1
|
Li M, Wang S, Huang H, Li L. Reliable estrogen-related prognostic signature for uterine corpus endometrial carcinoma. Comput Biol Chem 2024; 113:108216. [PMID: 39326337 DOI: 10.1016/j.compbiolchem.2024.108216] [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: 05/24/2024] [Revised: 09/04/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Uterine corpus endometrial carcinoma (UCEC) is a predominant gynecological malignancy worldwide. Overdosed estrogen exposure has been widely known as a crucial risk factor for UCEC patients. The purpose of this work is to explore crucial estrogen-related genes (ERGs) in UCEC. METHODS UCEC scRNA-seq data, bulk RNA data, and ERGs were obtained from GEO, TCGA, and Molecular Signature Database, respectively. Differential expression analysis and cross analysis determined the candidate genes, and optimal genes in risk score were obtained after univariate Cox regression analysis, LASSO Cox regression analysis, and multivariate Cox regression analysis. The functional information was revealed by GO, KEGG, and GSVA enrichment analyses. CCK8 assay was used to detect the drug sensitivity. RESULTS After cross analysis of the differentially expressed genes and the 8734 ERGs, 86 differentially expressed ERGs were identified in UCEC, which were significantly enriched in some immune related pathways and microbiota related pathways. Of them, the most optimal 8 ERGs were obtained to build prognostic risk score, including GAL, PHGDH, SLC7A2, HNMT, CLU, AREG, MACC1, and HMGA1. The risk score could reliably predict patient prognosis, and high-risk patients had worse prognosis. Higher HMGA1 gene expression exhibited higher sensitivity to Osimertinib. CONCLUSIONS Predictive risk score based on 8 ERGs exhibited excellent prognostic value in UCEC patients, and high-risk patients had inferior survival. UCEC patients with distinct prognoses showed different tumor immune microenvironment.
Collapse
Affiliation(s)
- Mojuan Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 511500, China; Department of Obstetrics and Gynecology, the Sixth Affiliated Hospital, South China University of Technology, Foshan, Guangdong 528000, China
| | - Shuai Wang
- Department of Obstetrics and Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong 511500, China
| | - Hao Huang
- Department of Obstetrics and Gynecology, the Sixth Affiliated Hospital, South China University of Technology, Foshan, Guangdong 528000, China
| | - Li Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 511500, China; Department of Obstetrics and Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong 511500, China.
| |
Collapse
|
2
|
Sun Y, Guo G, Zhang Y, Chen X, Lu Y, Hong R, Xiong J, Li J, Hu X, Wang S, Liu Y, Zhang Z, Yang X, Nan Y, Huang Q. IKBKE promotes the ZEB2-mediated EMT process by phosphorylating HMGA1a in glioblastoma. Cell Signal 2024; 116:111062. [PMID: 38242271 DOI: 10.1016/j.cellsig.2024.111062] [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: 08/08/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
IKBKE (Inhibitor of Nuclear Factor Kappa-B Kinase Subunit Epsilon) is an important oncogenic protein in a variety of tumors, which can promote tumor growth, proliferation, invasion and drug resistance, and plays a critical regulatory role in the occurrence and progression of malignant tumors. HMGA1a (High Mobility Group AT-hook 1a) functions as a cofactor for proper transcriptional regulation and is highly expressed in multiple types of tumors. ZEB2 (Zinc finger E-box Binding homeobox 2) exerts active functions in epithelial mesenchymal transformation (EMT). In our current study, we confirmed that IKBKE can increase the proliferation, invasion and migration of glioblastoma cells. We then found that IKBKE can phosphorylate HMGA1a at Ser 36 and/or Ser 44 sites and inhibit the degradation process of HMGA1a, and regulate the nuclear translocation of HMGA1a. Crucially, we observed that HMGA1a can regulate ZEB2 gene expression by interacting with ZEB2 promoter region. Hence, HMGA1a was found to promote the ZEB2-related metastasis. Consequently, we demonstrated that IKBKE can exert its oncogenic functions via the IKBKE/HMGA1a/ZEB2 signalling axis, and IKBKE may be a prominent biomarker for the treatment of glioblastoma in the future.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Gaochao Guo
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Yu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Xingjie Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Yalin Lu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Rujun Hong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Jinbiao Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Jiabo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Xue Hu
- Department of Clinical Nutrition, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, China
| | - Shuaishuai Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Yang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Zhimeng Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Neurosurgery, Ningbo Hospital of Zhejiang University, Ningbo, Zhejiang 315000, China
| | - Xuejun Yang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yang Nan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300052, China.
| |
Collapse
|
3
|
Li H, Cai L, Pan Q, Jiang X, Zhao J, Xiang T, Tang Y, Wang Q, He J, Weng D, Zhang Y, Liu Z, Xia J. N 6-methyladenosine-modified VGLL1 promotes ovarian cancer metastasis through high-mobility group AT-hook 1/Wnt/β-catenin signaling. iScience 2024; 27:109245. [PMID: 38439973 PMCID: PMC10910247 DOI: 10.1016/j.isci.2024.109245] [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: 07/17/2023] [Revised: 11/30/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
The main causes of death in patients with ovarian cancer (OC) are invasive lesions and the spread of metastasis. The present study aimed to explore the mechanisms that might promote OC metastasis. Here, we identified that VGLL1 expression was remarkably increased in metastatic OC samples. The role of VGLL1 in OC metastasis and tumor growth was examined by cell function assays and mouse models. Mechanistically level, METTL3-mediated N6-methyladenosine (m6A) modification contributed to VGLL1 upregulation in an IGF2BP2 recognition-dependent manner. Furthermore, VGLL1 directly interacts with TEAD4 and co-transcriptionally activates HMGA1. HMGA1 further activates Wnt/β-catenin signaling to enhance OC metastasis by promoting the epithelial-mesenchyme transition traits. Rescue assays indicated that the upregulation of HMGA1 was essential for VGLL1-induced metastasis. Collectively, these findings showed that the m6A-induced VGLL1/HMGA1/β-catenin axis might play a vital role in OC metastasis and tumor growth. VGLL1 might serve as a prognostic marker and therapeutic target against the metastasis of OC.
