1
|
Nadukkandy AS, Blaize B, Kumar CD, Mori G, Cordani M, Kumar LD. Non-coding RNAs as mediators of epithelial to mesenchymal transition in metastatic colorectal cancers. Cell Signal 2025; 127:111605. [PMID: 39842529 DOI: 10.1016/j.cellsig.2025.111605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/06/2025] [Accepted: 01/12/2025] [Indexed: 01/24/2025]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, necessitating the development of innovative treatment strategies. Recent research has underscored the significant role of non-coding RNAs (ncRNAs) in CRC pathogenesis, offering new avenues for diagnosis and therapy. In this review, we delve into the intricate roles of various ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in CRC progression, epithelial-mesenchymal transition (EMT), metastasis, and drug resistance. We highlight the interaction of these ncRNAs with and regulation of key signaling pathways, such as Wnt/β-catenin, Notch, JAK-STAT, EGFR, and TGF-β, and the functional relevance of these interactions in CRC progression. Additionally, the review highlights the emerging applications of nanotechnology in enhancing the delivery and efficacy of ncRNA-based therapeutics, which could address existing challenges related to specificity and side effects. Future research directions, including advanced diagnostic tools, targeted therapeutics, strategies to overcome drug resistance, and the integration of personalized medicine approaches are discussed. Integrating nanotechnology with a deeper understanding of CRC biology offers the potential for more effective, targeted, and personalized strategies, though further research is essential to validate these approaches.
Collapse
Affiliation(s)
- Aisha Shigna Nadukkandy
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Britny Blaize
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Chethana D Kumar
- Department of Surgical ICU, Christian Medical College, IDA Scudder Road, Vellore 632004, Tamil Nadu, India
| | - Giulia Mori
- Department Of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Lekha Dinesh Kumar
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India.
| |
Collapse
|
2
|
Han M, Zhou X, Cheng H, Qiu M, Qiao M, Geng X. Chitosan and hyaluronic acid in colorectal cancer therapy: A review on EMT regulation, metastasis, and overcoming drug resistance. Int J Biol Macromol 2025; 289:138800. [PMID: 39694373 DOI: 10.1016/j.ijbiomac.2024.138800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/04/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Up to 90% of cancer-related fatalities could be attributed to metastasis. Therefore, understanding the mechanisms that facilitate tumor cell metastasis is beneficial for improving patient survival and results. EMT is considered the main process involved in the invasion and spread of CRC. Essential molecular components like Wnt, TGF-β, and PI3K/Akt play a role in controlling EMT in CRC, frequently triggered by various factors such as Snail, Twist, and ZEB1. These factors affect not only the spread of CRC but also determine the reaction to chemotherapy. The influence of non-coding RNAs, especially miRNAs and lncRNAs, on the regulation of EMT is clear in CRC. Exosomes, involved in cell-to-cell communication, can affect the TME and metastasis of CRC. Pharmacological substances and nanoparticles demonstrate promise as efficient modulators of EMT in CRC. Chitosan and HA are two major carbohydrate polymers with considerable potential in inhibiting CRC. Chitosan and HA can be employed to modify nanoparticles to enhance cargo transport for reducing CRC. Additionally, chitosan and HA-modified nanocarriers, which can be utilized as potential approaches in suppressing EMT and reversing drug resistance in CRC, can inhibit EMT and chemoresistance, crucial components in tumorigenesis.
Collapse
Affiliation(s)
- Mingming Han
- Department of Pharmacy and Medical Devices, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Xi Zhou
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Hang Cheng
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Mengru Qiu
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Meng Qiao
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Xiao Geng
- Department of Party Committee Office, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| |
Collapse
|
3
|
Zhang Q, Yu M, Yang L, Sun D. MiR-875-5p suppresses Gli1 to alter the hedgehog signaling pathway, which in turn has hepatocellular cancer-related tumor suppressing properties. Heliyon 2024; 10:e37771. [PMID: 39381215 PMCID: PMC11459020 DOI: 10.1016/j.heliyon.2024.e37771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Background One of the most prevalent cancers worldwide is HCC, which has put patient health at risk. Increasing evidence indicated that messenger RNAs (mRNAs) played significant roles in modulating tumorigenesis. It has been established that Gli1 acts as an oncogene in a number of malignancies. However, more research was necessary to understand the Gli1 regulation mechanism in HCC. Methods Microarray technology was used to evaluate the expression of mRNAs. RT-qPCR was utilized to evaluate Gli1 and miR-875-5p expression. To investigate the role of Gli1, tests using CCK-8, EdU, transwell, immunofluorescence, and Western blot analysis was performed. RIP, RNA pull down, and luciferase reporter assays were employed to verify the interaction between Gli1 and miR-875-5p. Results In tissues and cells of HCC, Gli1 expression appeared to be upregulated, especially in metastatic samples and advanced stages of the disease. A worse outcome was predicted by elevated Gli1 expression. Additionally, in HCC, Gli1 inhibition impeded the growth, migration, and development of the EMT. Since miR-875-5p was shown to have a molecular target in Gli1, miR-875-5p mediated the negative regulation of Gli1. In HCC tissues, its expression pattern was less prominent. In HCC tissues, there was an inverse relationship between Gli1 expression and miR-875-5p expression. Overexpressing Gli1 helped to partially counteract the suppression of HCC migration, proliferation, and EMT formation by miR-875-5p overexpression. Conclusions MiR-875-5p in HCC suppresses tumors by downregulating Gli1, which supplies a novel treatment for HCC patients.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Pathology, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| | - Miao Yu
- Department of Clinical Laboratory, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| | - Leilei Yang
- Department of Hepatobiliary Surgery, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| | - Defeng Sun
- Department of Hepatobiliary Surgery, Central Hospital Affiliated to Jiangnan University, Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu Province, China
| |
Collapse
|
4
|
Shibui Y, Kohashi K, Hino Y, Tamaki A, Kinoshita I, Yamamoto H, Nakashima Y, Tajiri T, Oda Y. Expression of Forkhead Box M1 and Anticancer Effects of FOXM1 Inhibition in Epithelioid Sarcoma. J Transl Med 2024; 104:102093. [PMID: 38857782 DOI: 10.1016/j.labinv.2024.102093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
Epithelioid sarcoma (ES) is a rare aggressive sarcoma that, unlike most soft-tissue sarcomas, shows a tendency toward local recurrence and lymph node metastasis. Novel antitumor agents are needed for ES patients. Forkhead box transcription factor 1 (FOXM1) is a member of the Forkhead transcription factor family and is associated with multiple oncogenic functions; FOXM1 is known to be overexpressed and correlated with pathogenesis in various malignancies. In this study, we immunohistochemically analyzed FOXM1 expression levels and their clinical, clinicopathologic, and prognostic significance in 38 ES specimens. In addition, to investigate potential correlations between FOXM1 downregulation and oncologic characteristics, we treated ES cell lines with thiostrepton, a naturally occurring antibiotic that inhibits both small interfering RNA (siRNA) and FOXM1. In the analyses using ES samples, all 38 specimens were diagnosed as positive for FOXM1 by immunohistochemistry. We separated specimens into high (n = 19) and low (n = 19) FOXM1-protein expression groups by staining index score, and into large (n = 12), small (n = 25), and unknown (n = 1) tumor-size groups using a cutoff of 5 cm maximum diameter. Although there were significantly more samples with high FOXM1 expression in the large tumor group (P = .013), there were no significant differences with respect to age (P = 1.00), sex (P = .51), primary site of origin (P = .74), histologic subtypes (P = 1.00), depth (P = .74), or survival rate (P = .288) between the high and low FOXM1-protein expression groups. In the in vitro experiments using ES cell lines, FOXM1 siRNA and thiostrepton successfully downregulated FOXM1 mRNA and protein expression. Furthermore, downregulation of FOXM1 inhibited cell proliferation, drug resistance against chemotherapeutic agents, migration, and invasion and caused cell cycle arrest in the ES cell lines. Finally, cDNA microarray analysis data showed that FOXM1 regulated cIAP2, which is one of the apoptosis inhibitors activated by the TNFα-mediated NF-κB pathway. In conclusion, the FOXM1 gene may be a promising therapeutic target for ES.
