1
|
Alimohammadi M, Abolghasemi H, Cho WC, Reiter RJ, Mafi A, Aghagolzadeh M, Hushmandi K. Interplay between LncRNAs and autophagy-related pathways in leukemia: mechanisms and clinical implications. Med Oncol 2025; 42:154. [PMID: 40202565 DOI: 10.1007/s12032-025-02710-8] [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: 02/03/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025]
Abstract
Autophagy is a conserved catabolic process that removes protein clumps and defective organelles, thereby promoting cell equilibrium. Growing data suggest that dysregulation of the autophagic pathway is linked to several cancer hallmarks. Long non-coding RNAs (lncRNAs), which are key parts of gene transcription, are increasingly recognized for their significant roles in various biological processes. Recent studies have uncovered a strong connection between the mutational landscape and altered expression of lncRNAs in the tumor formation and development, including leukemia. Research over the past few years has emphasized the role of lncRNAs as important regulators of autophagy-related gene expression. These RNAs can influence key leukemia characteristics, such as apoptosis, proliferation, epithelial-mesenchymal transition (EMT), migration, and angiogenesis, by modulating autophagy-associated signaling pathways. With altered lncRNA expression observed in leukemia cells and tissues, they hold promise as diagnostic biomarkers and therapeutic targets. The current review focuses on the regulatory function of lncRNAs in autophagy and their involvement in leukemia, potentially uncovering valuable therapeutic targets for leukemia treatment.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Abolghasemi
- Department of Pediatrics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahboobeh Aghagolzadeh
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Huang M, Cai J, Zeng H, Zhu Y, Zhang F, Li S. miR-103 promotes esophageal squamous cell carcinoma metastasis by targeting FOXP1. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2025:1-14. [PMID: 40117454 DOI: 10.1080/15257770.2025.2478980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/17/2025] [Accepted: 03/08/2025] [Indexed: 03/23/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC), a prevalent malignancy within the digestive tract, is associated with a significantly high mortality rate. MicroRNAs were already demonstrated to work in a wide range of tumors. The objective of the present research was to elucidate the involvement of miR-103 in the pathogenesis of ESCC and to explore its underlying mechanisms of action. Real-time quantitative polymerase chain reaction was used to detect miR-103 expressions in ESCC tissues and cells. The clinical significance of these expressions was assessed by a series of statistical analyses. Transwell assay was used to study the impact of miR-103 on migration and invasion ability of ESCC cells. Furthermore, a dual luciferase reporter gene method was adopted to study the association of miR-103 with the targeting of forkhead box protein 1 (FOXP1). miR-103 was significantly up-regulation in ESCC tissues and cell lines. Clinically, high miR-103 expression was associated with negative prognosis in ESCC. The low miR-103 expression significantly inhibited cell proliferation, migration and invasion in ESCC cell lines. Furthermore, miR-103 regulated the mechanism of action of ESCC by targeting FOXP1. In this study, we found that miR-103 may serve as a biomarker for ESCC prognosis. miR-103 may promote ESCC cell metastasis by targeting FOXP1. These studies may elucidate the potential of miR-103 as a novel target for the treatment of ESCC.
