the precise role of EMT during renal cell carcinoma is still debatable and is under investigation. In this context, our study is designed to evaluate the role of cyclosporine (CsA) and transforming growth factor-β (TGFβ) administration in inducing EMT-like state in renal carcinoma cells. We also studied the associated phenotypic changes which may lead to tumor metastasis. Methods The morphological changes in renal cell carcinoma cells (A498) treated with TGF-β/CsA were observed by microscopy. Atomic force microscope was used to evaluate the changes in elasticity of cells treated with TGF-β/CsA. The expression of mesenchymal and chemoresistance genes were checked by RT-PCR. Assays for migration, invasion, sphere formation ability and expression of cancer stem cell-like phenotypes were done to evaluate the metastatic potential of these cells. Lineage specific differentiations were also done to determine the acquisition of stem-cell like phenotype. Results Our results showed that treatment with TGF-β/CsA led to loss of epithelial characteristics and gain of mesenchymal phenotype in vitro. Changes in shape and elastic properties of the cancer cells favoured metastatic progression, increased tumorisphere formation and invasiveness post treatment. We also observed higher expression of chemoresistance and stemness markers in EMT-induced cells. These cells also differentiated to various lineages like osteoblasts, adipocytes, neural and hepatic cells when induced with the respective differentiation media. Conclusion We concluded that TGF-β/CsA treatment led to acquisition of EMT-like cancer stem cells phenotype that enhanced local invasion and dissemination of renal carcinoma cells. This subpopulation of cells with EMT-like phenotype a can provide a better perception of the metastatic process. This can provide an in vitro system for testing pharmaceuticals for modulating EMT as a viable strategy within the therapeutic armamentarium for RCC patients. The results of our findings also suggest that CsA directly induced EMT like changes in epithelial cell which may be responsible for the potential risk of malignancy in transplant patients. Electronic supplementary material The online version of this article (10.1186/s12935-018-0555-6) contains supplementary material, which is available to authorized users.
Collapse
|
141
|
Lin WF, Lin XL, Fu SW, Yang L, Tang CT, Gao YJ, Chen HY, Ge ZZ. Pseudopod-associated protein KIF20B promotes Gli1-induced epithelial-mesenchymal transition modulated by pseudopodial actin dynamic in human colorectal cancer. Mol Carcinog 2018; 57:911-925. [PMID: 29573464 DOI: 10.1002/mc.22812] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 02/27/2018] [Accepted: 03/20/2018] [Indexed: 12/30/2022]
Abstract
Kinesin family member 20B (KIF20B) has been reported to have an oncogenic role in bladder and hepatocellular cancer cells, but its role in colorectal cancer (CRC) progression remains unclear. In this study, we assessed the mRNA and protein levels of KIF20B in CRC tissues using qRT-PCR and immunohistochemistry, respectively. KIF20B was overexpressed in CRC tissues and was associated with cancer invasion and metastasis. Mechanistically, KIF20B overexpression promoted the epithelial-mesenchymal transition (EMT) process mediated by glioma-associated oncogene 1 (Gli1) as well as CRC cell migration and invasion. Interestingly, KIF20B was localized in pseudopod protrusions of CRC cells and influenced the formation of cell protrusions, especially the EMT-related invadopodia. Moreover, intracellular actin dynamic participated in the modulation of the Gli1-mediated EMT and EMT-related cell pseudopod protrusion formation induced by KIF20B. We identified a role for KIF20B in CRC progression and revealed a correlation between KIF20B expression in CRC tissues and patient prognosis. The underlying mechanism was associated with the Gli1-mediated EMT and EMT-related cell protrusion formation modulated by intracellular actin dynamic. Thus, KIF20B may be a potential biomarker and promising treatment target for CRC.