Collapse
Affiliation(s)
- Yuichi Shibui
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Pediatric Surgery, Faculty of Medicine, University of Tsukuba Hospital, Ibaraki, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Pathology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yuko Hino
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihiko Tamaki
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Izumi Kinoshita
- Department of Pathology, Kokura Memorial Hospital, Fukuoka, Japan
| | - Hidetaka Yamamoto
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
5
|
Wang S, Huang X, Zhang G, Chen Z, Guan H, Zhou W. Tumor suppressor miR-361-3p inhibits prostate cancer progression through Gli1 and AKT/mTOR signaling pathway. Cell Signal 2024; 114:110998. [PMID: 38048859 DOI: 10.1016/j.cellsig.2023.110998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND The primary challenge in prostate cancer (PCa) is tumor metastasis, which seriously affects the survival time of patients. Growing evidence suggests that microRNAs play a crucial regulatory role in various malignancies and that the tumor suppressor miR-361-3p is responsible for regulating migration, proliferation, and invasion in different cancer types. However, the underlying regulatory mechanism of miR-361-3p in PCa remains unknown. METHODS The expression of miR-361-3p in PCa cells was analyzed using quantitative real time-polymerase chain reaction. The clinical utility of miR-361-3p in PCa was evaluated using in vitro assays. The mechanism of action of miR-361-3p was investigated using western blotting, luciferase reporter assays, immunofluorescence, and rescue studies. RESULTS The function, invasiveness, migration, and proliferation of PCa cells, as well as epithelial-mesenchymal transition (EMT), were aided by the downregulation of miR-361-3p, whereas its overexpression exerted the opposite effect. Repression of glioma-associated oncogene homolog 1 (Gli1) expression by miR-361-3p led to activation of the protein kinase B/mammalian target of rapamycin (AKT/mTOR) signaling pathway, triggering EMT and promoting PCa metastasis. CONCLUSIONS Downregulation of miR-361-3p along the Gli1 axis promoted tumor malignancy. Collectively, the results of this study imply that miR-361-3p has the potential to be both a biomarker and therapeutic target in PCa.
Collapse
Affiliation(s)
- Shaocheng Wang
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province 233099, China
| | - Xiaoyu Huang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province 233099, China
| | - Guangyu Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province 233099, China
| | - Zhijun Chen
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province 233099, China
| | - Han Guan
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province 233099, China.
| | - Wensheng Zhou
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui Province 233099, China.
| |
Collapse
|
6
|
Liu BW, Cao JL, Wang Y, Zhao X, Zeng Q, Liu WP, Zhang JH, Fan YZ, Dou J. GANT61, an inhibitor of Gli1, inhibits the proliferation and migration of hepatocellular carcinoma cells. J Investig Med 2024; 72:181-192. [PMID: 37724700 DOI: 10.1177/10815589231204056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Constitutive activation of Hedgehog (Hh) signaling has been implicated in many cancers including hepatocellular carcinoma (HCC). Among them, the terminal glioma-associated oncogene homolog 1 (Gli1) regulates the expression of critical genes in the Hh pathway. The current study aims to evaluate the anti-HCC effect of the Gli1 inhibitor, GANT61. In vitro analysis including cell counting kit-8 (CCK-8) assay, flow cytometry, and migration and invasion assay were adopted to evaluate the effect of GANT61 on HCC cell lines. In vivo, xenograft studies were also performed to verify the effect of GANT61 on HCC. By CCK-8 assay, we found that GANT61 could significantly reduce the growth of HCC cell lines Huh7 and hemophagocytic lymphohistiocytosis (HLE), and their IC50 concentrations were 4.481 and 6.734 μM, respectively. Flow cytometry shows that GANT61 induced cell cycle arrest in the G2/M phase and accelerated apoptosis of both HLE and Huh7 cells. While migration and invasion assay shows that GANT61 weakens cells' migration and invasion ability. Besides that, GANT61 inhibits the expression of Gli1, FoxM1, CyclinD1, and Bcl-2, upregulates the level of Bax protein, and also reverses the epithelial-mesenchymal transition program by downregulating the expression of Vimentin and N-Cadherin and upregulating the expression of epithelial E-Cadherin expression. Furthermore, GANT61 inhibits the growth of subcutaneous xenografts of Huh7 cells in nude mice. Overall, this study suggests that Gli1 is a potential target for therapy and GANT61 shows promising therapeutic potential for future treatment in HCC.
Collapse
Affiliation(s)
- Bao-Wang Liu
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jing-Lin Cao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yang Wang
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xin Zhao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Qiang Zeng
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Wen-Peng Liu
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jun-Hong Zhang
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yi-Ze Fan
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jian Dou
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
7
|
Li X, Li X, Liu G, Zhou L, Liu Y, Dou T, Chen X, Wang J. ANP32A Knockdown Attenuates the Malignant Biological Behavior of Colorectal Cancer Cells by Suppressing Epithelial-mesenchymal Transition and ERK Activation. J Cancer 2023; 14:2759-2770. [PMID: 37781083 PMCID: PMC10539559 DOI: 10.7150/jca.84687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/22/2023] [Indexed: 10/03/2023] Open
Abstract
Acidic leucine rich nuclear phosphoprotein-32A (ANP32A) protein has a variety of functions, such as regulating cell differentiation, influencing cell apoptosis and cell cycle progression. Our previous study demonstrated that high expression of ANP32A was found in the tumor tissues of colorectal cancer (CRC) patients and was positively associated with tumor grading. However, the function and underlying mechanisms of ANP32A in CRC metastasis have not been fully explored. In this study, we found that ANP32A knockdown significantly attenuated the migration and invasion, and epithelial-mesenchymal transition (EMT) in cells. Further mechanistic studies revealed that ANP32A knockdown inhibited the expression of β-catenin and phosphorylated-ERK. The immunofluorescent staining experiment has revealed that ANP32A was expressed in the cell membrane, cytosol and nucleus, and its expression was positively associated with β-catenin expression levels. Moreover, the ability of cell migration and invasion was inhibited, the expression of E-cadherin was enhanced following ANP32A knockdown, and these affects were abolished by an ERK activator PMA, enhanced by an ERK inhibitor PD98059. Moreover, our animal experiment also demonstrated that silenced ANP32A inhibited CRC cell growth, multi-organ metastasis, ERK activation and EMT progression in vivo. Collectively, these findings demonstrated that ANP32A promotes CRC progression and that may be a promising target for the anti-metastasis treatment of CRC.
Collapse
Affiliation(s)
- Xiaojuan Li
- Department of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, China
| | - Xumei Li
- Department of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, China
| | - Guoxiang Liu
- Department of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, China
| | - Luwei Zhou
- Department of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, China
| | - Yisa Liu
- Department of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, China
| | - Tong Dou
- Department of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, China
- School of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China
| | - Xu Chen
- Department of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541199, China
| | - Juan Wang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541001, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541001, China
- Faculty of Basic Medicine, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region 541004, China
| |
Collapse
|
8
|
Iyer AS, Shaik MR, Raufman JP, Xie G. The Roles of Zinc Finger Proteins in Colorectal Cancer. Int J Mol Sci 2023; 24:10249. [PMID: 37373394 DOI: 10.3390/ijms241210249] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Despite colorectal cancer remaining a leading worldwide cause of cancer-related death, there remains a paucity of effective treatments for advanced disease. The molecular mechanisms underlying the development of colorectal cancer include altered cell signaling and cell cycle regulation that may result from epigenetic modifications of gene expression and function. Acting as important transcriptional regulators of normal biological processes, zinc finger proteins also play key roles in regulating the cellular mechanisms underlying colorectal neoplasia. These actions impact cell differentiation and proliferation, epithelial-mesenchymal transition, apoptosis, homeostasis, senescence, and maintenance of stemness. With the goal of highlighting promising points of therapeutic intervention, we review the oncogenic and tumor suppressor roles of zinc finger proteins with respect to colorectal cancer tumorigenesis and progression.