Collapse
Affiliation(s)
- Min Huang
- Department of Oncology, The First People's Hospital of Jingzhou City, Jingzhou, China
| | - Jun Cai
- Department of Oncology, The First People's Hospital of Jingzhou City, Jingzhou, China
| | - Hai Zeng
- Department of Oncology, The First People's Hospital of Jingzhou City, Jingzhou, China
| | - Yan Zhu
- Department of Oncology, The First People's Hospital of Jingzhou City, Jingzhou, China
| | - Fan Zhang
- Department of Oncology, The First People's Hospital of Jingzhou City, Jingzhou, China
| | - Shuang Li
- Department of Oncology, The First People's Hospital of Jingzhou City, Jingzhou, China
| |
Collapse
|
3
|
An Z, Wang J, Li C, Tang C. Signal integrator function of CXXC5 in Cancer. Cell Commun Signal 2025; 23:25. [PMID: 39806388 PMCID: PMC11730785 DOI: 10.1186/s12964-024-02005-x] [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: 10/04/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
CXXC type zinc finger protein 5 (CXXC5) is a member of the ZF-CXXC family and plays a pivotal role in signal integration and information transfer within cell signaling network. CXXC5 acts as a regulator in various physiological processes, and abnormalities in its protein structure or function have been linked to multiple pathological processes. In this article, we correspondingly describe the composition of the ZF-CXXC family, emphatically introducing the features of the CXXC5 gene and protein, review the role of CXXC5 in cellular signaling networks, the physiological and pathological processes associated with CXXC5 dysregulation, and particularly focus on the correlation between CXXC5 and cancers. Finally, we summarize the current therapies targeting CXXC5 and their potential applications, and discuss the intriguing findings from current studies, and the opportunities and challenges in future.
Collapse
Affiliation(s)
- Zihao An
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Jiepu Wang
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chengzuo Li
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chao Tang
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
4
|
Xian D, Yang S, Liu Y, Liu Q, Huang D, Wu Y. MicroRNA-196a-5p facilitates the onset and progression via targeting ITM2B in esophageal squamous cell carcinoma. Pathol Int 2024; 74:129-138. [PMID: 38289121 DOI: 10.1111/pin.13408] [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: 09/15/2023] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 03/21/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent malignancy affecting the digestive tract, with an increasing incidence rate worldwide. Recently, numerous studies revealed that microRNAs were associated with gene expression regulation, particularly their involvement in the regulation of tumor cells, garnering widespread attention. Here, we discovered that miR-196a-5p was significantly upregulated in both ESCC tissues and cells, which was correlated with an unfavorable prognosis. Series functional in vitro investigations have confirmed that silencing miR-196a-5p obviously restrained the ESCC cells malignant phenotypes and promoted apoptosis. Bioinformatics analysis and rescue experiments revealed that miR-196a-5p directly targeted ITM2B, exerting influence on the development of ESCC cells through negative regulation of ITM2B expression. Xenograft mouse models were established for conducting in vivo experiments, providing further confirmation of the regulatory mechanism and biological significance of the miR-196a-5p/ITM2B axis in ESCC. Our research demonstrated miR-196a-5p promoted ESCC malignant progression by interacting with ITM2B, thereby providing novel clues and potential targets for the new diagnosis and thereby of ESCC.
Collapse
Affiliation(s)
- Dubiao Xian
- Cardiothoracic Surgery, The First Affiliated Hospital of Hainan Medical College, The Respiratory Medical Center of Hainan Province, Haikou, Hainan, China
| | - Shubo Yang
- Cardiothoracic Surgery, The First Affiliated Hospital of Hainan Medical College, The Respiratory Medical Center of Hainan Province, Haikou, Hainan, China
| | - Yunzhong Liu
- Cardiothoracic Surgery, The First Affiliated Hospital of Hainan Medical College, The Respiratory Medical Center of Hainan Province, Haikou, Hainan, China
| | - Qingfeng Liu