Collapse
Affiliation(s)
- Han Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Department of Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Liming Cai
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Qiuzhong Pan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Xingyu Jiang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jingjing Zhao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Tong Xiang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yan Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Qijing Wang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jia He
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Desheng Weng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yanna Zhang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Jianchuan Xia
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
4
|
Wu Z, Zhu J, Wen Y, Lei P, Xie J, Shi H, Wu R, Lou X, Hu Y. Hmga1-overexpressing lentivirus protects against osteoporosis by activating the Wnt/β-catenin pathway in the osteogenic differentiation of BMSCs. FASEB J 2023; 37:e22987. [PMID: 37555233 DOI: 10.1096/fj.202300488r] [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: 03/14/2023] [Revised: 04/26/2023] [Accepted: 05/09/2023] [Indexed: 08/10/2023]
Abstract
Postmenopausal osteoporosis is associated with bone formation inhibition mediated by the impaired osteogenic differentiation potential of bone marrow mesenchymal stem cells (BMSCs). However, identifying and confirming the essential genes in the osteogenic differentiation of BMSCs and osteoporosis remain challenging. The study aimed at revealing the key gene that regulated osteogenic differentiation of BMSCs and led to osteoporosis, thus exploring its therapeutic effect in osteoporosis. In the present study, six essential genes related to the osteogenic differentiation of BMSCs and osteoporosis were identified, namely, fibrillin 2 (Fbn2), leucine-rich repeat-containing 17 (Lrrc17), heat shock protein b7 (Hspb7), high mobility group AT-hook 1 (Hmga1), nexilin F-actin-binding protein (Nexn), and endothelial cell-specific molecule 1 (Esm1). Furthermore, the in vivo and in vitro experiments showed that Hmga1 expression was increased during the osteogenic differentiation of rat BMSCs, while Hmga1 expression was decreased in the bone tissue of ovariectomized (OVX) rats. Moreover, the expression of osteogenic differentiation-related genes, the activity of alkaline phosphatase (ALP), and the number of mineralized nodules were increased after Hmga1 overexpression, which was partially reversed by a Wnt signaling inhibitor (DKK1). In addition, after injecting Hmga1-overexpressing lentivirus into the bone marrow cavity of OVX rats, the bone loss, and osteogenic differentiation inhibition of BMSCs in OVX rats were partially reversed, while osteoclast differentiation promotion of BMSCs in OVX rats was unaffected. Taken together, the present study confirms that Hmga1 prevents OVX-induced bone loss by the Wnt signaling pathway and reveals that Hmga1 is a potential gene therapeutic target for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Zhixin Wu
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiayong Zhu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yinxian Wen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Pengfei Lei
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Xie
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Haifei Shi
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ronghuan Wu
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xianfeng Lou
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yihe Hu
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Yan B, Liu C, Li H, Wen N, Jiao W, Wang S, Zhang Y, Zhang T, Zhang H, Lv Y, Fan H, Liu X. Reversal of HMGA1-Mediated Immunosuppression Synergizes with Immunogenic Magnetothermodynamic for Improved Hepatocellular Carcinoma Therapy. ACS NANO 2023; 17:9209-9223. [PMID: 37162457 DOI: 10.1021/acsnano.3c00004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Magnetothermodynamic (MTD) therapy can activate antitumor immune responses by inducing potent immunogenic tumor cell death. However, tumor development is often accompanied by multifarious immunosuppressive mechanisms that can counter the efficacy of immunogenic MTD therapy. High-mobility group protein A1 (HMGA1) is overexpressed within hepatocellular carcinoma tissues and plays a crucial function in the generation of immunosuppressive effects. The reversal of HMGA1-mediated immunosuppression could enhance immunogenic tumor cell death-induced immune responses. A ferrimagnetic vortex-domain iron oxide (FVIO) nanoring-based nanovehicle was developed, which is capable of efficiently mediating an alternating magnetic field for immunogenic tumor cell death induction, while concurrently delivering HMGA1 small interfering (si)RNA (siHMGA1) to the cytoplasm of hepatocellular carcinoma Hepa 1-6 cells for HMGA1 pathway interference. Using siHMGA1-FVIO-mediated MTD therapy, the proliferation of hepatocellular carcinoma Hepa 1-6 tumors was inhibited, and the survival of a mouse model was improved. We also demonstrated that siHMGA1-FVIO-mediated MTD achieved synergistic antitumor effects in a subcutaneous hepatocellular carcinoma Hepa 1-6 and H22 tumor model by promoting dendritic cell maturation, enhancing antigen-presenting molecule expression (both major histocompatibility complexes I and II), improving tumor-infiltrating T lymphocyte numbers, and decreasing immunosuppressive myeloid-derived suppressor cells, interleukin-10, and transforming growth factor-β expression. The nanoparticle system outlined in this paper has the potential to target HMGA1 and, in combination with MTD-induced immunotherapy, is a promising approach for hepatocellular carcinoma treatment.
Collapse
Affiliation(s)
- Bin Yan
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Chen Liu
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Hugang Li
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Nana Wen
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Wangbo Jiao
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Siyao Wang
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yihan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Tingbin Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Huan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
- Department of Radiology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong 519000, China
| | - Yi Lv
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Haiming Fan
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Xiaoli Liu
- Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, Shaanxi 710069, China
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
6
|
Yu BY, Shi LG, Jiang C, Wang GK, Liu J, Wu TY. Kinesin family member C 1 overexpression exerts tumor-promoting properties in head and neck squamous cell carcinoma via the Rac1/Wnt/β-catenin pathway. J Transl Med 2023; 103:100134. [PMID: 36990154 DOI: 10.1016/j.labinv.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Kinesin family member C 1 (KIFC1) is a kinesin-14 motor protein, and its abnormal upregulation promotes the malignant behavior of cancer cells. N6-methyladenosine (m6A) RNA methylation is a common modification of eukaryotic mRNA and affects RNA expression. Herein, we explored how KIFC1 regulated head and neck squamous cell carcinoma (HNSCC) tumorigenesis and how m6A modification affected KIFC1 expression. Bioinformatics analysis was performed to screen for genes of interest, and in vitro and in vivo studies were carried out to investigate the function and mechanism of KIFC1 in HNSCC. We observed that the expression of KIFC1 in HNSCC tissues was significantly higher than in normal or adjacent normal tissues. Cancer patients with higher KIFC1 expression have lower tumor differentiation status. Demethylase alkB homolog 5 (ALKBH5), a cancer-promoting factor in HNSCC, could interact with KIFC1 mRNA and post-transcriptionally activated KIFC1 through m6A modification. KIFC1 downregulation suppressed HNSCC cell growth and metastasis in vivo and in vitro. However, overexpression of KIFC1 promoted these malignant behaviors. We demonstrated that KIFC1 overexpression activated the oncogenic Wnt/β-catenin pathway. KIFC1 interacted with the small GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1) at the protein level and increased activity. The Rho GTPase Rac1 was indicated to be an upstream activator of the Wnt/β-catenin signaling pathway, and its Rac1 inhibitor, NSC-23766, treatment reversed the effects caused by KIFC1 overexpression. Those observations demonstrate that abnormal expression of KIFC1 may be regulated by demethylase ALKBH5 in an m6A-dependent manner and promote HNSCC progression via the Rac1/Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Bo-Yu Yu
- Department of Otolaryngology, Head and Neck Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Ling-Gai Shi
- Department of Otolaryngology, Head and Neck Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Chang Jiang
- Department of Otolaryngology, Head and Neck Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Guang-Ke Wang
- Department of Otolaryngology, Head and Neck Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, China.
| | - Jun Liu
- Department of Otolaryngology, Head and Neck Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, China.
| | - Tian-Yi Wu
- Department of Otolaryngology, Head and Neck Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, China.
| |
Collapse
|
7
|
Wu Z, Huang Y, Yuan W, Wu X, Shi H, Lu M, Xu A. Expression, tumor immune infiltration, and prognostic impact of HMGs in gastric cancer. Front Oncol 2022; 12:1056917. [PMID: 36568211 PMCID: PMC9780705 DOI: 10.3389/fonc.2022.1056917] [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: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022] Open
Abstract
Background In the past decade, considerable research efforts on gastric cancer (GC) have been expended, however, little advancement has been made owing to the lack of effective biomarkers and treatment options. Herein, we aimed to examine the levels of expression, mutations, and clinical relevance of HMGs in GC to provide sufficient scientific evidence for clinical decision-making and risk management. Methods GC samples were obtained from The Cancer Genome Atlas (TCGA). University of California Santa Cruz (UCSC) XENA, Human Protein Atlas (HPA), Gene Expression Profiling Interactive Analysis (GEPIA), Kaplan-Meier Plotter, cBioPortal, GeneMANIA, STRING, LinkedOmics, and DAVID databases were employed. The "ggplot2" package in the R software (×64 3.6.3) was used to thoroughly analyze the effects of HMGs. qRT-PCR was performed to assess HMG levels in GC cell lines. Results A total of 375 GC tissues and 32 paraneoplastic tissues were analyzed. The levels of HMGA1, HMGA2, HMGB1, HMGB2, HMGB3, HMGN1, HMGN2, and HMGN4 expression were increased in GC tissues relative to normal gastric tissues. HMGA1, HMGA2, HMGB1, HMGB2, and HMGB3 were highly expressed in GC cell lines. The OS was significantly different in the group showing low expressions of HMGA1, HMGA2, HMGB1, HMGB2, HMGB3, HMGN2, HMGN3, and HMGN5. There was a significant difference in RFS between the groups with low HMGA2, HMGB3, and high HMGN2 expression. The levels of HMGA2, HMGB3, and HMGN1 had a higher accuracy for prediction to distinguish GC from normal tissues (AUC value > 0.9). HMGs were tightly associated with immune infiltration and tumor immune escape and antitumor immunity most likely participates in HMG-mediated oncogenesis in GC. GO and KEGG enrichment analyses showed that HMGs played a vital role in the cell cycle pathway. Conclusions Our results strongly suggest a vital role of HMGs in GC. HMGA2 and HMGB3 could be potential markers for prognostic prediction and treatment targets for GC by interrupting the cell cycle pathway. Our findings might provide renewed perspectives for the selection of prognostic biomarkers among HMGs in GC and may contribute to the determination of the optimal strategy for the treatment of these patients.