Collapse
Affiliation(s)
- Aishwarya S Iyer
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mohammed Rifat Shaik
- Department of Medicine, University of Maryland Medical Center Midtown Campus, Baltimore, MD 21201, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Guofeng Xie
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
9
|
Li H, Wang H, Cui Y, Jiang W, Zhan H, Feng L, Gao M, Zhao K, Zhang L, Xie X, Zhao N, Li Y, Liu P. EZH2 regulates pancreatic cancer cells through E2F1, GLI1, CDK3, and Mcm4. Hereditas 2023; 160:23. [PMID: 37198697 DOI: 10.1186/s41065-023-00280-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 04/06/2023] [Indexed: 05/19/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most common malignant tumors in digestive tract. To explore the role of epigenetic factor EZH2 in the malignant proliferation of PC, so as to provide effective medical help in PC. Sixty paraffin sections of PC were collected and the expression of EZH2 in PC tissues was detected by immunohistochemical assay. Three normal pancreas tissue samples were used as controls. The regulation of EZH2 gene on proliferation and migration of normal pancreatic cell and PC cell were determined by MTS, colony forming, Ki-67 antibody, scratch and Transwell assays. Through differential gene annotation and differential gene signaling pathway analysis, differentially expressed genes related to cell proliferation were selected and verified by RT-qPCR. EZH2 is mainly expressed in the nuclei of pancreatic tumor cells, but not in normal pancreatic cells. The results of cell function experiments showed that EZH2 overexpression could enhance the proliferation and migration ability of PC cell BXPC-3. Cell proliferation ability increased by 38% compared to the control group. EZH2 knockdown resulted in reduced proliferation and migration ability of cells. Compared with control, proliferation ability of cells reduced by 16%-40%. The results of bioinformatics analysis of transcriptome data and RT-qPCR demonstrated that EZH2 could regulate the expression of E2F1, GLI1, CDK3 and Mcm4 in normal and PC cells. The results revealed that EZH2 might regulate the proliferation of normal pancreatic cell and PC cell through E2F1, GLI1, CDK3 and Mcm4.
Collapse
Affiliation(s)
- Hongfeng Li
- Department of Clinical Laboratory, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Hailong Wang
- Department of Oncology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, No. 354 Beima Road, Hongqiao District, Tianjin, 300120, China
| | - Yunlong Cui
- Department of Hepatobiliary Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Wenhua Jiang
- Department of Radiotherapy, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hongjie Zhan
- Department of Gastric Cancer, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Lixia Feng
- Department of Nursing, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Konggang Hospital, Tianjin, 300300, China
| | - Mingyou Gao
- Department of Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Kuo Zhao
- Department of Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Limeng Zhang
- Department of Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xiaojing Xie
- Department of Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Ning Zhao
- Department of Oncology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Ying Li
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, No. 12 Health Road, Shijiazhuang, 050000, Hebei, China.
| | - Pengfei Liu
- Department of Oncology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, No. 354 Beima Road, Hongqiao District, Tianjin, 300120, China.
| |
Collapse
|
10
|
Xu M, Wang J, Li H, Zhang Z, Cheng Z. AIM2 inhibits colorectal cancer cell proliferation and migration through suppression of Gli1. Aging (Albany NY) 2020; 13:1017-1031. [PMID: 33291082 PMCID: PMC7835022 DOI: 10.18632/aging.202226] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022]
Abstract
Colorectal cancer (CRC) is a common malignant tumor and is one of the leading causes of cancer-related deaths worldwide. Absent in melanoma 2 (AIM2), as a member of the pyrin-HIN family proteins, plays contentious roles in different types of cancers. In the present work, we provide evidence that AIM2 was commonly downregulated in human CRC and loss of AIM2 significantly correlated with tumor size, depth of invasion, lymph node metastasis (LNM) and TNM (Tumor, Node, Metastases) stage in patients suffering from CRC. AIM2 knockdown promoted CRC cell proliferation, migration and epithelial-mesenchymal transition (EMT) progress, whereas AIM2 overexpression did the opposite. AIM2 inhibited glioma-associated oncogene-1 (Gli1) expression through Smoothened homolog (SMO)-independent pathway and regulated CRC cell proliferation and migration in a Gli1-dependent manner. Moreover, AIM2 could modulate Protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) signaling pathway and the increased Gli1 expression and EMT progress induced by AIM2 depletion was reversed after incubation with AKT inhibitor Ly294002 in CRC cells. In conclusion, our results define AIM2 as a novel regulator of Gli1 in CRC cell growth and metastasis, and suggest that the AIM2/AKT/mTOR/Gli1 signaling axis may serve as a potential target for treatment of CRC.
Collapse
Affiliation(s)
- Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| | - Junfeng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| | - Haoran Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| | - Zhengrong Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| |
Collapse
|
11
|
Shibui Y, Kohashi K, Tamaki A, Kinoshita I, Yamada Y, Yamamoto H, Taguchi T, Oda Y. The forkhead box M1 (FOXM1) expression and antitumor effect of FOXM1 inhibition in malignant rhabdoid tumor. J Cancer Res Clin Oncol 2020; 147:1499-1518. [PMID: 33221995 DOI: 10.1007/s00432-020-03438-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Malignant rhabdoid tumor (MRT) is a rare, highly aggressive sarcoma with an uncertain cell of origin. Despite the existing standard of intensive multimodal therapy, the prognosis of patients with MRT is very poor. Novel antitumor agents are needed for MRT patients. Forkhead box transcription factor 1 (FOXM1) is overexpressed and is correlated with the pathogenesis in several human malignancies. In this study, we identified the clinicopathological and prognostic values of the expression of FOXM1 and its roles in the progression of MRT. METHODS We investigated the FOXM1 expression levels and their clinical significance in 23 MRT specimens using immunohistochemistry and performed clinicopathologic and prognostic analyses. We also demonstrated correlations between the downregulation of FOXM1 and oncological characteristics using small interfering RNA (siRNA) and FOXM1 inhibitor in MRT cell lines. RESULTS Histopathological analyses revealed that primary renal MRTs showed significantly low FOXM1 protein expression levels (p = 0.032); however, there were no significant differences in other clinicopathological characteristics or the survival rate. FOXM1 siRNA and FOXM1 inhibitor (thiostrepton) successfully downregulated the mRNA and protein expression of FOXM1 in vitro and the downregulation of FOXM1 inhibited cell proliferation, drug resistance to chemotherapeutic agents, migration, invasion, and caused the cell cycle arrest and apoptosis of MRT cell lines. A cDNA microarray analysis showed that FOXM1 regulated FANCD2 and NBS1, which are key genes for DNA damage repair. CONCLUSION This study demonstrates that FOXM1 may serve as a promising therapeutic target for MRT.
Collapse
Affiliation(s)
- Yuichi Shibui
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akihiko Tamaki
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Izumi Kinoshita
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology Graduate School of Medical Sciences, Kyushu University, Maidashi3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
12
|
Xu M, Shao X, Li H, Zhang Z, Zhou C, Cheng Z. Clinical value and potential association of Rab1A and FoxM1 aberrant expression in colorectal cancer. Sci Rep 2020; 10:20160. [PMID: 33214609 PMCID: PMC7678875 DOI: 10.1038/s41598-020-77182-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 11/05/2020] [Indexed: 11/21/2022] Open
Abstract
Colorectal carcinoma (CRC) is one of the most common malignancies with a dismal 5-year survival rate. Our recent study indicated that Rab1A expression was closely related to GLI1 expression. A previous study shows that aberrant overexpression of GLI1 promotes colorectal cancer metastasis via FoxM1 overexpression. However, the potential correlation between Rab1A and FoxM1 in CRC remains elusive. Immunohistochemistry was performed to investigate the association of the expression of Rab1A and FoxM1 and to determine the prognosis in 135 CRC tissue and adjacent normal tissues. Using Oncomine datasets, we found that Rab1A and FoxM1 mRNA were obviously upregulated in CRC tissues compared to normal tissues. Additionally, the expression of Rab1A and FoxM1 was significantly higher in CRC tissues than that in normal tissues. Rab1A expression was positively correlated with FoxM1 expression in CRC, especially in TNM stage III. In addition, Rab1A and FoxM1 overexpression was found to be significantly correlated with poor prognosis in CRC patients. Besides, both high expression of Rab1A and FoxM1 led to a worse prognosis than anyone low group, and both low expression of Rab1A and FoxM1 had a better prognosis than the anyone low group. Therefore, Rab1A and FoxM1 play crucial roles and could be used as clinical biomarkers in CRC.