- Cardiothoracic Surgery, The First Affiliated Hospital of Hainan Medical College, The Respiratory Medical Center of Hainan Province, Haikou, Hainan, China
| | - Ding Huang
- Cardiothoracic Surgery, The First Affiliated Hospital of Hainan Medical College, The Respiratory Medical Center of Hainan Province, Haikou, Hainan, China
| | - Yuechang Wu
- Cardiothoracic Surgery, The First Affiliated Hospital of Hainan Medical College, The Respiratory Medical Center of Hainan Province, Haikou, Hainan, China
| |
Collapse
|
5
|
Wang H. The RNA m6A writer RBM15 contributes to the progression of esophageal squamous cell carcinoma by regulating miR-3605-5p/KRT4 pathway. Heliyon 2024; 10:e24459. [PMID: 38312624 PMCID: PMC10835169 DOI: 10.1016/j.heliyon.2024.e24459] [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: 10/17/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
Cancer progression can be modulated by N6-methyladenosine (m6A) modification. RNA binding motif protein 15 (RBM15) is an essential RNA m6A writer that influences carcinogenesis, however its significance in esophageal squamous cell carcinoma (ESCC) is uncertain. This research is intended to examine how RBM15 regulates the development of ESCC. We performed qRT-PCR analysis to evaluate the expression of RBM15, microRNA (miR-3605-5p) as well as keratin 4 (KRT4) in ESCC. Target relationship between miR-3605-5p and KRT4 was validated by dual luciferase reporter assay. Western blotting analyzed the protein levels of KRT4, p53, and p21. To demonstrate that RBM15 is responsible for the m6A alteration of miR-3605-5p, RIP and Me-RIP experiments were carried out concurrently. m6A content was measured by m6A quantification assay. Cell growth and migration were assessed using the CCK-8 and transwell assays. In addition, the role of RBM15 in vivo was examined using a mouse tumor xenograft model. RBM15 and miR-3605-5p were both substantially expressed in ESCC, however KRT4 was not expressed highly. Overexpressed RBM15 triggered cell proliferation and migration in ESCC. Besides, RBM15/m6A could mediate pri-3605-5p to form the mature miR-3605-5p, and miR-3605-5p further targeted KRT4. Further investigations showed that upregulation of KRT4 overturned the promoting impact of RBM15 overexpression on cell proliferation as well as on cell migration in ESCC by activating p53 signaling pathway. This work implied the carcinogenic activity of RBM15/m6A in ESCC via miR-3605-5p/KRT4 pathway, providing a novel m6A modification pattern in the tumorigenesis of ESCC.
Collapse
Affiliation(s)
- Huan Wang
- General practice section, Wuhan University of Science and Technology Hospital, Wuhan, 430070, Hubei, China
| |
Collapse
|
6
|
Liu WJ, Zhao Y, Chen X, Miao ML, Zhang RQ. Epigenetic modifications in esophageal cancer: An evolving biomarker. Front Genet 2023; 13:1087479. [PMID: 36704345 PMCID: PMC9871503 DOI: 10.3389/fgene.2022.1087479] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Esophageal cancer is a widespread cancer of the digestive system that has two main subtypes: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EA). In the diverse range of cancer therapy schemes, the side effects of conventional treatments remain an urgent challenge to be addressed. Therefore, the pursuit of novel drugs with multiple targets, good efficacy, low side effects, and low cost has become a hot research topic in anticancer therapy. Based on this, epigenetics offers an attractive target for the treatment of esophageal cancer, where major mechanisms such as DNA methylation, histone modifications, non-coding RNA regulation, chromatin remodelling and nucleosome localization offer new opportunities for the prevention and treatment of esophageal cancer. Recently, research on epigenetics has remained at a high level of enthusiasm, focusing mainly on translating the basic research into the clinical setting and transforming epigenetic alterations into targets for cancer screening and detection in the clinic. With the increasing emergence of tumour epigenetic markers and antitumor epigenetic drugs, there are also more possibilities for anti-esophageal cancer treatment. This paper focuses on esophageal cancer and epigenetic modifications, with the aim of unravelling the close link between them to facilitate precise and personalized treatment of esophageal cancer.