Collapse
Affiliation(s)
- Wen-Feng Lin
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China.,Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, China
| | - Xiao-Lu Lin
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China.,Department of Digestive Endoscopy, Fujian Provincial Hospital, Provincial Clinic Medical College, Fujian Medical University, Fuzhou, China
| | - Seng-Wang Fu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Li Yang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Chao-Tao Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Yun-Jie Gao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Hao-Yan Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhi-Zheng Ge
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
142
|
Hua Q, Mi B, Huang G. The emerging co-regulatory role of long noncoding RNAs in epithelial-mesenchymal transition and the Warburg effect in aggressive tumors. Crit Rev Oncol Hematol 2018; 126:112-120. [PMID: 29759552 DOI: 10.1016/j.critrevonc.2018.03.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/03/2018] [Accepted: 03/29/2018] [Indexed: 12/12/2022] Open
Abstract
Malignant tumor cells have several unique characteristics, and their ability to undergo epithelial-mesenchymal transition (EMT) is a molecular gateway to invasive behavior. Rapid proliferation and increased invasiveness during EMT enhance aberrant glucose metabolism in tumor cells. Meanwhile, aerobic glycolysis provides energy, biosynthesis precursors, and an appropriate microenvironment to facilitate EMT. Reciprocal crosstalk between the processes synergistically contributes to malignant cancer behaviors, but the regulatory mechanisms underlying this interaction remain unclear. Long non-coding RNAs (lncRNAs) are a recently recognized class of RNAs involved in multiple physiological and pathological tumor activities. Increasing evidence indicates that lncRNAs play overlapping roles in both EMT and cancer metabolism. In this review, we describe the lncRNAs reportedly involved in the two biological processes and explore the detailed mechanisms that could help elucidate this co-regulatory network and provide a theoretical basis for clinical management of EMT-related malignant phenotypes.
Collapse
Affiliation(s)
- Qian Hua
- Department of Nuclear Medicine, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Baoming Mi
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University (Wuxi 4th People's Hospital), Wuxi, Jiangsu, 214062, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
143
|
Leo C, Cotic C, Pomp V, Fink D, Varga Z. Overexpression of Lox in triple-negative breast cancer. Ann Diagn Pathol 2018; 34:98-102. [PMID: 29661738 DOI: 10.1016/j.anndiagpath.2018.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/24/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Triple negative breast cancer (TNBC) accounts for approximately 15% of breast cancers. It is associated with a poor prognosis and typically earlier onset of metastasis in comparison with other breast cancer subtypes. Since TNBC lacks the expression of estrogen and progesterone receptors and Her2 status is also negative, there is currently no target that can be used for systemic therapy. Epithelial-mesenchymal transition (EMT) plays an important role in tumor progression and metastasis. In this study, we examined a subset of EMT markers consisting of Snail, Twist-1 and Lox in TNBC and non-TNBC breast cancer subtypes and analyzed their expression pattern in regard to subtype, clinico-pathological parameters and prognosis. EXPERIMENTAL DESIGN We analyzed 659 breast cancer samples from two tissue microarrays. Breast cancer samples were categorized into two groups according to hormone receptor expression and Her2 status (n = 146 were triple negative, n = 513 were non triple-negative). Immunohistochemical expression of Snail, Twist-1 and Lox was semi-quantitatively analyzed using a three-tiered (weak-moderate-strong) scoring system. Results were statistically analyzed and correlated to clinico-pathological parameters and overall survival. RESULTS Strong overexpression of Lox was significantly higher in triple negative breast cancers when compared to non triple-negative breast cancers (p < 0.001). No difference was seen between the groups regarding Snail and Twist expression (p > 0.05). In addition, Lox expression was significantly stronger in poorly differentiated (G3) breast cancers (p < 0.001 for Lox). CONCLUSIONS The EMT marker Lox has a differential expression pattern in breast cancer, being significantly overexpressed in triple negative breast cancers. We could not link this expression to prognosis, however, this marker might be explored in future studies as possible target for systemic therapy of TNBC.
Collapse
Affiliation(s)
- Cornelia Leo
- Department of Gynecology, Kantonsspital Baden, Baden, Switzerland.
| | - Christine Cotic
- Department of Gynecology, University Hospital Zurich, 8010 Zurich, Switzerland.
| | - Victoria Pomp
- Department of Pathology and Molecularpathology, University Hospital Zurich, 8010 Zurich, Switzerland
| | - Daniel Fink
- Department of Gynecology, University Hospital Zurich, 8010 Zurich, Switzerland.
| | - Zsuzsanna Varga
- Department of Pathology and Molecularpathology, University Hospital Zurich, 8010 Zurich, Switzerland.