Collapse
Affiliation(s)
- Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Xinyu Shao
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, No.242 Guangji Road, Suzhou, 215006, Jiangsu Province, China
| | - Haoran Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui Province, China
| | - Zhengrong Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui Province, China
| | - Chunli Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, No.242 Guangji Road, Suzhou, 215006, Jiangsu Province, China.
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui Province, China.
| |
Collapse
|
13
|
Chang Y, Chen H, Duan J, Wu W, Le F, Mou F. The inhibitory effect and safety of GANT61 on HeLa cells in nude mice. Exp Mol Pathol 2019; 113:104352. [PMID: 31809711 DOI: 10.1016/j.yexmp.2019.104352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 11/19/2019] [Accepted: 12/03/2019] [Indexed: 12/25/2022]
Abstract
The Hedgehog (Hh) pathway effector Gli1 plays an important role in cervical cancer, and GANT61 is an Hh signaling inhibitor. In this study, we aimed to investigate the inhibitory effect of GANT61 on cervical cancer and to study its safety in nude mice. We used in vivo experiments to assess the effect of GANT61 on the growth of cervical cancer HeLa cells, and we measured the WBC, HGB, PLT, ALT, AST and Cre levels in nude mice. Next, we examined the organ and tumor morphology and distant metastasis by HE staining. We used immunohistochemistry to monitor the expression levels of Gli1, FoxM1, Ki-67, cyclinD1, E-cadherin, vimentin, survivin, caspase-3 and CD34+. Western blotting and RT-RCR were used to measure Gli1 expression. GANT61 inhibited the growth and metastasis of HeLa cervical cancer cells upon their transplantation into nude mice, and we preliminarily propose that GANT61 is safe for nude mice. These findings suggest that GANT61 could be used as a Hedgehog inhibitor to inhibit EMT and proliferation and to promote apoptosis via Gli1 downregulation.
Collapse
Affiliation(s)
- Yanan Chang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, PR China
| | - Hong Chen
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, PR China.
| | - Jie Duan
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, PR China
| | - Wang Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, PR China
| | - Fangshu Le
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, PR China
| | - Fen Mou
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, PR China
| |
Collapse
|
14
|
Expression analysis and implication of Rab1A in gastrointestinal relevant tumor. Sci Rep 2019; 9:13384. [PMID: 31527621 PMCID: PMC6746845 DOI: 10.1038/s41598-019-49786-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 08/31/2019] [Indexed: 12/20/2022] Open
Abstract
Gastrointestinal cancers have become increasingly prevalent worldwide. Previous studies have reported an oncogenic function of Rab1A in colorectal cancer and hepatocellular carcinomas via the mTOR pathway. However, the exact role of Rab1A in gastrointestinal cancers remains elusive. We detected significantly higher expression of Rab1A in the gastrointestinal tumor tissues compared to that in other cancer types following an in silico analysis of TGCA and GTEX databases. Furthermore, Rab1A was overexpressed in the gastrointestinal tumor tissues compared to the para-tumor tissues. Although Rab1A expression levels were not associated with the tumor-lymph node-metastasis (TNM) stage, Rab1A overexpression in the tumor tissues of a gastric cancer (GC) cohort was strongly correlated with poor prognosis in the patients. In addition, Rab1A knockdown significantly inhibited the in vitro proliferation and migration abilities of GC cells, as well as the growth of GC xenografts in vivo. Furthermore, a positive correlation was observed between Rab1A expression levels and that of different upstream/downstream mTOR targets. Taken together, Rab1A regulates the PI3K-AKT-mTORC1 pathway through the mTORC1 complex consisting of mTORC1, Rheb and Rab1A, and is a promising therapeutic target in GC.
Collapse
|
15
|
Lin J, Tan H, Nie Y, Wu D, Zheng W, Lin W, Zhu Z, Yang B, Chen X, Chen T. Krüppel-like factor 2 inhibits hepatocarcinogenesis through negative regulation of the Hedgehog pathway. Cancer Sci 2019; 110:1220-1231. [PMID: 30719823 PMCID: PMC6447955 DOI: 10.1111/cas.13961] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. The most important reason for the occurrence of HCC is hepatitis C or B infection. Moreover, genetic factors play an important role in the tumorigenesis of HCC. Here, we demonstrated that Krüppel-like factor 2 (KLF2) expression was downregulated in HCC samples compared with adjacent tissues. Additionally, KLF2 was shown to inhibit the growth, migration and colony-formation ability of liver cancer cells. Further mechanistic studies revealed that KLF2 can compete with Gli1 for interaction with HDAC1 and restrains Hedgehog signal activation. Together, our results suggest that KLF2 has potential as a diagnostic biomarker and therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- JinBo Lin
- Longgang Central Hospital of Shenzhen, Affiliated Longgang Hospital of Zunyi Medical University, Shenzhen, China
| | - Huifang Tan
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingjie Nie
- Clinical Research Lab Center, Guizhou Provincial People's Hospital, Guiyang, China
| | - Dongwen Wu
- The Third Xiangya Hospital, Central South University, Changsha, China
| | - Weiji Zheng
- Longgang Central Hospital of Shenzhen, Affiliated Longgang Hospital of Zunyi Medical University, Shenzhen, China
| | - Wensong Lin
- Longgang Central Hospital of Shenzhen, Affiliated Longgang Hospital of Zunyi Medical University, Shenzhen, China
| | - Zheng Zhu
- Longgang Central Hospital of Shenzhen, Affiliated Longgang Hospital of Zunyi Medical University, Shenzhen, China
| | - Bing Yang
- Longgang Central Hospital of Shenzhen, Affiliated Longgang Hospital of Zunyi Medical University, Shenzhen, China
| | - Xiaoliang Chen
- Shenzhen Guangming District Center for Disease Control and Prevention, Guangdong, China
| | - Tao Chen
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Clinical Research Lab Center, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
16
|
Yao Y, Zhou D, Shi D, Zhang H, Zhan S, Shao X, Sun K, Sun L, Wu G, Tian K, Zhu X, He S. GLI1 overexpression promotes gastric cancer cell proliferation and migration and induces drug resistance by combining with the AKT-mTOR pathway. Biomed Pharmacother 2019; 111:993-1004. [PMID: 30841479 DOI: 10.1016/j.biopha.2019.01.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/05/2019] [Accepted: 01/06/2019] [Indexed: 02/07/2023] Open
Abstract
Hedgehog (HH) pathway significantly affected the pathogenesis of Gastric cancer (GC), but the multiple uncanonical HH pathways that are mediated by Zinc Finger protein GLI1 (GLI1) are still unclear. In the present work, we evaluated GLI1 and p-AKT expression in GC using immunohistochemistry (IHC) analysis. GLI1 and AKT specific shRNA was transfected into GC cell lines to investigate the cross-regulation between HH pathway and AKT-mTOR pathway. The effect of GLI1 and p-AKT on proliferation, migration, and drug resistance were examined. Moreover, a mouse xenograft model of GC was established to verify the role of GLI1 and p-AKT in promoting drug sensitivity in vivo. Our results suggested the clinicopathological factors and prognosis by the differential expression of GLI1 and p-AKT in GC patients. GLI1 was activated by the AKT-mTOR pathway. Co-expression of GLI1 and p-AKT was associated with cell viability, migration, and drug resistance and indicated a poor prognosis in GC patients. Agents targeted against both GLI1 and p-AKT may reverse drug-resistance and achieve better inhibition than agents targeted against a single molecule. There was a significant correlation between the high expression of GLI1 and p-AKT in GC. Additionally, our study confirmed the activity of the AKT-mTOR-GLI1 axis, which provided a new viable field for GC treatment.