Collapse
Affiliation(s)
- Wen-Jian Liu
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuan Zhao
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xu Chen
- School of Basic Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Man-Li Miao
- School of Basic Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ren-Quan Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
7
|
Pan L, Liu W, Zhao H, Chen B, Yue X. MiR-191-5p inhibits KLF6 to promote epithelial-mesenchymal transition in breast cancer. Technol Health Care 2023; 31:2251-2265. [PMID: 37545272 DOI: 10.3233/thc-230217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) exert certain functions in the development of several cancers and can be a potential hallmark for cancer diagnosis and prognosis. MiR-191-5p has been proven to have high expression in breast cancer (BC), while its biological role and potential regulatory mechanisms in BC remain an open issue. OBJECTIVE Bioinformatics was utilized to assay miR-191-5p level in BC tissues and predict its downstream target gene as well as the enriched signaling pathways of the target gene. METHODS qRT-PCR was carried out to assay miR-191-5p and KLF6 levels in BC cells as well as miR-191-5p level in blood-derived exosomes from BC patients. Western blot was to examine the expression of proteins linked with cell adhesion, epithelial-mesenchymal transition (EMT), and exosome markers. A dual luciferase reporter assay was utilized to verify the interaction between miR-191-5p and KLF6. Abilities of cell phenotypes of BC cells were detected by CCK8, Transwell, and cell adhesion assay, separately. RESULTS Upregulated miR-191-5p expression and downregulated KLF6 expression were observed in BC cells. There was a targeting relationship between miR-191-5p and KLF6. MiR-191-5p negatively regulated KLF6 to promote EMT and malignant progression of BC cells. Additionally, we described a dramatically high level of miR-191-5p in the blood exosomes of BC patients. CONCLUSION MiR-191-5p advances the EMT of BC by targeting KLF6, indicating that miR-191-5p and KLF6 may be new biomarkers for BC.
Collapse
|
8
|
miR-145-3p Hampers the Malignant Progression of Esophageal Carcinoma via CXCL5 Downregulation. Anal Cell Pathol (Amst) 2022; 2022:5418356. [PMID: 35936390 PMCID: PMC9355783 DOI: 10.1155/2022/5418356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/20/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
Esophageal carcinoma (EC) is the most prevalent malignant tumor that occurs frequently worldwide. The early diagnostic biomarkers are crucial for EC treatment. miRNA can regulate EC progression, with diagnostic and prognostic value. Herein, differentially expressed miRNAs and mRNAs (DEmRNAs) in EC were predicted based on TCGA database. The target mRNAs of miRNA were predicted through databases, which were then intersected with DEmRNAs. Next, the correlation between miRNA and candidate mRNAs was analyzed. qRT-PCR was introduced to analyze expression of miR-145-3p and CXCL5 mRNA in EC cell lines, and western blot was performed to assess protein expression of CXCL5. Cell proliferation, migration, invasion, and apoptosis in EC were examined through CCK-8, wound healing, Transwell invasion, and flow cytometry assays. Moreover, targeting relationship between miR-145-3p and CXCL5 was verified through luciferase reporter gene analysis. The experimental results revealed a decreased miR-145-3p expression and an increased CXCL5 expression in EC. Enforced expression of miR-145-3p hindered proliferation, migration, invasion, and stimulated apoptosis of EC cells by repressing CXCL5. This study manifested that miR-145-3p may be a tumor suppressor in EC, and miR-145-3p/CXCL5 axis restrained the malignant progression of EC. These results supply an underlying target for prognosis and treatment of EC patients.
Collapse
|
9
|
Exosomal miRNA Expression Profiling and the Roles of Exosomal miR-4741, miR-32, miR-3149, and miR-6727 on Gastric Cancer Progression. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1263812. [PMID: 35832845 PMCID: PMC9273446 DOI: 10.1155/2022/1263812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 01/03/2023]
Abstract
Objective Accumulated evidence highlights the biological implications of exosomes in gastric cancer. Herein, we conducted the exosomal miRNA expression profiling and identified potential diagnostic markers for gastric cancer. Methods Plasma exosomes were isolated and identified from three gastric cancer patients and three healthy participants. Microarrays of exosomal miRNAs were then analyzed. Differentially expressed exosomal miRNAs were screened with fold − change|≥2.0 and p ≤ 0.05. Among them, miR-4741, miR-32, miR-3149, and miR-6727 expressions were verified in tissues and plasma of patients and healthy subjects. ROC curves were conducted for evaluating the diagnostic performance. The roles of miR-32, miR-3149, miR-6727, and miR-4741 on gastric cancer progression were observed by cellular experiments. Results Isolated exosomes were well characterized by Western blot and transmission electron microscopy as well as nanoparticle-tracking analyses. According to the microarrays, 142 exosomal miRNAs were upregulated, and 34 were downregulated in gastric cancer than healthy subjects. miR-4741 upregulation and miR-32, miR-3149, and miR-6727 downregulations were found in tissues and plasma of gastric cancer patients. The AUCs of miR-4741, miR-32, miR-3149, and miR-6727 were separately 0.8554, 0.9456, 0.7683, and 0.8923. Upregulated miR-32, miR-3149, and miR-6727 as well as downregulated miR-4741 lowered proliferative, migratory, and invasive capacities as well as elevated apoptotic levels of gastric cancer cells. Conclusion Our study successfully isolated and verified exosomes from plasma of gastric cancer as well as proposed four exosomal miRNAs that could act as promising diagnostic markers and suppress gastric cancer progression.