Collapse
Affiliation(s)
- Zhiheng Wu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Yang Huang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Weiwei Yuan
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Xiong Wu
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, PR China, State Key Laboratory of Optometry, Ophthalmology, and Visual Science, Wenzhou, Zhejiang, China
| | - Hui Shi
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ming Lu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Aman Xu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| |
Collapse
|
8
|
Therachiyil L, Hussein OJ, Uddin S, Korashy HM. Regulation of the aryl hydrocarbon receptor in cancer and cancer stem cells of gynecological malignancies: An update on signaling pathways. Semin Cancer Biol 2022; 86:1186-1202. [PMID: 36252938 DOI: 10.1016/j.semcancer.2022.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 01/27/2023]
Abstract
Gynecological malignancies are a female type of cancers that affects the reproductive system. Cancer metastasis or recurrence mediated by cellular invasiveness occurs at advanced stages of cancer progression. Cancer Stem Cells (CSCs) enrichment in tumors leads to chemoresistance, which results in cancer mortality. Exposure to environmental pollutants such as polycyclic aromatic hydrocarbons is associated with an increased the risk of CSC enrichment in gynecological cancers. One of the important pathways that mediates the metabolism and bioactivation of these environmental chemicals is the transcription factor, aryl hydrocarbon receptor (AhR). The present review explores the molecular mechanisms regulating the crosstalk and interaction of the AhR with cancer-related signaling pathways, such as apoptosis, epithelial-mesenchymal transition, immune checkpoints, and G-protein-coupled receptors in several gynecological malignancies such as ovarian, uterine, endometrial, and cervical cancers. The review also discusses the potential of targeting the AhR pathway as a novel chemotherapy for gynecological cancers.
Collapse
Affiliation(s)
- Lubna Therachiyil
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Ola J Hussein
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
9
|
Li Y, Li F, Wang Y, Song F, Qi L, Hu Q. circ-LIMK1 regulates cisplatin resistance in lung adenocarcinoma by targeting miR-512-5p/HMGA1 axis. Open Med (Wars) 2022; 17:1568-1583. [PMID: 36304135 PMCID: PMC9547352 DOI: 10.1515/med-2022-0542] [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: 01/08/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/12/2022] Open
Abstract
This study aimed to unveil the detailed role and new mechanism of circ-LIMK1 in lung adenocarcinoma. Real-time quantitative polymerase chain reaction was performed to analyze the expression of circ-LIMK1, miR-512-5p, and HMGA1. 3-(4,5)-Dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide assay was employed to test the half maximal inhibitory concentration of cisplatin (DDP). Western blot was used to measure the expression of HMGA1, multidrug resistance protein 1, mitochondrial 37S ribosomal protein, and vascular endothelial growth factor A. Colony formation assay, flow cytometry, transwell assay, and tube formation assay were performed to analyze cell functions. Animal models were established to assay the role of circ-LIMK1 in vivo. The expression of circ-LIMK1 was up-regulated in DDP-resistant tumor tissues and cells. Knockdown of circ-LIMK1 reduced DDP resistance, impaired cancer cell growth, migration, invasion, and angiogenesis. circ-LIMK1 targeted miR-512-5p, and HMGA1 was targeted by miR-512-5p. MiR-512-5p absence could restore the repressive effects of circ-LIMK1 knockdown on lung adenocarcinoma cell phenotypes. Overexpression of HMGA1 could restore the inhibitory effects of miR-512-5p enrichment on lung adenocarcinoma cell malignant phenotypes. Knockdown of circ-LIMK1 could reduce growth of DDP-resistant tumors in vivo. Collectively, circ-LIMK1 regulated DDP resistance in lung adenocarcinoma by targeting miR-512-5p/HMGA1 axis.
Collapse
Affiliation(s)
- Ya Li
- Department of Respiratory and Critical Care Medicine, Xi’an Gaoling District Hospital, Xi’an, China
| | - Fangfang Li
- Department of Respiratory and Critical Care Medicine, Xi’an Gaoling District Hospital, Xi’an, China
| | - Yaya Wang
- Department of Respiratory and Critical Care Medicine, Xi’an Gaoling District Hospital, Xi’an, China
| | - Fangyu Song
- Department of Respiratory and Critical Care Medicine, Xi’an Gaoling District Hospital, Xi’an, China
| | - Lin Qi
- Department of Respiratory and Critical Care Medicine, Xi’an Gaoling District Hospital, Xi’an, China
| | - Qiang Hu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Xi’an Medical College, No. 277 Youyi West Road, Beilin District, Xi’an 710068, China
| |
Collapse
|
10
|
Wang L, Zhang J, Xia M, Liu C, Zu X, Zhong J. High Mobility Group A1 (HMGA1): Structure, Biological Function, and Therapeutic Potential. Int J Biol Sci 2022; 18:4414-4431. [PMID: 35864955 PMCID: PMC9295051 DOI: 10.7150/ijbs.72952] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022] Open
Abstract
High mobility group A1 (HMGA1) is a nonhistone chromatin structural protein characterized by no transcriptional activity. It mainly plays a regulatory role by modifying the structure of DNA. A large number of studies have confirmed that HMGA1 regulates genes related to tumours in the reproductive system, digestive system, urinary system and haematopoietic system. HMGA1 is rare in adult cells and increases in highly proliferative cells such as embryos. After being stimulated by external factors, it will produce effects through the Wnt/β-catenin, PI3K/Akt, Hippo and MEK/ERK pathways. In addition, HMGA1 also affects the ageing, apoptosis, autophagy and chemotherapy resistance of cancer cells, which are linked to tumorigenesis. In this review, we summarize the mechanisms of HMGA1 in cancer progression and discuss the potential clinical application of targeted HMGA1 therapy, indicating that targeted HMGA1 is of great significance in the diagnosis and treatment of malignancy.
Collapse
Affiliation(s)
- Lu Wang
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Ji Zhang
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China
| | - Min Xia
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Chang Liu
- Department of Endocrinology and Metabolism, The First People's Hospital of Chenzhou, First School of Clinical Medicine, University of Southern Medical, Guangzhou 510515, Guangdong, China
| | - Xuyu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jing Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.,Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| |
Collapse
|
11
|
Banz-Jansen C, Helweg LP, Kaltschmidt B. Endometrial Cancer Stem Cells: Where Do We Stand and Where Should We Go? Int J Mol Sci 2022; 23:ijms23063412. [PMID: 35328833 PMCID: PMC8955970 DOI: 10.3390/ijms23063412] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/08/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
Endometrial cancer is one of the most common malignant diseases in women worldwide, with an incidence of 5.9%. Thus, it is the most frequent cancer of the female genital tract, with more than 34,000 women dying, in Europe and North America alone. Endometrial Cancer Stem Cells (CSC) might be drivers of carcinogenesis as well as metastatic and recurrent disease. Therefore, targeting CSCs is of high interest to improve prognosis of patients suffering of advanced or recurrent endometrial cancer. This review describes the current evidence of molecular mechanisms in endometrial CSCs with special emphasis on MYC and NF-κB signaling as well as mitochondrial metabolism. Furthermore, the current status of immunotherapy targeting PD-1 and PD-L1 in endometrial cancer cells and CSCs is elucidated. The outlined findings encourage novel therapies that target signaling pathways in endometrial CSCs as well as immunotherapy as a promising therapeutic approach in the treatment of endometrial cancer to impede cancer progression and prevent recurrence.
Collapse
Affiliation(s)
- Constanze Banz-Jansen
- Department of Gynecology and Obstetrics, and Perinatal Center, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33617 Bielefeld, Germany;
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany;
| | - Laureen P. Helweg
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany;
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Correspondence:
| | - Barbara Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany;
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
12
|
Zhou Y, Pan A, Zhang Y, Li X. Hsa_circ_0039569 facilitates the progression of endometrial carcinoma by targeting the miR-197/high mobility group protein A1 axis. Bioengineered 2022; 13:4212-4225. [PMID: 35130798 PMCID: PMC8973714 DOI: 10.1080/21655979.2022.2027060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Circular RNAs are novel regulators in endometrial carcinoma. Hsa_circ_0039569 was reportedly upregulated in endometrial carcinoma; however, the functional roles and mechanisms of hsa_circ_0039569 need further investigation. Therefore, we used quantitative real-time PCR (qRT–PCR) to determine the mRNA levels of hsa_circ_0039569, miR-197 and high mobility group protein A1 (HMGA1). The protein level of HMGA1 was determined by Western blot. Cell Counting Kit-8 and colony formation assays were used to assess cell proliferation. Cell migration was measured via wound healing and Transwell assays. Transwell assay was also performed to determine cell invasion ability. Direct binding of the indicated molecules were verified by RNA binding protein immunoprecipitation (RIP) assay and dual luciferase reporter assay. The results revealed that hsa_circ_0039569 and HMGA1 were elevated, while miR-197 was downregulated in endometrial carcinoma. Moreover, hsa_circ_0039569 was positively correlated with the expression of HMGA1 and was negatively correlated with the level of miR-197. In addition, hsa_circ_0039569 facilitated the proliferation, migration and invasion of endometrial carcinoma cells. The underlying mechanism is that hsa_circ_0039569 serves as a sponge of miR-197 to repress the inhibitory effect of miR-197 on HMGA1. Furthermore, the miR-197/HMGA1 axis was implicated in endometrial carcinoma progression accelerated by hsa_circ_0039569. Collectively, hsa_circ_0039569 may promote the development of endometrial carcinoma by serving as an endogenous sponge of miR-197, increasing HMGA1 expression and identifying a novel target for endometrial carcinoma treatment.