Collapse
Affiliation(s)
- Yizhou Yao
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Diyuan Zhou
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Dongtao Shi
- Departments of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Hui Zhang
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China; Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, PR China
| | - Shenghua Zhan
- Departments of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Xinyu Shao
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215006, PR China
| | - Kang Sun
- Department of General Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, PR China
| | - Liang Sun
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Guangting Wu
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Kangjun Tian
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Xinguo Zhu
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
| | - Songbing He
- Departments of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
17
|
Laissue P. The forkhead-box family of transcription factors: key molecular players in colorectal cancer pathogenesis. Mol Cancer 2019; 18:5. [PMID: 30621735 PMCID: PMC6325735 DOI: 10.1186/s12943-019-0938-x] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/01/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly occurring cancer worldwide and the fourth most frequent cause of death having an oncological origin. It has been found that transcription factors (TF) dysregulation, leading to the significant expression modifications of genes, is a widely distributed phenomenon regarding human malignant neoplasias. These changes are key determinants regarding tumour’s behaviour as they contribute to cell differentiation/proliferation, migration and metastasis, as well as resistance to chemotherapeutic agents. The forkhead box (FOX) transcription factor family consists of an evolutionarily conserved group of transcriptional regulators engaged in numerous functions during development and adult life. Their dysfunction has been associated with human diseases. Several FOX gene subgroup transcriptional disturbances, affecting numerous complex molecular cascades, have been linked to a wide range of cancer types highlighting their potential usefulness as molecular biomarkers. At least 14 FOX subgroups have been related to CRC pathogenesis, thereby underlining their role for diagnosis, prognosis and treatment purposes. This manuscript aims to provide, for the first time, a comprehensive review of FOX genes’ roles during CRC pathogenesis. The molecular and functional characteristics of most relevant FOX molecules (FOXO, FOXM1, FOXP3) have been described within the context of CRC biology, including their usefulness regarding diagnosis and prognosis. Potential CRC therapeutics (including genome-editing approaches) involving FOX regulation have also been included. Taken together, the information provided here should enable a better understanding of FOX genes’ function in CRC pathogenesis for basic science researchers and clinicians.
Collapse
Affiliation(s)
- Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 N° 63C-69, Bogotá, Colombia.
| |
Collapse
|
18
|
Park S, Kim H, Kim K, Roh S. Sonic hedgehog signalling regulates the self-renewal and proliferation of skin-derived precursor cells in mice. Cell Prolif 2018; 51:e12500. [PMID: 30151845 PMCID: PMC6528853 DOI: 10.1111/cpr.12500] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The sonic hedgehog (Shh) signalling pathway has an important role in the maintenance of various stem cells and organogenesis during development. However, the effect of Shh in skin-derived precursors (SKPs), which have the capacity for multipotency and self-renewal, is not yet clear. The present study investigated the effects of the Shh signalling pathway on the proliferation and self-renewal of murine SKPs (mSKPs). METHODS The Shh signalling pathway was activated by treatment with purmorphamine (Shh agonist) or recombinant Shh in mSKPs. Cyclopamine (Shh antagonist) or GANT-61 (Gli inhibitor) was used to inhibit the pathway. Western blot, qPCR, and immunofluorescence were used to analyse the expression of genes related to self-renewal, stemness, epithelial-mesenchymal transition (EMT) and the Shh signalling pathway. In addition, cell proliferation and apoptosis were examined. RESULTS Inhibiting the Shh signalling pathway reduced mSKP proliferation and sphere formation, but increased apoptosis. Activating this signalling pathway produced opposite results. The Shh signalling pathway also controlled the EMT phenotype in mSKPs. Moreover, purmorphamine recovered the self-renewal and proliferation of aged mSKPs. CONCLUSION Our results suggest that the Shh signalling pathway has an important role in the proliferation, self-renewal and apoptosis of mSKPs. These findings also provide a better understanding of the cellular mechanisms underlying SKP self-renewal and apoptosis that allow more efficient expansion of SKPs.
Collapse
Affiliation(s)
- Sangkyu Park
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| | - Hyewon Kim
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| | - Kichul Kim
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology LaboratoryDental Research Institute, BK21, Seoul National University School of DentistrySeoulKorea
| |
Collapse
|
19
|
Yu X, Zheng Y, Zhu X, Gao X, Wang C, Sheng Y, Cheng W, Qin L, Ren N, Jia H, Dong Q. Osteopontin promotes hepatocellular carcinoma progression via the PI3K/AKT/Twist signaling pathway. Oncol Lett 2018; 16:5299-5308. [PMID: 30250599 PMCID: PMC6144913 DOI: 10.3892/ol.2018.9281] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 11/16/2017] [Indexed: 12/12/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) serves critical roles in the migration, invasion and metastasis of human cancer cells. This process is initiated by regulation of E-cadherin expression by the major inducers of EMT. Previous studies reported that osteopontin (OPN) is essential for hepatocellular carcinoma (HCC) metastasis as it facilitates the EMT in HCC. However, the role and clinical significance of OPN as an EMT regulator in HCC remains unknown. The present study revealed that OPN regulated the expression of Twist by activating RAC serine/threonine-protein kinase (Akt), a critical EMT regulator. Interfering with the phosphoinositide 3-kinase (PI3K)/Akt pathway may suppress the expression of Twist enhanced by OPN. Increased Twist levels in HCC were associated with poor survival and tumor recurrence in patients with HCC following surgery. A significant association was observed between OPN expression and Twist levels in HCC, and a combination of these two parameters was revealed to be a more powerful predictor of poor patient prognosis. The findings of the present study indicate that Twist serves an notable role in OPN-mediated metastasis of HCC through activation of the PI3K/Akt pathway. Twist may be a potential therapeutic target for the prevention of HCC metastasis in patients exhibiting high OPN expression.
Collapse
Affiliation(s)
- Xinxin Yu
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Yan Zheng
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
- Cancer Metastasis Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xuchao Zhu
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Xiaomei Gao
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Chaoqun Wang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
- Cancer Metastasis Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yuanyuan Sheng
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
- Cancer Metastasis Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Wei Cheng
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Lunxiu Qin
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
- Cancer Metastasis Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Ning Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Huliang Jia
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
- Cancer Metastasis Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Qiongzhu Dong
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
- Cancer Metastasis Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
20
|
Wang X, Yao Y, Zhu X. The influence of aberrant expression of GLI1/p-S6K on colorectal cancer. Biochem Biophys Res Commun 2018; 503:3198-3204. [PMID: 30143258 DOI: 10.1016/j.bbrc.2018.08.124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/18/2018] [Indexed: 12/14/2022]
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. Recent studies have reported that PI3K/AKT/mTOR pathway regulated the GLI1 expression level via SMO-independent pathway in a variety of tumor types. We detected the expression level of GLI1/p-S6K in CRC tissues. We found the expression of GLI1/p-S6K was apparently close with lymph node metastasis and TNM stage and patients with positive GLI1/p-S6K expression had shorter survival time and patients with both GLI1 and p-S6K positive expression had an even worse overall survival than those with single positive expression. Moreover, GLI1 and p-S6K expression was considered to be independent prognostic factors in CRC patient and the positive co-expression of GLI1/p-S6K had greater influence than single expression positive on the prognosis of postoperative patients with tumor size≥5 cm, well differentiation, positive lymph node metastasis, venous invasion, neural invasion and TNM III-IV. Meanwhile, the GLI1/p-S6K expression had impact on more clinicopathologic features in colon-side carcinoma than in rectum-side carcinoma and the mTOR/S6K/GLI1 axis played an important role in CRC especially in advanced stage. Hence, further studies are underway to explore the molecular mechanism between GLI1 and p-S6K in CRC, and in addition, it offers novel facilities for molecular targeting therapy for CRC.