Collapse
|
10
|
Weidle UH, Nopora A. MicroRNAs and Corresponding Targets in Esophageal Cancer as Shown In Vitro and In Vivo in Preclinical Models. Cancer Genomics Proteomics 2022; 19:113-129. [PMID: 35181582 DOI: 10.21873/cgp.20308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/08/2023] Open
Abstract
Squamous cell carcinoma of the esophagus is associated with a dismal prognosis. Therefore, identification of new targets and implementation of new treatment modalities are issues of paramount importance. Based on a survey of the literature, we identified microRNAs conferring antitumoral activity in preclinical in vivo experiments. In the category of miRs targeting secreted factors and transmembrane receptors, four miRs were up-regulated and 10 were down-regulated compared with five out of nine in the category transcription factors, and six miRs were down-regulated in the category enzymes, including metabolic enzymes. The down-regulated miRs have targets which can be inhibited by small molecules or antibody-related entities, or re-expressed by reconstitution therapy. Up-regulated miRs have targets which can be reconstituted with small molecules or inhibited with antagomirs.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
11
|
Zeng ZL, Zhu Q, Zhao Z, Zu X, Liu J. Magic and mystery of microRNA-32. J Cell Mol Med 2021; 25:8588-8601. [PMID: 34405957 PMCID: PMC8435424 DOI: 10.1111/jcmm.16861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/25/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are a group of endogenous, small (∼22 nts in length) noncoding RNA molecules that function specifically by base pairing with the mRNA of genes and regulate gene expression at the post-transcriptional level. Alterations in miR-32 expression have been found in numerous diseases and shown to play a vital role in cell proliferation, apoptosis, oncogenesis, invasion, metastasis and drug resistance. MiR-32 has been documented as an oncomiR in the majority of related studies but has been also verified as a tumour suppressor miRNA in conflicting reports. Moreover, it has a crucial role in metabolic and cardiovascular disorders. This review provides an in-depth look into the most recent finding regarding miR-32, which is involved in the expression, regulation and functions in different diseases, especially tumours. Additionally, this review outlines novel findings suggesting that miR-32 may be useful as a noninvasive biomarker and as a targeted therapeutic in several diseases.