Collapse
Affiliation(s)
- Yi Zhou
- Third Department of Gynecology and Oncology, Hunan Cancer Hospital, Changsha, Hunan Province, China
| | - Anyi Pan
- Third Department of Gynecology and Oncology, Hunan Cancer Hospital, Changsha, Hunan Province, China
| | - Yudong Zhang
- Department of General Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xinchun Li
- Third Department of Gynecology and Oncology, Hunan Cancer Hospital, Changsha, Hunan Province, China
| |
Collapse
|
13
|
Wei YG, Yang CK, Wei ZL, Liao XW, He YF, Zhou X, Huang HS, Lan CL, Han CY, Peng T. High-Mobility Group AT-Hook 1 Served as a Prognosis Biomarker and Associated with Immune Infiltrate in Hepatocellular Carcinoma. Int J Gen Med 2022; 15:609-621. [PMID: 35058711 PMCID: PMC8765458 DOI: 10.2147/ijgm.s344858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The protein high-mobility group AT-hook 1 (HMGA1) has been demonstrated that modulated cellular proliferation, invasion, and apoptosis with a poor prognosis in miscellaneous carcinomas. However, the mechanism of circumstantial carcinogenesis and association with the immune microenvironment of HMGA1 in hepatocellular carcinoma (HCC) had not been extensively explored. METHODS The gene expression, clinicopathological correlation, and prognosis analysis were performed in the data obtained from TCGA. The results were further validated by ICGC and GEO database and external validation cohort from Guangxi. The HMGA1 protein expression was further examined in the HPA database. Biological function analyses were conducted by GSEA, STRING database, and Coexpedia online tool. Using TIMER and CIBERSORT method, the relationship between immune infiltrate and HMGA1 was investigated. RESULTS In HCC, HMGA1 had much higher transcriptional and proteomic expression than in corresponding paraneoplastic tissue. Patients with high HMGA1 expression had a poor prognosis and unpromising clinicopathological features. High HMGA1 expression was closely related to the cell cycle, tumorigenesis, substance metabolism, and immune processes by regulating complex signaling pathways. Notably, HMGA1 may be associated with TP53 mutational carcinogenesis. Moreover, increased HMGA1 expression may lead to an increase in immune infiltration and a decrease in tumor purity in HCC. CIBERSORT analysis elucidated that the amount of B cell naive, B cell memory, T cells gamma delta, macrophages M2, and mast cell resting decreased when HMGA1 expression was high, whereas T cells follicular helper, macrophages M0, and Dendritic cells resting increased. CONCLUSION In conclusions, HMGA1 is a potent prognostic biomarker and a sign of immune infiltration in HCC, which may be a potential immunotherapy target for HCC.
Collapse
Affiliation(s)
- Yong-Guang Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Cheng-Kun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Zhong-Liu Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xi-Wen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yong-Fei He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Hua-Sheng Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Chen-Lu Lan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Chuang-Ye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
14
|
Development of Biomarker Signatures Associated with Anoikis to Predict Prognosis in Endometrial Carcinoma Patients. JOURNAL OF ONCOLOGY 2022; 2021:3375297. [PMID: 34992654 PMCID: PMC8727165 DOI: 10.1155/2021/3375297] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023]
Abstract
Purpose To generate a signature based on anoikis-related genes (ARGs) for endometrial carcinoma (EC) patients and elucidate the molecular mechanisms in EC. Methods On the basis of TCGA-UCEC dataset, we identified specific anoikis-related genes (ARGs) in EC. Cox-relative regression methods were used to generate an anoikis-related signature (ARS). The possible biological pathways of ARS-related genes were analyzed by GSEA. The clinical potency and immune status of ARS were analyzed by CIBERSORT method, ssGSEA algorithm, Tumor Immune Dysfunction and Exclusion (TIDE) analysis. Moreover, the expression patterns of ARS genes were verified by HPA database. Results Seven anoikis genes (CDKN2A, E2F1, ENDOG, EZH2, HMGA1, PLK1, and SLC2A1) were determined to develop a prognostic ARS. Both genes of ARS were closely bound up with the prognosis of EC patients. The ARS could accurately classify EC cases with different clinical outcome and mirror the specific immune status of EC. We observed that ARS-high patients could not benefit from immunotherapy. Finally, all the hub genes of ARS were proved to be upregulated in EC tissues by immunohistology. Conclusion ARS can be used to stratify the risk and forecast the survival outcome of EC patients and provide prominent reference for individualized treatment in EC.
Collapse
|
15
|
KIFC1 promotes aerobic glycolysis in endometrial cancer cells by regulating the c-myc pathway. J Bioenerg Biomembr 2021; 53:703-713. [PMID: 34729671 DOI: 10.1007/s10863-021-09924-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/29/2021] [Indexed: 10/19/2022]
Abstract
Endometrial cancer (EC) is a common gynecological malignant tumor worldwide. It is imperative to study pathogenesis and therapeutic targets for improving the prognosis of EC. The present study aimed to explore the function and mechanism of kinesin family member C1 (KIFC1) in EC. EC tumor and adjacent normal tissues were collected from 68 pairs of patients. The expression of KIFC1 in tissues and EC cells was analyzed by immunohistochemistry, qRT-PCR or western blot. MTT assay was used to test the cell viability. Flow cytometry was used to determine apoptosis and the cell cycle. Glucose uptake, lactate production, ATP contents and lactate dehydrogenase (LDH) activity were evaluated by a glucose metabolism kit. The expression of HMGA1, c-myc and glycolytic genes was assessed using western blot or qRT-PCR. A mouse xenograft model was established in BALB/c mice to detect tumor growth in vivo. KIFC1 was significantly upregulated in EC tumor tissues compared to adjacent normal control tissues. The upregulated expression of KIFC1 was correlated with poor prognosis in patients. Lentiviral-mediated overexpression of KIFC1 observably enhanced cell viability and reduced the apoptotic rate of Ishikawa and HEC-1B cells. Cell cycle progression was also expedited in the KIFC1 vector group. Moreover, overexpression of KIFC1 elevated glucose uptake, lactate production, ATP contents and LDH activity. However, knockdown of KIFC1 by short hairpin RNA (shRNA) showed the reverse effect on cellular functions. In addition, the expression of c-myc, GLUT1, LDHA and HK2 was increased by the KIFC1 vector. Moreover, HMGA1 regulated the expression of c-myc and glycolytic genes. Upregulated HMGA1 could rescue the effect of KIFC1 knockdown on cellular functions and the expression of glycolytic genes. Finally, KIFC1 knockdown inhibits tumor growth in vivo. The upregulation of KIFC1 was correlated with poor prognosis in EC. KIFC1 promoted aerobic glycolysis in endometrial cancer cells by regulating the HMGA1/c-myc pathway. KIFC1 may be a potential target for the diagnosis and therapy of EC.
Collapse
|
16
|
Wu X, Wang X, Shan L, Zhou J, Zhang X, Zhu E, Yuan H, Wang B. High-mobility group AT-Hook 1 mediates the role of nuclear factor I/X in osteogenic differentiation through activating canonical Wnt signaling. Stem Cells 2021; 39:1349-1361. [PMID: 34028135 DOI: 10.1002/stem.3418] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/29/2021] [Indexed: 11/09/2022]
Abstract
It was previously reported that the loss of the transcription factor nuclear factor I/X (NFIX) gene in mice impaired endochondral ossification and mineralization in bone. However, the cellular and molecular basis for the defect remains unexplored. In this study, we investigated if and how NFIX regulates osteoblast differentiation. Nfix mRNA was induced during osteogenic and adipogenic differentiation of progenitor cells. Loss-of-function and gain-of-function studies revealed that NFIX induced osteoblast differentiation and impaired adipocyte formation from progenitor cells. RNA-seq and promoter analysis revealed that NFIX transcriptionally stimulated the expression of high-mobility group AT-Hook 1 (HMGA1). We then demonstrated that HMGA1 stimulated osteogenic differentiation of progenitor cells at the expense of adipogenic differentiation. The effect of Nfix siRNA on the differentiation of progenitor cells could be attenuated when HMGA1 was simultaneously overexpressed. Further investigations revealed the stimulatory effect of NFIX and HMGA1 on canonical wingless-type MMTV integration site family (Wnt) signaling. HMGA1 transcriptionally activates the expression of low-density lipoprotein receptor-related protein 5. Finally, in vivo transfection of Nfix siRNA to the marrow of mice reduced osteoblasts and increased fat accumulation in the marrow, and inactivated HMGA1/β-catenin signaling in bone marrow mesenchymal stem cells. This study suggests that HMGA1 plays a role in osteoblast commitment and mediates the function of NFIX through transcriptionally activating canonical Wnt signaling.