Collapse
Affiliation(s)
- Xuchao Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
21
|
Grindel BJ, Martinez JR, Tellman TV, Harrington DA, Zafar H, Nakhleh L, Chung LW, Farach-Carson MC. Matrilysin/MMP-7 Cleavage of Perlecan/HSPG2 Complexed with Semaphorin 3A Supports FAK-Mediated Stromal Invasion by Prostate Cancer Cells. Sci Rep 2018; 8:7262. [PMID: 29740048 PMCID: PMC5940808 DOI: 10.1038/s41598-018-25435-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Interrupting the interplay between cancer cells and extracellular matrix (ECM) is a strategy to halt tumor progression and stromal invasion. Perlecan/heparan sulfate proteoglycan 2 (HSPG2) is an extracellular proteoglycan that orchestrates tumor angiogenesis, proliferation, differentiation and invasion. Metastatic prostate cancer (PCa) cells degrade perlecan-rich tissue borders to reach bone, including the basement membrane, vasculature, reactive stromal matrix and bone marrow. Domain IV-3, perlecan’s last 7 immunoglobulin repeats, mimics native proteoglycan by promoting tumoroid formation. This is reversed by matrilysin/matrix metalloproteinase-7 (MMP-7) cleavage to favor cell dispersion and tumoroid dyscohesion. Both perlecan and Domain IV-3 induced a strong focal adhesion kinase (FAK) dephosphorylation/deactivation. MMP-7 cleavage of perlecan reversed this, with FAK in dispersed tumoroids becoming phosphorylated/activated with metastatic phenotype. We demonstrated Domain IV-3 interacts with the axon guidance protein semaphorin 3A (Sema3A) on PCa cells to deactivate pro-metastatic FAK. Sema3A antibody mimicked the Domain IV-3 clustering activity. Direct binding experiments showed Domain IV-3 binds Sema3A. Knockdown of Sema3A prevented Domain IV-3-induced tumoroid formation and Sema3A was sensitive to MMP-7 proteolysis. The perlecan-Sema3A complex abrogates FAK activity and stabilizes PCa cell interactions. MMP-7 expressing cells destroy the complex to initiate metastasis, destroy perlecan-rich borders, and favor invasion and progression to lethal bone disease.
Collapse
Affiliation(s)
- Brian J Grindel
- Department of BioSciences, Rice University, Houston, TX, 77005, USA.,Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA.,Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jerahme R Martinez
- Department of BioSciences, Rice University, Houston, TX, 77005, USA.,Department of Mechanical Engineering, University of Delaware, Newark, DE, 19706, USA
| | - Tristen V Tellman
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA
| | - Daniel A Harrington
- Department of BioSciences, Rice University, Houston, TX, 77005, USA.,Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA
| | - Hamim Zafar
- Department of Computer Science, Rice University, Houston, TX, 77005, USA
| | - Luay Nakhleh
- Department of Computer Science, Rice University, Houston, TX, 77005, USA
| | - Leland W Chung
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Mary C Farach-Carson
- Department of BioSciences, Rice University, Houston, TX, 77005, USA. .,Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA.
| |
Collapse
|
22
|
Abstract
Previous studies have found that G-protein-coupled receptor 116 (GPR116) is a regulator of breast cancer metastasis. However, the role of GPR116 in colorectal carcinoma (CRC) carcinogenesis and progression is unknown. In this study, We found GPR116 expression was significantly up-regulated in CRC specimens compared with corresponding non-cancerous tissues. Increased GPR116 expression in CRC was correlated with histological differentiation and distant metastasis. In addition, high expression of GPR116 was significantly associated with poor overall survival of CRC patients, which was also confirmed by GSE14333, GSE17536 and GSE33113 datasets from the Gene Expression Omnibus (GEO). Furthermore, we demonstrated that the ability of proliferation and invasion of CRC cell lines HCT116 and LOVO was markedly reduced after transfected with siRNA-GPR116. Meanwhile, GPR116 may drive EMT in CRC cells through AKT/EKR signaling pathway, resulting in metastasis. Thus, GPR116 may be a novel reliable prognostic indicator and a risk factor in CRC progression.
Collapse
|
23
|
Yu T, Jia W, An Q, Cao X, Xiao G. Bioinformatic Analysis of GLI1 and Related Signaling Pathways in Chemosensitivity of Gastric Cancer. Med Sci Monit 2018; 24:1847-1855. [PMID: 29596399 PMCID: PMC5890825 DOI: 10.12659/msm.906176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background This study assessed the prognostic value of GLI1 in gastric cancer and analyzed the possible GLI1-related signaling network in chemosensitivity. Material/Methods Bioinformatic data mining was performed by using data in the TCGA-Stomach Cancer (TCGA-STAD) and the Kaplan-Meier plotter. GLI1 co-expressed genes in TCGA-STAD were subjected to KEGG pathway analysis. The genes enriched in the KEGG pathways were further subjected to Protein-Protein Interaction (PPI) analysis. Results In TCGA-STAD, high GLI1 gene/exon expression was associated with significantly worse survival (p=0.016 and 0.0023 respectively). In the Kaplan-Meier plotter, high GLI1 expression was associated with unfavorable overall survival (OS) (HR: 1.68, 95%CI: 1.42–2, p<0.0001) and first progression-free survival (FPS) (HR: 1.72, 95%CI: 1.4–2.11, p<0.0001). In TCGA-STAD, 600 GLI1 co-expressed genes were identified (absolute Pearson’s r ≥0.5). The most significant pathways were pathways in cancer (p=230.0E-12) and the Hedgehog signaling pathway (p=6.9E-9). PI3K-AKT pathway (p=17.0E-9) has the largest proportion of gene enrichment. Some GLI1 co-expressed genes in the PI3K-AKT pathway are central nodes in the PPI network and also play important roles in chemosensitivity of gastric cancer. Nevertheless, the mechanisms underlying their co-expression are still largely unexplored. Conclusions High GLI1 expression is associated with unfavorable OS and FPS in patients with gastric cancer. As a member of the Hedgehog signaling pathway, GLI1 co-expressed genes are also largely enriched in PI3K/AKT pathway in gastric cancer, which is closely related to chemoresistance. The underlying mechanisms are still largely unexplored and need further study.
Collapse
Affiliation(s)
- Tao Yu
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Beijing, China (mainland)
| | - Wenzhuo Jia
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Beijing, China (mainland)
| | - Qi An
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Beijing, China (mainland)
| | - Xianglong Cao
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Beijing, China (mainland)
| | - Gang Xiao
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Beijing, China (mainland)
| |
Collapse
|
24
|
Hou Y, Hu Q, Huang J, Xiong H. Omeprazole Inhibits Cell Proliferation and Induces G0/G1 Cell Cycle Arrest through Up-regulating miR-203a-3p Expression in Barrett's Esophagus Cells. Front Pharmacol 2018; 8:968. [PMID: 29375376 PMCID: PMC5767174 DOI: 10.3389/fphar.2017.00968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022] Open
Abstract
Existing data suggest that proton pump inhibitors (PPIs), particularly omeprazole, have significant anti-tumor action in monotherapy and or combination chemotherapy. Hedgehog (Hh) signaling pathway represents a leading candidate as a molecular mediator of Barrett's esophagus (BE). Studies have indicated reduced miRNAs in BE progression, however, little is known about the latent anti-neoplasm effects of miRNAs in BE cells. Here, we investigated whether omeprazole could inhibit BE progression by regulating Hh pathway and explored the promising Hh-targeted miRNAs in BE cells. We conducted qRT-PCR and immunoblotting assay to evaluate the effects of omeprazole on the expression of Hh signaling components and miR-203a-3p in CP-A and CP-B cells. The promising target genes of miR-203a-3p were predicted by bioinformatics methods, and verified by luciferase assays and qRT-PCR. The effects of omeprazole on BE cell proliferation and cell cycle distribution were determined. The overexpression or silencing of miR-203a-3p was performed to test its anti-proliferative effects. Finally, rescue experiments that miR-203a-3p inhibitor alleviated the effects of omeprazole on decreasing the levels of Gli1 mRNA, protein and luciferase were performed. Mechanistic studies showed that omeprazole could inhibit the expression of Gli1 and the nuclear localization of Gli1. Moreover, we determined that omeprazole could selectively up-regulated the expression of miR-203a-3p, and Gli1 was a bona fide target of miR-203a-3p. miR-203a-3p inhibitor alleviated the suppressing effects of omeprazole on Gli1 luciferase activity, mRNA and protein level. The functional assay suggested that omeprazole could dose-dependently inhibit BE cell growth and induce cell cycle arrest in G0/G1 phase. Additionally, overexpression and silencing of miR-203a-3p in BE cells disrupted cell cycle progress, resulting in suppressing and accelerating cell proliferation, respectively. Taken together, these data provide a novel mechanism of potentially anti-neoplastic effects for omeprazole through modulation of miR-203a-3p expression and thus suppressing Hh/Gli1 signaling in BE cells.