Collapse
Affiliation(s)
- ZL Zeng
- The First Affiliated HospitalDepartment of Metabolism and EndocrinologyHengyang Medical SchoolUniversity of South ChinaHengyangChina
- The First Affiliated HospitalDepartment of Clinical MedicineHengyang Medical SchoolUniversity of South ChinaHengyangChina
- Key Laboratory for Arteriosclerology of Hunan ProvinceDepartment of Cardiovascular DiseaseHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Qingyun Zhu
- The First Affiliated HospitalDepartment of Metabolism and EndocrinologyHengyang Medical SchoolUniversity of South ChinaHengyangChina
- The First Affiliated HospitalDepartment of Clinical MedicineHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Zhibo Zhao
- The First Affiliated HospitalDepartment of Metabolism and EndocrinologyHengyang Medical SchoolUniversity of South ChinaHengyangChina
- The First Affiliated HospitalDepartment of Clinical MedicineHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Xuyu Zu
- The First Affiliated HospitalDepartment of Metabolism and EndocrinologyHengyang Medical SchoolUniversity of South ChinaHengyangChina
- The First Affiliated HospitalDepartment of Clinical MedicineHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Jianghua Liu
- The First Affiliated HospitalDepartment of Metabolism and EndocrinologyHengyang Medical SchoolUniversity of South ChinaHengyangChina
- The First Affiliated HospitalDepartment of Clinical MedicineHengyang Medical SchoolUniversity of South ChinaHengyangChina
| |
Collapse
|
12
|
Chen X, Wang X, Yi L, Song Y. The KN Motif and Ankyrin Repeat Domains 1/CXXC Finger Protein 5 Axis Regulates Epithelial-Mesenchymal Transformation, Metastasis and Apoptosis of Gastric Cancer via Wnt Signaling. Onco Targets Ther 2020; 13:7343-7352. [PMID: 32801759 PMCID: PMC7395690 DOI: 10.2147/ott.s240991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/12/2020] [Indexed: 12/26/2022] Open
Abstract
Background Emerging research indicates that CXXC finger protein 5 (CXXC5) is involved in the development of various cancers. Besides, KN motif and ankyrin repeat domains 1 (KANK1) was proved as a tumor suppressor in multiple cancers. Our study aimed to illustrate the functional role and mechanism of CXXC5 and KANK1 in gastric cancer (GC) pathogenesis. Methods The tissues of 55 GC patients and six GC cell lines were used to investigate CXXC5 and KANK1 expression using RT-qPCR. Western blot assay was conducted to measure the protein levels of CXXC5, KANK1, epithelial-mesenchymal transformation (EMT) proteins (Vimentin, E-cadherin) and Wnt signaling proteins (β-catenin, Axin2). The correlation between KANK1 and CXXC5 was estimated by Pearson’s correlation analysis. The results of Transwell assays showed the migration and invasion abilities of GC cells, while the apoptosis rate was detected by flow cytometry. Results The expressions of CXXC5 and KANK1 were both decreased in GC tissues and cells, compared with the normal ones (P < 0.01). Overexpressing CXXC5 significantly induced apoptosis (P < 0.05) and inhibited EMT, migration (P < 0.05) and invasion (P < 0.01) in GC cells. Wnt/β-catenin/Axin2 signaling was suppressed by CXXC5 overexpression, and activating Wnt/β-catenin/Axin2 signaling reversed the effects of CXXC5. The expression of KANK1 was found to be positively correlated with CXXC5 (r2 = 0.4024). KANK1 presented similar effects with CXXC5 on GC cells; however, silencing CXXC5 or activating Wnt/β-catenin/Axin2 signaling antagonized the effects of KANK1 overexpression on EMT and apoptosis in GC (P < 0.05). Conclusion Our study suggested that CXXC5 was downregulated in GC and participated in EMT and apoptosis regulations via the Wnt/β-catenin/Axin2 pathway. Besides, the decreased expression of CXXC5 in GC was caused by KANK1 dysregulation.