| |
Collapse
|
144
|
Qian H, Ding X, Zhang J, Mao F, Sun Z, Jia H, Yin L, Wang M, Zhang X, Zhang B, Yan Y, Zhu W, Xu W. Cancer stemness and metastatic potential of the novel tumor cell line K3: an inner mutated cell of bone marrow-derived mesenchymal stem cells. Oncotarget 2018; 8:39522-39533. [PMID: 28465472 PMCID: PMC5503629 DOI: 10.18632/oncotarget.17133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 02/20/2017] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) transplantation has been used for therapeutic applications in various diseases. Here we report MSCs can malignantly transform in vivo. The novel neoplasm was found on the tail of female rat after injection with male rat bone marrow-derived MSCs (rBM-MSCs) and the new tumor cell line, K3, was isolated from the neoplasm. The K3 cells expressed surface antigens and pluripotent genes similar to those of rBM-MSCs and presented tumor cell features. Moreover, the K3 cells contained side population cells (SP) like cancer stem cells (CSCs), which might contribute to K3 heterogeneity and tumorigenic capacity. To investigate the metastatic potential of K3 cells, we established the nude mouse models of liver and lung metastases and isolated the corresponding metastatic cell lines K3-F4 and K3-B6. Both K3-F4 and K3-B6 cell lines with higher metastatic potential acquired more mesenchymal and stemness-related features. Epithelial-mesenchymal transition is a potential mechanism of K3-F4 and K3-B6 formation.
Collapse
Affiliation(s)
- Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Xiaoqing Ding
- Faculty of Medical Laboratory Science, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Jiao Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Fei Mao
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Zixuan Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Haoyuan Jia
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Lei Yin
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Mei Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Bin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Yongmin Yan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Wei Zhu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, P. R. China
| |
Collapse
|
145
|
Liu GM, Zhang YM. Targeting FBPase is an emerging novel approach for cancer therapy. Cancer Cell Int 2018; 18:36. [PMID: 29556139 PMCID: PMC5845355 DOI: 10.1186/s12935-018-0533-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/05/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of death in both developed and developing countries. Metabolic reprogramming is an emerging hallmark of cancer. Glucose homeostasis is reciprocally controlled by the catabolic glycolysis and anabolic gluconeogenesis pathways. Previous studies have mainly focused on catabolic glycolysis, but recently, FBPase, a rate-limiting enzyme in gluconeogenesis, was found to play critical roles in tumour initiation and progression in several cancer types. Here, we review recent ideas and discoveries that illustrate the clinical significance of FBPase expression in various cancers, the mechanism through which FBPase influences cancer, and the mechanism of FBPase silencing. Furthermore, we summarize some of the drugs targeting FBPase and discuss their potential use in clinical applications and the problems that remain unsolved.
Collapse
Affiliation(s)
- Gao-Min Liu
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, No. 38 Huangtang Road, Meizhou, 514000 China
| | - Yao-Ming Zhang
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, No. 38 Huangtang Road, Meizhou, 514000 China
| |
Collapse
|
146
|
Yu J, Fang Q, Meng S. Knockdown of Long Noncoding RNA ENST457720 Inhibits Proliferation of Non-Small Cell Lung Cancer Cells In Vitro and In Vivo. Oncol Res 2018; 27:47-53. [PMID: 29495976 PMCID: PMC7848414 DOI: 10.3727/096504018x15193843443255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) represents the leading cause of cancer-related mortality worldwide. More and more reports have identified important roles for long noncoding RNAs (lncRNAs) in cancer development. ENST457720 expression was upregulated in lung adenocarcinoma in a microarray-based lncRNA screen. We determined the expression levels of ENST457720 in NSCLC tissues with quantitative real-time PCR and then studied their clinical significance. We explored the biological significance of ENST457720 with gain- and loss-of-function analyses in vitro and in vivo. In this study, ENST457720 was expressed at higher levels in NSCLC tissues than in paired normal tissues. Higher ENST457720 expression was associated with larger tumor sizes, lymph node metastasis, and advanced TNM stage. ENST457720 silencing suppressed NSCLC cell proliferation in vitro and in vivo. Moreover, ENST457720 knockdown inhibited NSCLC invasion and reversed the epithelial-to-mesenchymal transition. ENST457720 promoted NSCLC proliferation and invasion, which may be a novel potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Jia Yu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Qiyu Fang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Shuyan Meng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
147
|
Cui X, Li Z, Gao J, Gao PJ, Ni YB, Zhu JY. Elevated CXCL1 increases hepatocellular carcinoma aggressiveness and is inhibited by miRNA-200a. Oncotarget 2018; 7:65052-65066. [PMID: 27542259 PMCID: PMC5323138 DOI: 10.18632/oncotarget.11350] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023] Open
Abstract
In this study, we investigated the value of measurement of the chemokine CXCL1 in clinical management of hepatocellular carcinoma (HCC) and its possible role in the molecular pathogenesis of HCC. High CXCL1 expression predicted recurrence in HCC patients and promoted tumor progression in both in vivo and in vitro experimental systems. Overexpression of CXCL1 increased mitochondrial metabolism and activated the epithelial-to-mesenchymal transition (EMT). Using computational analysis we identified the microRNA miR-200a as a putative post-transcriptional regulator of CXCL1. We found that levels of miR-200a were inversely correlated with CXCL1 expression in HCC patient tissue samples by northern blot and qRT-PCR. Furthermore, CXCL1 was identified as a direct target which was bound and inhibited by miR- 200a. These findings provide new insights into the role of CXCL1 in HCC and its post-transcriptional regulation and suggest it may be a prognostic indicator for poor outcomes and a potential target for therapy.