Collapse
Affiliation(s)
- Yichao Hou
- 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
| | - Qiang Hu
- 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
| | - Jiao Huang
- 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
| | - Hua Xiong
- 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
| |
Collapse
|
25
|
Yue M, Li S, Yan G, Li C, Kang Z. Paeoniflorin inhibits cell growth and induces cell cycle arrest through inhibition of FoxM1 in colorectal cancer cells. Cell Cycle 2018; 17:240-249. [PMID: 29301438 DOI: 10.1080/15384101.2017.1407892] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Paeoniflorin (PF) exhibits tumor suppressive functions in a variety of human cancers. However, the function of PF and molecular mechanism in colorectal cancer are elusive. In the present study, we investigated whether PF could exert its antiproliferative activity, anti-migration, and anti-invasive function in colorectal cancer cells. We found that PF inhibited cell growth and induced apoptosis and blocked cell cycle progression in the G0/G1 phase in colorectal cancer cells. Moreover, we found that PF suppressed cell migration and invasion in colorectal cancer cells. FoxM1 has been reported to play an important oncogenic role in human cancers. We also determine whether PF inhibited the expression of FoxM1, leading to its anti-cancer activity. We found that PF treatment in colorectal cancer cells resulted in down-regulation of FoxM1. The rescue experiments showed that overexpression of FoxM1 abrogated the tumor suppressive function induced by PF treatment. Notably, depletion of FoxM1 promoted the anti-tumor activity of PF in colorectal cancer cells. Therefore, inhibition of FoxM1 could participate in the anti-tumor activity of PF in colorectal cancer cells.
Collapse
Affiliation(s)
- Meng Yue
- a Department of Colorectal and Anal Surgery , The First Hospital of Jilin University , Changchun , Jilin , 130021 , China
| | - Shiquan Li
- a Department of Colorectal and Anal Surgery , The First Hospital of Jilin University , Changchun , Jilin , 130021 , China
| | - Guoqiang Yan
- a Department of Colorectal and Anal Surgery , The First Hospital of Jilin University , Changchun , Jilin , 130021 , China
| | - Chenyao Li
- a Department of Colorectal and Anal Surgery , The First Hospital of Jilin University , Changchun , Jilin , 130021 , China
| | - Zhenhua Kang
- a Department of Colorectal and Anal Surgery , The First Hospital of Jilin University , Changchun , Jilin , 130021 , China
| |
Collapse
|
26
|
Lou W, Liu J, Gao Y, Zhong G, Chen D, Shen J, Bao C, Xu L, Pan J, Cheng J, Ding B, Fan W. MicroRNAs in cancer metastasis and angiogenesis. Oncotarget 2017; 8:115787-115802. [PMID: 29383201 PMCID: PMC5777813 DOI: 10.18632/oncotarget.23115] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/17/2017] [Indexed: 12/15/2022] Open
Abstract
Cancer metastasis is a malignant process by which tumor cells migrate from their primary site of origin to other organs. It is the main cause of poor prognosis in cancer patients. Angiogenesis is the process of generating new blood capillaries from pre-existing vasculature. It plays a vital role in primary tumor growth and distant metastasis. MicroRNAs are small non-coding RNAs involved in regulating normal physiological processes as well as cancer pathogenesis. They suppress gene expression by specifically binding to the 3′-untranslated region (3′-UTR) of their target genes. They can thus act as oncogenes or tumor suppressors depending on the function of their target genes. MicroRNAs have shown great promise for use in anti-metastatic cancer therapy. In this article, we review the roles of various miRNAs in cancer angiogenesis and metastasis and highlight their potential for use in future therapies against metastatic cancer.
Collapse
Affiliation(s)
- Weiyang Lou
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Jingxing Liu
- Department of Intensive Care Unit, Changxing People's Hospital of Zhejiang, Zhejiang Province, Huzhou 313100, China
| | - Yanjia Gao
- Department of Anesthesiology, International Hospital of Zhejiang University, Shulan (Hangzhou) Hospital, Zhejiang Province, Hangzhou 310003, China
| | - Guansheng Zhong
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Danni Chen
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Jiaying Shen
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Chang Bao
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Liang Xu
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Jie Pan
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Junchi Cheng
- Department of Chemotherapy, Zhejiang Cancer Hospital, Zhejiang Province, Hangzhou 310003, China
| | - Bisha Ding
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
27
|
MicroRNA-761 promotes the sensitivity of colorectal cancer cells to 5-Fluorouracil through targeting FOXM1. Oncotarget 2017; 9:321-331. [PMID: 29416616 PMCID: PMC5787468 DOI: 10.18632/oncotarget.20109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/25/2017] [Indexed: 12/14/2022] Open
Abstract
Resistance to chemotherapy is a big challenge for treatment of patients with colorectal cancer; however; the mechanism underlying chemoresistance in colorectal cancer cell has not been elucidated. MicroRNAs (miRNAs) are new players in the development of drug chemoresistance. In our study, we indicated that overexpression of miR-761 promoted the sensitivity of colorectal cancer cells to 5-Fluorouracil (5-FU). miR-761 expression was downregulated in colorectal cancer cell lines and tissues. miR-761 expression was lower in patients with low grade than in patients with high grade. In additon, we showed that elevated expression of miR-761 suppressed colorectal cancer cell proliferation, cell cycle, colony formation and cell invasion. We identified that FOXM1 was a direct target gene of miR-761 in colorectal cancer cell. FOXM1 expression was upregulated in colorectal cancer tissues compare to the adjacent non-tumor tissues. MiR-761 expression was negatively associated with the expression of FOXM1 in colorectal cancer tissues. Elevated expression of FOXM1 suppressed the sensitivity of miR-761-overexpressing HT29 cells to 5-FU. We also indicated that FOXM1 overexpression promoted cell proliferation, cycle and invasion of miR-761-overexpressing HT29 cells. These data suggested that miR-761 played a tumor suppressor miRNA in colorectal cancer progression and reduced miR-761 expression might be a major mechanism for 5-FU resistance in colorectal cancer cell.