Collapse
Affiliation(s)
- Xin Chen
- Gastroenteric Medicine and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, Jilin 13000, People's Republic of China
| | - Xiaodong Wang
- Gastroenteric Medicine and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, Jilin 13000, People's Republic of China
| | - Lanjuan Yi
- Department of Gastroenterology, Yantaishan Hospital of Yantai City, Yantai, Shandong 264000, People's Republic of China
| | - Ying Song
- Gastroenteric Medicine and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, Jilin 13000, People's Republic of China
| |
Collapse
|
13
|
Su X, Gao C, Feng X, Jiang M. miR-613 suppresses migration and invasion in esophageal squamous cell carcinoma via the targeting of G6PD. Exp Ther Med 2020; 19:3081-3089. [PMID: 32256796 PMCID: PMC7086187 DOI: 10.3892/etm.2020.8540] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 01/17/2020] [Indexed: 12/20/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common cancer in China and has a high mortality rate. MicroRNAs (miRs) are a family of post-transcriptional regulators, which negatively regulate target gene expression. miR-613 has been revealed to be a diagnostic and prognostic biomarker in ESCC. However, the role of miR-613 in ESCC remains unclear. In the present study, miR-613 expression was identified to be reduced in tumor tissues in comparison with corresponding adjacent normal tissues. TargetScan and a dual-luciferase reporter assay verified glucose-6-phosphate dehydrogenase (G6PD) as a direct target of miR-613. In contrast with miR-613, G6PD expression was increased in tumor tissues compared with matched healthy tissues. Furthermore, overexpression of miR-613 inhibited cell migration and invasion of Eca109 cells compared with controls, while G6PD overexpression reversed the inhibition induced by miR-613, as determined by wound healing and Transwell assays. In addition, miR-613 overexpression decreased the mRNA and protein expression of G6PD, matrix metalloproteinase (MMP)2 and MMP9, and reduced the phosphorylation of signal transducer and activator of transcription 3 (STAT3) compared with controls, while G6PD reversed the effects of miR-613. However, miR-613 and G6PD did not affect the expression of STAT3. In conclusion, the aforementioned results suggest that miR-613 targets G6PD to suppress ESCC cell migration and invasion through reduced MMP2 and MMP9 expression and inactivation of the STAT3 signaling pathway. Thus, the present study may provide a new molecular foundation for treatment of ESCC.
Collapse
Affiliation(s)
- Xiangyu Su
- Department of Oncology, Zhongda Hospital, The Affiliated Hospital of Southeast University, Nanjing, Jiangsu 210009, P.R. China
- Clinical Medicine, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Chanchan Gao
- Department of Oncology, Zhongda Hospital, The Affiliated Hospital of Southeast University, Nanjing, Jiangsu 210009, P.R. China
- Clinical Medicine, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiaoyao Feng
- Department of Radiation Oncology, General Hospital of Eastern Theater Command, Nanjing, Jiangsu 210002, P.R. China
| | - Ming Jiang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
14
|
Gu JF, Liu SG, Pan Q, Qin F, Li YY. Negative regulation of CDK6 expression by microRNA-126-5p and its influence on the proliferation and invasion of esophageal cancer cells. Anat Rec (Hoboken) 2020; 303:2811-2820. [PMID: 31916689 DOI: 10.1002/ar.24362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/10/2019] [Accepted: 11/24/2019] [Indexed: 12/18/2022]
Abstract
The present study aimed to investigate the expression of cyclin-dependent kinase 6 (CDK6) and microRNA-126-5p (miR-126-5p) in esophageal cancer tissues and cells, and their effect on esophageal cancer cell proliferation and invasion, and to explore the potential molecular mechanisms. The relative expression levels of CDK6 and miR-126-5p in esophageal cancer tissue, paracancerous tissue, and HEEC and EC109 cells were determined and compared using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). A miR-126-5p overexpression vector was constructed and a stable EC109 cell line expressing miR-126-5p was established. The EC109 cell line was transfected with a CDK6 small interfering RNA sequence. The rate of cell proliferation was determined using the WST-8 method, and cell invasion was determined using a Transwell assay. In addition, the relative expression levels of genes were determined using RT-qPCR; the relative expression levels of proteins were determined by western blot analysis; the binding sites of CDK6 and miR-126-5p were analyzed using TargetScan software; and the interaction of CDK6 and miR-126-5p was verified using dual-fluorescence reporter gene expression. Esophageal tissues and EC109 cells expressed higher levels of CDK6 but significantly lower levels of miR-126-5p compared with adjacent tissues and HEEC cells, and their correlation coefficient between esophageal tissues and matched adjacent tissues was -7.526. The overexpression of miR-126-5p and CDK6 knockdown in the EC109 cell line inhibited cell proliferation and invasion compared with the control and NC (negative control) groups. miR-126-5p overexpression reduced the relative expression level of CDK6, and CDK6 knockdown by siRNA increased the expression of miR-126-5p. miR-126-5p regulated CDK6 expression by binding to the 3'-untranslated region of its mRNA. Overexpression miR-126-5p inhibited the proliferation and migration of esophageal cancer cells by targeting CDK6 and negatively regulating its expression. These findings contribute to the understanding of the underlying molecular mechanism of esophageal cancer.