Collapse
Affiliation(s)
- Xiaowen Wu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Xiaochen Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Liying Shan
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Jie Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Xin Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Endong Zhu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Hairui Yuan
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Baoli Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
17
|
Matsubara K, Matsubara Y, Uchikura Y, Takagi K, Yano A, Sugiyama T. HMGA1 Is a Potential Driver of Preeclampsia Pathogenesis by Interference with Extravillous Trophoblasts Invasion. Biomolecules 2021; 11:biom11060822. [PMID: 34072941 PMCID: PMC8227282 DOI: 10.3390/biom11060822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia (PE) is a serious disease that can be fatal for the mother and fetus. The two-stage theory has been proposed as its cause, with the first stage comprising poor placentation associated with the failure of fertilized egg implantation. Successful implantation and placentation require maternal immunotolerance of the fertilized egg as a semi-allograft and appropriate extravillous trophoblast (EVT) invasion of the decidua and myometrium. The disturbance of EVT invasion during implantation in PE results in impaired spiral artery remodeling. PE is thought to be caused by hypoxia during remodeling failure-derived poor placentation, which results in chronic inflammation. High-mobility group protein A (HMGA) is involved in the growth and invasion of cancer cells and likely in the growth and invasion of trophoblasts. Its mechanism of action is associated with immunotolerance. Thus, HMGA is thought to play a pivotal role in successful pregnancy, and its dysfunction may be related to the pathogenesis of PE. The evaluation of HMGA function and its changes in PE might confirm that it is a reliable biomarker of PE and provide prospects for PE treatment through the induction of EVT proliferation and invasion during the implantation.
Collapse
Affiliation(s)
- Keiichi Matsubara
- Department of Regional Pediatrics and Perinatology, Graduate School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan
- Correspondence:
| | - Yuko Matsubara
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Yuka Uchikura
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Katsuko Takagi
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Akiko Yano
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| | - Takashi Sugiyama
- Department of Obstetrics and Gynecology, School of Medicine, Ehime University, Ehime, Toon-shi 791-0295, Shitsukawa, Japan; (Y.M.); (Y.U.); (K.T.); (A.Y.); (T.S.)
| |
Collapse
|
18
|
Christakoudi S, Evangelou E, Riboli E, Tsilidis KK. GWAS of allometric body-shape indices in UK Biobank identifies loci suggesting associations with morphogenesis, organogenesis, adrenal cell renewal and cancer. Sci Rep 2021; 11:10688. [PMID: 34021172 PMCID: PMC8139988 DOI: 10.1038/s41598-021-89176-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/21/2021] [Indexed: 02/08/2023] Open
Abstract
Genetic studies have examined body-shape measures adjusted for body mass index (BMI), while allometric indices are additionally adjusted for height. We performed the first genome-wide association study of A Body Shape Index (ABSI), Hip Index (HI) and the new Waist-to-Hip Index and compared these with traditional indices, using data from the UK Biobank Resource for 219,872 women and 186,825 men with white British ancestry and Bayesian linear mixed-models (BOLT-LMM). One to two thirds of the loci identified for allometric body-shape indices were novel. Most prominent was rs72959041 variant in RSPO3 gene, expressed in visceral adipose tissue and regulating adrenal cell renewal. Highly ranked were genes related to morphogenesis and organogenesis, previously additionally linked to cancer development and progression. Genetic associations were fewer in men compared to women. Prominent region-specific associations showed variants in loci VEGFA and HMGA1 for ABSI and KLF14 for HI in women, and C5orf67 and HOXC4/5 for ABSI and RSPO3, VEGFA and SLC30A10 for HI in men. Although more variants were associated with waist and hip circumference adjusted for BMI compared to ABSI and HI, associations with height had previously been reported for many of the additional variants, illustrating the importance of adjusting correctly for height.
Collapse
Affiliation(s)
- Sofia Christakoudi
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK. .,MRC Centre for Transplantation, King's College London, Great Maze Pond, London, SE1 9RT, UK.
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK.,Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK.,Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| |
Collapse
|
19
|
Cai Y, Hao M, Chang Y, Liu Y. LINC00665 enhances tumorigenicity of endometrial carcinoma by interacting with high mobility group AT-hook 1. Cancer Cell Int 2021; 21:8. [PMID: 33407473 PMCID: PMC7789558 DOI: 10.1186/s12935-020-01657-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/16/2020] [Indexed: 11/22/2022] Open
Abstract
Background Endometrial carcinoma is a frequently diagnosed cancer among females. LncRNAs are reported to be associated with various cancers. Their biological roles in endometrial carcinoma progression is an emerging scientific area. LINC00665 can exert a significant role in many cancers. However, its potential function in endometrial carcinoma is still poorly known. Method qRT-PCR was carried out to test expression of LINC00665 and HMGA1. Western blot analysis was carried out to detect protein expression of HMGA1. Cell proliferation was evaluated using Cell Counting Kit-8 (CCK-8) and EdU assay. Flow cytometry assay was used to determine cell apoptosis and cell cycle. Wound healing and transwell invasion assay was carried out to test cell migration and invasion. Immunohistochemical staining and HE staining were conducted to assess Ki-67 and tumor growth respectively. Results Expression of LINC00665 in clinical endometrial carcinoma tissues and cells was obviously up-regulated. Loss of LINC00665 could repress endometrial carcinoma cell viability, induce cell apoptosis and block cell cycle in G1 phase. KLE and HHUA cell migration and invasion ability were depressed by LINC00665 shRNA. Decrease of LINC00665 suppressed endometrial carcinoma tumorigenicity in vivo. RIP assay proved that LINC00665 directly bound with HMGA1 protein. shRNA of HMGA1 obviously restrained endometrial carcinoma cell growth and cell invasion. Conclusions LINC00665 might promote endometrial carcinoma progression by positively modulating HMGA1.
Collapse
Affiliation(s)
- Yixuan Cai
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Min Hao
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yue Chang
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yun Liu
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
Cui X, Zhang H, Chen T, Yu W, Shen K. Long Noncoding RNA SNHG22 Induces Cell Migration, Invasion, and Angiogenesis of Gastric Cancer Cells via microRNA-361-3p/HMGA1/Wnt/β-Catenin Axis. Cancer Manag Res 2020; 12:12867-12883. [PMID: 33364835 PMCID: PMC7751299 DOI: 10.2147/cmar.s281578] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background The correlation between long non-coding RNAs (lncRNAs) and gastric cancer (GC) has been indicated. As a newly found lncRNA, small nucleolar RNA host gene 22 (SNHG22) functions as an oncogene in ovarian carcinoma and breast cancer. However, its action has not been explored in GC. Herein, the purpose of the current research was to examine the influence of SNHG22 on GC development. Methods RT-qPCR was used to identify SNHG22 and microRNA-361-3p (miR-361-3p) in GC tissues and cells. Functional assays were implemented to measure changes on biological activities of GC cells under different transfections. Besides, after human umbilical vein endothelial cells (HUVECs) were co-cultured with supernatant of transfected GC cells, angiogenesis was assessed by tube formation assay in vitro. HMGA1 and β-catenin expression were determined. Finally, mechanistic assays, including RNA pull-down assay and dual-luciferase reporter assay, were employed to assess relationships among SNHG22, miR-361-3p, and HMGA1. Results SNHG22 and HMGA1 were highly expressed but miR-361-3p was poorly expressed in GC tissues. Mechanistically, SNHG22 bound to miR-361-3p, and miR-361-3p targeted HMGA1 to disrupt the Wnt/β-catenin pathway. Following SNHG22 or HMGA1 silencing or miR-361-3p upregulation, we observed a decline of proliferation, migration, and invasion of GC cells and HUVEC angiogenesis but acceleration of GC cell apoptosis and cell cycle arrest. Conclusion Collectively, SNHG22 silencing possessed tumor-suppressing potentials in GC development via Wnt/β-catenin pathway by binding to miR-361-3p and downregulating HMGA1, highlighting a new promising road for GC treatment development.