Collapse
|
28
|
Chen Y, Liu Y, Ni H, Ding C, Zhang X, Zhang Z. FoxM1 overexpression promotes cell proliferation and migration and inhibits apoptosis in hypopharyngeal squamous cell carcinoma resulting in poor clinical prognosis. Int J Oncol 2017; 51:1045-1054. [PMID: 28848994 PMCID: PMC5592873 DOI: 10.3892/ijo.2017.4094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/21/2017] [Indexed: 01/08/2023] Open
Abstract
Forkhead box M1 (FoxM1), a member of the Fox family of transcriptional factors, is involved in the development of various human malignancies. However, the expression level of FoxM1 and its functional role in hypopharyngeal squamous cell carcinoma (HSCC) remained unclear to date. The aim of the present study was to investigate the FoxM1 expression in 63 HSCC and 20 adjacent normal tissues, as well as to evaluate its association with the clinicopathological parameters and its diagnostic value in HSCC. To further explore the biological function of FoxM1 in vitro, siRNAs were used to knockdown the expression of FoxM1 in the HSCC cell line Fadu. The results revealed that FoxM1 protein was highly expressed in HSCC tissues and that its high expression was closely associated with HSCC tumor differentiation (P=0.004), tumor size (P=0.002), clinical stage (P=0.001), lymph node metastasis (P=0.002), treatment (P=0.045) and expression of the proliferation marker Ki-67 (P<0.001). Additionally, the elevated expression of FoxM1 in HSCC patients consistently predicted a poor survival time. Knockdown of FoxM1 expression blocked Fadu cell proliferation and promoted apoptosis, and also led to the down-regulation of cyclin A1 expression. Furthermore, decreased expression of FoxM1 markedly impeded cell migration and reversed the epithelial-mesenchymal transition phenotype, as indicated by decreased expression of vimentin and increased expression of E-cadherin in Fadu cells. These results indicate that FoxM1 may act as an oncogene and serve as a therapeutic target against malignant progression in HSCC.
Collapse
Affiliation(s)
- Yan Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Haosheng Ni
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chuanjin Ding
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaobo Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhenxin Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
29
|
Zhang X, Lv QL, Huang YT, Zhang LH, Zhou HH. Akt/FoxM1 signaling pathway-mediated upregulation of MYBL2 promotes progression of human glioma. J Exp Clin Cancer Res 2017; 36:105. [PMID: 28784180 PMCID: PMC5547476 DOI: 10.1186/s13046-017-0573-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/26/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND MYB-related protein B (B-MYB/MYBL2), a member of the myeloblastosis family of transcription factors, has been reported for its role in the genesis and progression of tumors. Forkhead box M1 (FoxM1), another transcriptional factor, is considered to be an independent predictor of poor survival in many solid cancers. The aim of the present study was to investigate the clinical significance of the correlation between MYBL2 and FoxM1 in glioma and the possible mechanism of FoxM1and MYBL2 expression. METHODS MYBL2 and FoxM1expression in cancerous tissues and cell lines were determined by reverse transcription-PCR (RT-PCR), Western blotting and immunostaining. The co-expression of MYBL2 and FoxM1 was analyzed in low-grade glioma (LGG) and glioblastoma (HGG) cohorts of TCGA using cBioportal and UCSC Xena. And, the role of MYBL2 and FoxM1 in glioma cell progression and the underlying mechanisms were studied by using small interfering RNA (si-RNA) and pcDNA3.1 + HAvectors. Furthermore, the effects of MYBL2 and FoxM1 in cell proliferation, cell cycle progression, apoptosis, migration, invasion, and adhesion were determined by cell proliferation assays, flow cytometry analysis, transwell migration and cell adhesion assay. RESULTS MYBL2 and FoxM1 expression are significantly associated with clinical stages and overall survival of glioma patients. In cohorts of TCGA, patients with high MYBL2 but without radio-chemotherapy had the highest hazard ratio (adjusted HR = 5.29, 95% CI = 1.475-18.969, P < 0.05). Meanwhile, MYBL2 closely related to the FoxM1 expression in 79 glioma tissues (r = 0.742, p < 0.05) and LGG (r = 0.83) and HGG (r = 0.74) cohorts of TCGA. Down regulation of FoxM1 and MYBL2 by siRNAs induced the cell cycle arrest, apoptosis and EMT of glioma cells. Furthermore, inactivations of Akt/FoxM1 signaling by Akt inhibitor and siRNA-FoxM1 reduce the expression of MYBL2 in glioma cells. CONCLUSIONS MYBL2 is a key downstream factor of Akt/FoxM1 signaling to promote progression of human glioma, and could be a new candidate gene for molecular targeting therapy and biomarker for radiotherapy of glioma. TRIAL REGISTRATION CTXY-1300041-3-2. ChiCTR-COC-15006186 . Registered date: 13 September 2013.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, Hunan, People's Republic of China
| | - Qiao-Li Lv
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, Hunan, People's Republic of China
| | - Yuan-Tao Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, Hunan, People's Republic of China
- Department of Neurology, The Brain Hospital of Hunan Province, Changsha, 410007, Hunan, People's Republic of China
| | - Li-Hua Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, Hunan, People's Republic of China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, Hunan, People's Republic of China.
| |
Collapse
|
30
|
Wang D, Hu G, Du Y, Zhang C, Lu Q, Lv N, Luo S. Aberrant activation of hedgehog signaling promotes cell proliferation via the transcriptional activation of forkhead Box M1 in colorectal cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:23. [PMID: 28148279 PMCID: PMC5288899 DOI: 10.1186/s13046-017-0491-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 01/10/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Recent evidence suggests that the aberrant activation of Hedgehog (Hh) signaling by Gli transcription factors is characteristic of a variety of aggressive human carcinomas, including colorectal cancer (CRC). Forkhead box M1 (FoxM1) controls the expression of a number of cell cycle regulatory proteins, and FoxM1 expression is elevated in a broad range of human malignancies, which suggests that it plays a crucial role in tumorigenesis. However, the mechanisms underlying FoxM1 expression are not fully understood. Here, we aim to further investigate the molecular mechanism by which Gli1 regulates FoxM1 in CRC. METHODS Western blotting and immunohistochemistry (IHC) were used to evaluate FoxM1 and Gli1 protein expression, respectively, in CRC tissues and matched adjacent normal mucosa. BrdU (5-bromo-2'-deoxyuridine) and clone formation assays were used to clarify the influence of FoxM1 on CRC cell growth and proliferation. Chromatin immunoprecipitation (ChIP) and luciferase experiments were performed to explore the potential mechanisms by which Gli1 regulates FoxM1. Additionally, the protein and mRNA expression levels of Gli1 and FoxM1 in six CRC cell lines were measured using Western blotting and real-time PCR. Finally, the effect of Hh signaling on the expression of FoxM1 was studied in cell biology experiments, and the effects of Hh signaling activation and FoxM1 inhibition on the distribution of CRC cells among cell cycle phases was assessed by flow cytometry. RESULTS Gli1 and FoxM1 were abnormally elevated in human CRC tissues compared with matched adjacent normal mucosa samples, and FoxM1 is a downstream target gene of the transcription factor Gli1 in CRC and promoted CRC cell growth and proliferation. Moreover, the aberrant activation of Hh signaling promoted CRC cell proliferation by directly binding to the promoter of FoxM1 and transactivating the activity of FoxM1 in CRC cells. CONCLUSION The dysregulation of the Hh-Gli1-FoxM1 axis is essential for the proliferation and growth of human CRC cells and offers a potent target for therapeutic intervention in CRC.
Collapse
Affiliation(s)
- DeJie Wang
- Institute of Digestive Disease, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Guohui Hu
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.,Jiangxi Key Laboratory of Molecular Diagnosis and Precision Medicine, Nancahng, Jiangxi, 330006, China
| | - Ying Du
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.,Jiangxi Key Laboratory of Molecular Diagnosis and Precision Medicine, Nancahng, Jiangxi, 330006, China
| | - Cheng Zhang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.,Jiangxi Key Laboratory of Molecular Diagnosis and Precision Medicine, Nancahng, Jiangxi, 330006, China
| | - Quqin Lu
- Department of Epidemiology & Biostatistics, School of Public Health, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Nonghua Lv
- Institute of Digestive Disease, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Donghu District, Nanchang, Jiangxi, 330006, China.
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China. .,Jiangxi Key Laboratory of Molecular Diagnosis and Precision Medicine, Nancahng, Jiangxi, 330006, China.
| |
Collapse
|