Collapse
Affiliation(s)
- Jian-Fa Gu
- Department of Oncology, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Song-Ge Liu
- Department of Oncology, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Qiong Pan
- Department of Oncology, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Fengying Qin
- Department of Oncology, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Yan-Yan Li
- Department of Oncology, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
15
|
Wang D, Zeng T, Lin Z, Yan L, Wang F, Tang L, Wang L, Tang D, Chen P, Yang M. Long non-coding RNA SNHG5 regulates chemotherapy resistance through the miR-32/DNAJB9 axis in acute myeloid leukemia. Biomed Pharmacother 2019; 123:109802. [PMID: 31884339 DOI: 10.1016/j.biopha.2019.109802] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/10/2019] [Accepted: 12/15/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a common hematopoietic malignancy with invasive activity. Drug resistance greatly contributes to the poor efficacy of chemotherapy in AML treatment. Recent research indicates that long non-coding RNAs (LncRNAs) regulates chemotherapy resistance in malignancy. METHODS Microarray analysis was used to screen out AML related genes, and interaction between small nucleolar RNA host gene 5(SNHG5) and miR-32, as well as that between miR-32 and DNAJB9. Quantitative real-time PCR (qRT-PCR) and In situ hybridization(ISH) were used to determine the expression levels of SNHG5, miR-32 and DNAJB9 mRNA in AML cell lines and clinic samples. Western blot was performed to detect protein expression levels. After being treated with varying concentrations of Adriamycin(ADM), cell viability was evaluated using a cell counting kit-8(CCK8). RESULTS We carried out a genome-wide LncRNA expression study and found SNHG5 aberrantly overexpressed in AML comparing to the donors. Knock-down of SNHG5 promoted sensitivity of AML cells to chemotherapy. In addition, miR-32 was identified as the downstream target of SNHG5 and miR-32 inhibitor abrogated the inhibiting effects of downregulated SNHG5 on AML cell viability. Furthermore, inhibited SNHG5 decreased DNAJB9 expression levels by sponging miR-32. The SNHG5/miR-32/DNAJB9 axis targeted autophagy to regulate chemotherapy resistance. CONCLUSION SHNG5 regulates chemotherapy resistance by targeting the miR-32/DNAJB9 axis in acute myeloid leukemia, which provided a novel potential target for AML and revealed an important mechanism of chemotherapy resistance.
Collapse
Affiliation(s)
- Dan Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ting Zeng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhi Lin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lu Yan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Fenglin Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lanlan Tang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Leyuan Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pan Chen
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China.
| | - Minghua Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
16
|
Luo J, Chen XQ, Li P. The Role of TGF-β and Its Receptors in Gastrointestinal Cancers. Transl Oncol 2019; 12:475-484. [PMID: 30594036 PMCID: PMC6314240 DOI: 10.1016/j.tranon.2018.11.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Early detection of gastrointestinal tumors improves patient survival. However, patients with these tumors are typically diagnosed at an advanced stage and have poor prognosis. The incidence and mortality of gastrointestinal cancers, including esophageal, gastric, liver, colorectal, and pancreatic cancers, are increasing worldwide. Novel diagnostic and therapeutic agents are required to improve patient survival and quality of life. The tumor microenvironment, which contains nontumor cells, signaling molecules such as growth factors and cytokines, and extracellular matrix proteins, plays a critical role in cancer cell proliferation, invasion, and metastasis. Transforming growth factor beta (TGF-β) signaling has dual roles in gastrointestinal tumor development and progression as both a tumor suppressor and tumor promoter. Here, we review the dynamic roles of TGF-β and its receptors in gastrointestinal tumors and provide evidence that targeting TGF-β signaling may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Jingwen Luo
- Oncology Department, West China Hospital of Medicine, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Xu-Qiao Chen
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ping Li
- Oncology Department, West China Hospital of Medicine, Sichuan University, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|