Collapse
Affiliation(s)
- Xiaofeng Cui
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Huaiyu Zhang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Tong Chen
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Wei Yu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Kexin Shen
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, People's Republic of China
| |
Collapse
|
21
|
Chen M, Xu K, Li B, Wang N, Zhang Q, Chen L, Zhang D, Yang L, Xu Z, Xu H. HMGA1 Regulates the Stem Cell-Like Properties of Circulating Tumor Cells from GIST Patients via Wnt/β-Catenin Pathway. Onco Targets Ther 2020; 13:4943-4956. [PMID: 32606726 PMCID: PMC7296980 DOI: 10.2147/ott.s249063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022] Open
Abstract
Background Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the digestive system. Circulating tumor cells (CTCs) have been proven to be critical in the recurrence and metastasis of diseases; however, the characteristics of CTCs of GIST are still unclear. Methods We sorted out and verified the validity of CTCs from peripheral blood of gastrointestinal stromal tumor (GIST) patients with or without heterochronous liver metastasis using flow cytometry (FCM). Differential genes were analyzed between the GIST patients with and without liver metastasis using next-generation sequencing (NGS). Results The preliminary study on the characteristics of CTCs revealed that CTCs of GIST patients with heterochronous liver metastasis had stronger stem cell-like properties (SC-like properties) than CTCs of those without liver metastasis. Furthermore, NGS followed with a series of assays revealed that HMGA1 played a critical role in regulating the SC-like properties of CTCs. Mechanistically, HMGA1 could activate Wnt/β-catenin pathway in vitro and vivo. Moreover, we found that the expression level of HMGA1 in CTCs was an independent risk factor probably influencing the prognosis of GIST patients. Conclusion Our findings indicate the significant role of HMGA1 in SC-like properties, IM resistance and eventually hepatic metastasis formation of CTCs. Targeting HMGA1 in CTCs may be a therapeutic strategy for GIST patients with hepatic metastasis.
Collapse
Affiliation(s)
- Ming Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Kangjing Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Nuofan Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Qiang Zhang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222000, People's Republic of China
| | - Liang Chen
- Department of General Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, People's Republic of China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| | - Hao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medical University, Nanjing, Jiangsu 211166, People's Republic of China
| |
Collapse
|
22
|
Xie Y, Zhong L, Duan D, Li T. Casticin inhibits invasion and proliferation via downregulation of β-catenin and reversion of EMT in oral squamous cell carcinoma. J Oral Pathol Med 2019; 48:897-905. [PMID: 31318467 DOI: 10.1111/jop.12930] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/14/2019] [Accepted: 07/04/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Casticin expresses multiple anti-cancer activities, whereas the effect of casticin on oral squamous cell carcinoma (OSCC) is still unclear. β-catenin signaling plays a crucial role in the epithelial-mesenchymal transition which is closely related to tumorigenesis. Herein, we aimed to study the functions of casticin on invasion and migration of OSCC, and clarify whether the effect of casticin on OSCC has a relationship with β-catenin signaling. METHODS Human OSCC cell lines UM1 and HSC-3 were treated with different concentrations of casticin. The cell viability was evaluated by MTT and soft agar colony formation. Transwell assay and wound-healing assay were performed to measure the ability of cell invasion and migration. The protein expression was assessed by Western blotting. RESULTS Casticin displayed inhibitory activities of cell viability, invasion, and migration on OSCC cell lines. Meanwhile, casticin could reverse EMT process and inhibit the expression of β-catenin in OSCC. Knock-down or overexpression of β-catenin could alter the effect of casticin on OSCC. CONCLUSIONS Casticin impaired invasion and migration of OSCC by inhibition of β-catenin and reversal of EMT and could be a potential anti-cancer bioactive agent.
Collapse
Affiliation(s)
- Yaxin Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liang Zhong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dingyu Duan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Taiwen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Zhang K, Li H, Yan Y, Zang Y, Li K, Wang Y, Xue F. Identification of key genes and pathways between type I and type II endometrial cancer using bioinformatics analysis. Oncol Lett 2019; 18:2464-2476. [PMID: 31452737 PMCID: PMC6676660 DOI: 10.3892/ol.2019.10550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/17/2019] [Indexed: 12/24/2022] Open
Abstract
Endometrial carcinoma (EC) is a common malignant neoplasm of the female reproductive tract. The malignant degree of type II EC is much greater than that of type I EC, usually presenting with a high recurrence rate and a poor prognosis. Therefore, the present study aimed to examine the principal genes associated with the degree of differentiation in type I and type II EC and reveal their potential mechanisms. Differentially expressed genes (DEGs) were selected from the gene expression profiles derived from The Cancer Genome Atlas. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted. In the present study, the KEGG pathway enrichment analysis revealed that 5,962 upregulated DEGs were significantly enriched in the ‘p53 signaling pathway’ and involved in ‘lysine degradation’. In addition, 3,709 downregulated DEGs were enriched in ‘pathways in cancer’, as well as ‘tight junction regulation’, the ‘cell cycle’ and the ‘Wnt signaling pathway’. The 13 top hub genes MAPK1, PHLPP1, ESR1, MDM2, CDKN2A, CDKN1A, AURKA, BCL2L1, POLQ, PIK3R3, RHOQ, EIF4E and LATS2 were identified via the protein-protein interaction network. Furthermore, the OncoPrint algorithm from cBioPortal declared that 25% of EC cases carried genetic alterations. The altered DEGs (MAPK1, MDM2, AURKA, EIF4E and LATS2) may be involved in tumor differentiation and may be valuable diagnostic biomarkers. In conclusion, a number of principal genes were identified in the present study that may be determinants of poorly differentiated type II EC carcinogenesis, which may contribute to future research into potential molecular mechanisms. In addition, these genes may help identify candidate biomarkers and novel therapeutic targets for type II EC.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Huiyang Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yuqin Zang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Ke Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
24
|
Huang X, Zhong R, He X, Deng Q, Peng X, Li J, Luo X. Investigations on the mechanism of progesterone in inhibiting endometrial cancer cell cycle and viability via regulation of long noncoding RNA NEAT1/microRNA-146b-5p mediated Wnt/β-catenin signaling. IUBMB Life 2018; 71:223-234. [PMID: 30452118 DOI: 10.1002/iub.1959] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 11/12/2022]
Abstract
Progesterone is often used to protect the endometrium and prevent endometrial cancer. An intensive study on its molecular mechanism in endometrial cancer would contribute to the development of more promising therapies. Relevant lncRNAs and mRNAs expression data in endometrial cancer cell line Ishikawa pretreated and post-treated with progesterone were derived from Gene Expression Omnibus (accession no. GSE29435), and then we analyzed long noncoding RNAs and mRNAs with differential expressions in two different conditions. The Cytoscape software, TargetScan, miRanda, and Human microRNA Disease Database (HMDD) websites were employed. Gene set enrichment analysis (GSEA) was used to determine related Kyoto Encyclopedia of Genes and Genomes pathways alteration in Ishikawa cells treated with progesterone. In addition to bioinformatics analysis, Reverse Transcription-Polymerase Chain Reaction (RT-PCR), Western blot, and dual-luciferase reporter assays were performed. The impact of progesterone on cell propagation and cell cycle was testified by colony formation and flow cytometry analysis. LncRNA nuclear enriched abundant transcript 1 (NEAT1) was the most significantly downregulated lncRNA in endometrial cancer cells treated with progesterone. Lymphoid enhancing factor 1 (LEF1) was positively associated with NEAT1, and eventually hsa_miR-146b-5p was validated to target both LEF1 and NEAT1. Wnt/β-catenin signaling pathway was identified to involve in endometrial cancer. NEAT1 or LEF1 was overexpressed in endometrial cancer cells while downregulated following post-treatment with progesterone. Conversely, miR-146b-5p was notably decreased in Ishikawa cells while upregulated after treatment with progesterone. Downstream gene c-myc or MMP9 regulated by upstream gene LEF1 in Wnt/β-catenin signaling pathway was remarkably increased in Ishikawa cells and positively related with NEAT1. Progesterone inhibited cell cycle and viability through regulating NEAT1/miR-146b-5p axis via Wnt/β-catenin signaling pathway. Progesterone exerted suppressive influence on endometrial cancer progression via regulation of lncRNA NEAT1/miR-146b-5p-mediated Wnt/β-catenin signaling pathway, which might reveal new strategies for developing more effective therapeutics. © 2018 IUBMB Life, 71(1):223-234, 2019.