Collapse
Affiliation(s)
- Xiao Cui
- Department of Hepatobilliary Surgery, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing 100044, China.,Department of General Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhao Li
- Department of Hepatobilliary Surgery, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing 100044, China
| | - Jie Gao
- Department of Hepatobilliary Surgery, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing 100044, China
| | - Peng-Ji Gao
- Department of Hepatobilliary Surgery, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing 100044, China
| | - Yan-Bing Ni
- Department of Hepatobilliary Surgery, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing 100044, China
| | - Ji-Ye Zhu
- Department of Hepatobilliary Surgery, Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
148
|
A novel non-canonical Wnt signature for prostate cancer aggressiveness. Oncotarget 2018; 8:9572-9586. [PMID: 28030815 PMCID: PMC5354754 DOI: 10.18632/oncotarget.14161] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/23/2016] [Indexed: 01/22/2023] Open
Abstract
Activation of the Canonical Wnt pathway (CWP) has been linked to advanced and metastatic prostate cancer, whereas the Wnt5a-induced non-canonical Wnt pathway (NCWP) has been associated with both good and poor prognosis. A newly discovered NCWP, Wnt5/Fzd2, has been shown to induce epithelial-to-mesenchymal transition (EMT) in cancers, but has not been investigated in prostate cancer. The aim of this study was to investigate if the CWP and NCWP, in combination with EMT, are associated with metabolic alterations, aggressive disease and biochemical recurrence in prostate cancer. An initial analysis was performed using integrated transcriptomics, ex vivo and in vivo metabolomics, and histopathology of prostatectomy samples (n=129), combined with at least five-year follow-up. This analysis detected increased activation of NCWP through Wnt5a/ Fzd2 as the most common mode of Wnt activation in prostate cancer. This activation was associated with increased expression of EMT markers and higher Gleason score. The transcriptional association between NCWP and EMT was confirmed in five other publicly available patient cohorts (1519 samples in total). A novel gene expression signature of concordant activation of NCWP and EMT (NCWP-EMT) was developed, and this signature was significantly associated with metastasis and shown to be a significant predictor of biochemical recurrence. The NCWP-EMT signature was also associated with decreased concentrations of the metabolites citrate and spermine, which have previously been linked to aggressive prostate cancer. Our results demonstrate the importance of NCWP and EMT in prostate cancer aggressiveness, suggest a novel gene signature for improved risk stratification, and give new molecular insight.