Collapse
Affiliation(s)
- Xiaohui Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People's Republic of China.,Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Rui Zhong
- Department of Ultrasound, Guangdong Province Traditional Chinese Medical Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Xiukui He
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Qingshan Deng
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Xiuhong Peng
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Jieming Li
- Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Xiping Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People's Republic of China.,Department of Gynecology, Guangdong Women and Children Hospital, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
25
|
Wang C, Wang X, Tang J, Chen H, Zhang J, Li Y, Lei S, Ji H, Yang B, Ren J, Ding N. Genome-wide association studies for two exterior traits in Chinese Dongxiang spotted pigs. Anim Sci J 2018; 89:868-875. [DOI: 10.1111/asj.13003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/11/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Chengbin Wang
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Xiaopeng Wang
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Jianhong Tang
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Hao Chen
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Junjie Zhang
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Yiping Li
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Shengrong Lei
- National Conservation Farm of Dongxiang Spotted Pigs; Dongxiang China
| | - Huayuan Ji
- Institute of Animal Husbandry and Veterinary; Jiangxi Academy of Agricultural Science; Nanchang China
| | - Bin Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Jun Ren
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| | - Nengshui Ding
- State Key Laboratory of Pig Genetic Improvement and Production Technology; Jiangxi Agricultural University; Nanchang China
| |
Collapse
|
26
|
Du Q, Liu J, Zhang X, Zhang X, Zhu H, Wei M, Wang S. Propofol inhibits proliferation, migration, and invasion but promotes apoptosis by regulation of Sox4 in endometrial cancer cells. ACTA ACUST UNITED AC 2018; 51:e6803. [PMID: 29490000 PMCID: PMC5856446 DOI: 10.1590/1414-431x20176803] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/14/2017] [Indexed: 01/27/2023]
Abstract
Propofol is an intravenous sedative hypnotic agent of which the growth-inhibitory effect has been reported on various cancers. However, the roles of propofol in endometrial cancer (EC) remain unclear. This study aimed to explore the effects of propofol on EC in vitro and in vivo. Different concentrations of propofol were used to treat Ishikawa cells. Colony number, cell viability, cell cycle, apoptosis, migration, and invasion were analyzed by colony formation, MTT, flow cytometry, and Transwell assays. In addition, the pcDNA3.1-Sox4 and Sox4 siRNA plasmids were transfected into Ishikawa cells to explore the relationship between propofol and Sox4 in EC cell proliferation. Tumor weight in vivo was measured by xenograft tumor model assay. Protein levels of cell cycle-related factors, apoptosis-related factors, matrix metalloproteinases 9 (MMP9), matrix metalloproteinases 2 (MMP2) and Wnt/β-catenin pathway were examined by western blot. Results showed that propofol significantly decreased colony numbers, inhibited cell viability, migration, and invasion but promoted apoptosis in a dose-dependent manner in Ishikawa cells. Moreover, propofol reduced the expression of Sox4 in a dose-dependent manner. Additionally, propofol significantly suppressed the proportions of Ki67+ cells, but Sox4 overexpression reversed the results. Furthermore, in vivo assay results showed that propofol inhibited tumor growth; however, the inhibitory effect was abolished by Sox4 overexpression. Moreover, propofol inhibited Sox4 expression via inactivation of Wnt/β-catenin signal pathway. Our study demonstrated that propofol inhibited cell proliferation, migration, and invasion but promoted apoptosis by regulation of Sox4 in EC cells. These findings might indicate a novel treatment strategy for EC.
Collapse
Affiliation(s)
- Qing Du
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Xuezhi Zhang
- Department of Emergency, Qingdao University, Qingdao, China
| | - Xin Zhang
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - He Zhu
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Ming Wei
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Shilei Wang
- Department of Anesthesiology, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Xie F, Xiang X, Huang Q, Ran P, Yuan Y, Li Q, Qi G, Guo X, Xiao C, Zheng S. Reciprocal control of lncRNA-BCAT1 and β-catenin pathway reveals lncRNA-BCAT1 long non-coding RNA acts as a tumor suppressor in colorectal cancer. Oncotarget 2018; 8:23628-23637. [PMID: 28416735 PMCID: PMC5410332 DOI: 10.18632/oncotarget.15466] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/27/2016] [Indexed: 12/15/2022] Open
Abstract
β-catenin plays a major role in tumor development and progression. The present study found that β-catenin was upregulated in 30 samples of colorectal cancer (CRC) tissue as compared to adjacent non-tumor tissues. Analysis of long non-coding RNA (lncRNA) expression profiles using the GSE18560 and GSE44097 datasets, which were generated via the Affymetrix plus 2.0 microarray platform and downloaded from the GEO database, revealed 20 differentially expressed lncRNAs following β-catenin knockdown. We focused on AK091631, a novel lncRNA, which we named lncRNA-β-catenin associated transcript 1 (LncRNA-BCAT1). lncRNA-BCAT1 expression was decreased in CRC tissues, and was negatively associated with β-catenin in both CRC tissues and cell lines. lncRNA-BCAT1 overexpression suppressed CRC cell growth and invasion by downregulating cyclin D1, c-Myc, and MMP-2. These results suggest that lncRNA-BCAT1 overexpression inhibits CRC cell growth and invasion via Wnt/β-catenin pathway blockade, and that lncRNA-BCAT1 is repressed by Wnt/β-catenin signaling. This evidence suggests that lncRNA-BCAT1 is a tumor suppressor and that lncRNA-BCAT1 may be an effective prognostic biomarker in CRC.
Collapse
Affiliation(s)
- Fei Xie
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Xudong Xiang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Qionglin Huang
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Pengzhan Ran
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Yuncang Yuan
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Qian Li
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Guoxiang Qi
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Xiaopeng Guo
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Chunjie Xiao
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Shangyong Zheng
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| |
Collapse
|
28
|
Ong MS, Cai W, Yuan Y, Leong HC, Tan TZ, Mohammad A, You ML, Arfuso F, Goh BC, Warrier S, Sethi G, Tolwinski NS, Lobie PE, Yap CT, Hooi SC, Huang RY, Kumar AP. 'Lnc'-ing Wnt in female reproductive cancers: therapeutic potential of long non-coding RNAs in Wnt signalling. Br J Pharmacol 2017; 174:4684-4700. [PMID: 28736855 PMCID: PMC5727316 DOI: 10.1111/bph.13958] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/30/2017] [Accepted: 07/17/2017] [Indexed: 02/06/2023] Open
Abstract
Recent discoveries in the non-coding genome have challenged the original central dogma of molecular biology, as non-coding RNAs and related processes have been found to be important in regulating gene expression. MicroRNAs and long non-coding RNAs (lncRNAs) are among those that have gained attention recently in human diseases, including cancer, with the involvement of many more non-coding RNAs (ncRNAs) waiting to be discovered. ncRNAs are a group of ribonucleic acids transcribed from regions of the human genome, which do not become translated into proteins, despite having essential roles in cellular physiology. Deregulation of ncRNA expression and function has been observed in cancer pathogenesis. Recently, the roles of a group of ncRNA known as lncRNA have gained attention in cancer, with increasing reports of their oncogenic involvement. Female reproductive cancers remain a leading cause of death in the female population, accounting for almost a third of all female cancer deaths in 2016. The Wnt signalling pathway is one of the most important oncogenic signalling pathways which is hyperactivated in cancers, including female reproductive cancers. The extension of ncRNA research into their mechanistic roles in human cancers has also led to novel reported roles of ncRNAs in the Wnt pathway and Wnt-mediated oncogenesis. This review aims to provide a critical summary of the respective roles and cellular functions of Wnt-associated lncRNAs in female reproductive cancers and explores the potential of circulating cell-free lncRNAs as diagnostic markers and lncRNAs as therapeutic targets. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Mei S Ong
- Departments of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Wanpei Cai
- Departments of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
| | - Yi Yuan
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
| | - Hin C Leong
- Departments of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
| | - Tuan Z Tan
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
| | - Asad Mohammad
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
| | - Ming L You
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research InstituteCurtin UniversityPerthWAAustralia
| | - Boon C Goh
- Departments of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
- National University Cancer InstituteNational University Health SystemSingapore
- Department of Haematology‐OncologyNational University Health SystemSingapore
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal UniversityBangaloreIndia
- School of Biomedical Sciences, Curtin Health Innovation Research InstituteCurtin UniversityPerthWAAustralia
| | - Gautam Sethi
- Departments of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- School of Biomedical Sciences, Curtin Health Innovation Research InstituteCurtin UniversityPerthWAAustralia
| | - Nicholas S Tolwinski
- Division of ScienceYale‐NUS CollegeSingapore
- Department of Biological ScienceNational University of SingaporeSingapore
| | - Peter E Lobie
- Departments of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Departments of Anatomy, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Tsinghua Berkeley Shenzhen Institute and Division of Life Science and HealthTsinghua University Graduate SchoolShenzhenChina
| | - Celestial T Yap
- Departments of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- National University Cancer InstituteNational University Health SystemSingapore
| | - Shing C Hooi
- Departments of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Ruby Y Huang
- Departments of Anatomy, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
- National University Cancer InstituteNational University Health SystemSingapore
- Department of Obstetrics and GynaecologyNational University HospitalSingapore
| | - Alan P Kumar
- Departments of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
- National University Cancer InstituteNational University Health SystemSingapore
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal UniversityBangaloreIndia
- Curtin Medical School, Faculty of Health ScienceCurtin UniversityPerthWAAustralia
- Department of Biological SciencesUniversity of North TexasDentonTXUSA
| |
Collapse
|
29
|
Wang J, Zhao X, Guo Z, Ma X, Song Y, Guo Y. Regulation of NEAT1/miR-214-3p on the growth, migration and invasion of endometrial carcinoma cells. Arch Gynecol Obstet 2017; 295:1469-1475. [PMID: 28447190 DOI: 10.1007/s00404-017-4365-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/30/2017] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the function and mechanism of lnc NEAT1 in regulating the growth, migration and invasion of endometrial carcinoma (EC) cells. MATERIALS AND METHODS NEAT1 and miR-214-3p levels were measured by qRT-PCR. The protein levels of HMGA1, β-catenin, c-myc and MMP9 were evaluated by Western blot. The effects of NEAT1, HMGA1, miR-214-3p on the viability, migration and invasion of HEC-1A cells were accessed by WST-1 assay and transwell migration/invasion assay. The effect of miR-214-3p on Wnt signaling activity was tested by luciferase reporter assay. RESULTS NEAT1, HMGA1 and β-catenin were significantly upregulated in EC tissues, and miR-214-3p was significantly downregulated. NEAT1 promoted the growth, migration and invasion of HEC-1A cells, and mRNA level of Wnt/β-catenin downstream genes c-myc and MMP9. In addition, HMGA1 upregualted the protein and mRNA levels of Wnt/β-catenin downstream genes c-myc and MMP9, and could improve cell viability, and increase numbers of migration and invasion of HEC-1A cells. miR-214-3p overexpression inhibited the proliferation, migration and invasion of HEC-1A cells, while NEAT1 overexpression reversed these effects. miR-214-3p overexpression inhibited the activity of Wnt/β-catenin pathway, while NEAT1 overexpression reversed this effect. Then, si-HMGA1 reduced the activity of Wnt/β-catenin pathway. Moreover, we found NEAT1 and HMGA1 bound to miR-214-3p by luciferase reporter assay, and NEAT1 and HMGA1 expression were negatively correlated with miR-214-3p. CONCLUSION NEAT1 regulates HMGA1 via miR-214-3p to regulate Wnt/β-catenin pathway, thus promotes the growth, migration and invasion of HEC-1A cells.
Collapse
Affiliation(s)
- Jian Wang
- Department of Obstetric and Gynecology, The Third Hospital of Hebei Medical University, 139 Ziqiang Rd., Shijiazhuang, Hebei, 050051, People's Republic of China.
| | - Xiangzhai Zhao
- Department of Obstetric and Gynecology, The Third Hospital of Hebei Medical University, 139 Ziqiang Rd., Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Zhaojun Guo
- Department of Obstetric and Gynecology, The Third Hospital of Hebei Medical University, 139 Ziqiang Rd., Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Xiaolin Ma
- Department of Obstetric and Gynecology, The Third Hospital of Hebei Medical University, 139 Ziqiang Rd., Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Yueqing Song
- Department of Obstetric and Gynecology, The Third Hospital of Hebei Medical University, 139 Ziqiang Rd., Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Ying Guo
- Department of Obstetric and Gynecology, The Third Hospital of Hebei Medical University, 139 Ziqiang Rd., Shijiazhuang, Hebei, 050051, People's Republic of China
| |
Collapse
|
30
|
Komina A, Palkina N, Aksenenko M, Tsyrenzhapova S, Ruksha T. Antiproliferative and Pro-Apoptotic Effects of MiR-4286 Inhibition in Melanoma Cells. PLoS One 2016; 11:e0168229. [PMID: 28005927 PMCID: PMC5179095 DOI: 10.1371/journal.pone.0168229] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 11/28/2016] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION MicroRNAs are essential regulators of gene expression at the post-transcriptional level. Their expression is altered in cancer tissues, and evaluation of these alterations is considered a promising tool used to diagnose and identify prognostic markers. MATERIALS AND METHODS The microRNA expression profiles of formalin-fixed, paraffin-embedded melanoma and melanocytic nevi samples were estimated with a microarray and subsequently validated by real-time PCR. Melanoma cells were transfected with miR-4286 inhibitor to evaluate the influence of this microRNA on the viability, proliferation, apoptosis, migration, and invasion of melanoma cells. RESULTS The microarray revealed that the expression of 1,171 microRNAs was altered in melanoma samples compared to melanocytic nevi. Real-time PCR validation experiments found the microRNA expression levels to correspond to the melanoma/melanocytic nevi microarray results. The pathway analysis identified 52 modulated pathways in melanoma. Moreover, the application of miR-4286 inhibitor to BRO melanoma cells resulted in a 2.6-fold increase in the apoptosis rate and a 1.7-fold decrease in the cell proliferation/viability but did not affect the invasiveness and migration of these cells. Furthermore, the use of miR-4286 inhibitor altered the mRNA expression of several miR-4286 gene targets: folylpolyglutamate synthase, RNA polymerase I-specific transcription initiation factor, apelin, G-protein-coupled receptor 55, and high-mobility group A1 protein, which have been implicated in cell proliferation/apoptosis regulation. Lastly, the transiently transfected SK-MEL-1 cells with miR-4286 inhibitor decreased proliferation rate and modulated folylpolyglutamate synthase rates of these cells. CONCLUSION Our results demonstrate that miR-4286 mediates proliferation and apoptosis in melanoma cells, these findings may represent a novel mechanism underlying these processes.
Collapse
Affiliation(s)
- Anna Komina
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation
| | - Nadezhda Palkina
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation
| | - Mariya Aksenenko
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation
| | - Seseg Tsyrenzhapova
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation
| | - Tatiana Ruksha
- Department of Pathophysiology, Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation
- * E-mail:
| |
Collapse
|
31
|
Yang Y, Bai YS, Wang Q. CDGSH Iron Sulfur Domain 2 Activates Proliferation and EMT of Pancreatic Cancer Cells via Wnt/β-Catenin Pathway and Has Prognostic Value in Human Pancreatic Cancer. Oncol Res 2016; 25:605-615. [PMID: 27983920 PMCID: PMC7841247 DOI: 10.3727/096504016x14767450526417] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Recently, increasing evidence has shown that CDGSH iron sulfur domain 2 (CISD2) is involved in the initiation and metastasis of several cancers. However, the evidence of its potential role in pancreatic cancer is still lacking. In our present study, CISD2 was found to be increased in pancreatic cancer samples and multiple cell lines. Moreover, statistical analysis revealed that a high level of CISD2 was related to advanced clinical stage, advanced T-stage, positive vascular invasion, positive distant metastasis, and larger tumor size. In addition, multivariate analysis suggests that CISD2 was an independent prognostic factor in pancreatic cancer. Importantly, downregulation of CISD2 was capable of inhibiting the survival and growth of pancreatic cancer cells. Mechanistic study showed that inactivation of the Wnt/β-catenin pathway contributed to the CISD2 deficit-induced death of pancreatic cancer cells. Furthermore, we showed that CISD2 silencing significantly inhibited EMT via the Wnt/β-catenin pathway. Finally, in nude mice, the CISD2 deficit suppressed the tumorigenesis of pancreatic cancer cells. Collectively, our study demonstrated that CISD2 could be an independent prognostic factor for pancreatic cancer and suggested that the CISD2/Wnt/β-catenin pathway contributes to the proliferation of pancreatic cancer cells and EMT, hinting at a novel promising molecular target in the therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Yuan-Song Bai
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Qing Wang
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|