Collapse
|
149
|
Li S, Zhang HY, Du ZX, Li C, An MX, Zong ZH, Liu BQ, Wang HQ. Induction of epithelial-mesenchymal transition (EMT) by Beclin 1 knockdown via posttranscriptional upregulation of ZEB1 in thyroid cancer cells. Oncotarget 2018; 7:70364-70377. [PMID: 27683118 PMCID: PMC5342558 DOI: 10.18632/oncotarget.12217] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 09/02/2016] [Indexed: 12/21/2022] Open
Abstract
Beclin 1 has emerged as a haploinsufficient tumor suppression gene in a variety of human carcinomas. In order to clarify the role of Beclin 1 in thyroid cancer, Beclin 1 was knockdown in thyroid cancer cell lines. The current study demonstrated that knockdown of Beclin 1 resulted in morphological and molecular changes of thyroid cancer cells consistent with epithelial-mesenchymal transition (EMT), a morphogenetic procedure during which cells lose their epithelial characteristics and acquire mesenchymal properties concomitantly with gene expression reprogramming. In addition, the current study presented evidence demonstrating that Beclin 1 knockdown triggered this prometastatic process via stabilization of the EMT inducer ZEB1 mRNA through upregulation of AU-binding factor 1 (AUF1), which is recruited to the 3′-untranslated region (UTR) of the ZEB1 mRNA and decreases its degradation. We also found a negative correlation of Beclin 1 with AUF1 or ZEB1 in thyroid cancer tissues. These results indicated that at least some tumor suppressor functions of Beclin 1 were mediated through posttranscriptional regulation of ZEB1 via AUF1 in thyroid cancers.
Collapse
Affiliation(s)
- Si Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110001, China
| | - Hai-Yan Zhang
- Department of Geriatrics, the 1st Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Zhen-Xian Du
- Department of Endocrinology and Metabolism, the 1st Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Chao Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110001, China
| | - Ming-Xin An
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110001, China
| | - Zhi-Hong Zong
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110001, China
| | - Bao-Qin Liu
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110001, China
| | - Hua-Qin Wang
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang 110001, China
| |
Collapse
|
150
|
Li A, Gu Y, Li X, Sun H, Zha H, Xie J, Zhao J, Huang M, Chen L, Peng Q, Zhang Y, Weng Y, Zhou L. S100A6 promotes the proliferation and migration of cervical cancer cells via the PI3K/Akt signaling pathway. Oncol Lett 2018; 15:5685-5693. [PMID: 29552203 PMCID: PMC5840553 DOI: 10.3892/ol.2018.8018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/13/2017] [Indexed: 02/02/2023] Open
Abstract
Cervical cancer is the second most common gynecological cancer worldwide and remains one of the leading causes of cancer-associated mortality among women. S100A6 has been reported to be associated with the development of many types of cancer. The aim of the present study was to investigate the effect of S100A6 on the proliferation, apoptosis and migration of cervical cancer cells and its underlying molecular mechanisms. Quantative polymerase chain reaction (qPCR) was used to detect the basic mRNA level of S100A6 in HeLa, SiHa and CaSki cells. Western blot analysis was used to detect the protein level of S100A6, epithelial cadherin, neuronal cadherin, phosphorylated protein kinase B (p-Akt), t-Akt, p-glycogen synthase kinase 3β (GSK3β), t-GSK3β and β-catenin. Semi-qPCR was used to detect the mRNA level of Snail, Twist and Vimentin. MTT and Hoechst staining assays were used to detect the proliferation and apoptosis of cells, and wound healing and Transwell assays were used to detect the migration of cells. The results of the present study demonstrate that the levels of S100A6 were decreased in HeLa cells compared with in SiHa and CaSki cells. Overexpression of S100A6 in HeLa and CaSki cells promoted the proliferative and migratory ability, and had no significant effect on cellular apoptosis. Whereas the knockdown of S100A6 in SiHa and CaSki cells inhibited the proliferative and migratory ability, it had no significant effect on apoptosis. The overexpression of S100A6 in HeLa cells increased the levels of neuronal (N)-cadherin, vimentin, Snail and Twist. Conversely, knockdown of S100A6 in SiHa cells decreased the levels of N-cadherin, vimentin, Snail and Twist and increased the levels of epithelial (E)-cadherin. Furthermore, overexpression of S100A6 in HeLa cells activated the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway, and treatment with the PI3K inhibitor LY294002 partially repressed S100A6-enhanced proliferation and migration of cervical cancer cells. These results indicate that S100A6 facilitates the malignant potential of cervical cancer cells, particularly metastatic ability and epithelial-mesenchymal transition, which is mediated by activating the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Aifang Li
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yue Gu
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xueru Li
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hui Sun
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - He Zha
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jiaqing Xie
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jiali Zhao
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Mao Huang
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lu Chen
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qi Peng
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yan Zhang
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yaguang Weng
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lan Zhou
- Key Laboratory of Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|