1
|
Li S, Zhang Z, Li Z, Yang L, Liu J, Liu Y, Liu Y, Hou Y, Mei M, Huang Y. CENPA promotes glutamine metabolism and tumor progression by up-regulating SLC38A1 in endometrial cancer. Cell Signal 2024; 117:111110. [PMID: 38382691 DOI: 10.1016/j.cellsig.2024.111110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/28/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
Glutamine addiction is a significant hallmark of metabolic reprogramming in tumors and is crucial to the progression of cancer. Nevertheless, the regulatory mechanisms of glutamine metabolism in endometrial cancer (EC) remains elusive. In this research, we found that elevated expression of CENPA and solute carrier family 38 member 1 (SLC38A1) were firmly associated with worse clinical stage and unfavorable outcomes in EC patients. In addition, ectopic overexpression or silencing of CENPA could either enhance or diminish glutamine metabolism and tumor progression in EC. Mechanistically, CENPA directly regulated the transcriptional activity of the target gene, SLC38A1, leading to enhanced glutamine uptake and metabolism, thereby promoting EC progression. Notably, a prognostic model utilizing the expression levels of CENPA and SLC38A1 genes independently emerged as a prognostic factor for EC. More importantly, CENPA and SLC38A1 were significantly elevated and positively correlated, as well as indicative of poor prognosis in multiple cancers. In brief, our study confirmed that CENPA is a critical transcription factor involved in glutamine metabolism and tumor progression through modulating SLC38A1. This revelation suggests that targeting CENPA could be an appealing therapeutic approach to address pan-cancer glutamine addiction.
Collapse
Affiliation(s)
- Shuang Li
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China
| | - Zihui Zhang
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China
| | - Zhifang Li
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China
| | - Lian Yang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jianfeng Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yujie Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yancai Liu
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China
| | - Yanmei Hou
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China
| | - Mei Mei
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China.
| | - Yuqin Huang
- Department of Gynecology and Obstetrics, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei Province, China.
| |
Collapse
|
2
|
Liu HT, Zhao Y, Wang HC, Liu QL. METTL3-mediated m 6A methylation of SLC38A1 stimulates cervical cancer growth. Biochem Biophys Res Commun 2024; 716:150039. [PMID: 38701556 DOI: 10.1016/j.bbrc.2024.150039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/14/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
The objective of this study was to better characterize the role of the glutamine transporter SLC38A1 in cervical cancer and explore the underlying mechanisms. Data from public databases and clinical cervical cancer tissue samples were used to assess the expression of SLC38A1 and its prognostic significance. Immunohistochemical staining, qRT-PCR, and Western blotting were used to evaluate the expression of relevant genes and proteins. Cell viability, cell cycle, apoptosis, and intracellular glutamine content were measured using CCK-8, flow cytometry, and biochemical assays. Additionally, the RNA immunoprecipitation (RIP) assay was used to examine the impact of METTL3/IGF2BP3 on the m6A modification of the SLC38A1 3'UTR. Both cervical cancer specimens and cells showed significantly increased expression of SLC38A1 and its expression correlated with an unfavorable prognosis. Knockdown of SLC38A1 inhibited cell viability and cell cycle progression, induced apoptosis, and suppressed tumor growth in vivo. Glutaminase-1 inhibitor CB-839 reversed the effects of SLC38A1 overexpression. METTL3 promoted m6A modification of SLC38A1 and enhanced its mRNA stability through IGF2BP3 recruitment. Moreover, METTL3 silencing inhibited cell viability, cell cycle progression, intracellular glutamine content, and induced apoptosis, but these effects were reversed by SLC38A1 overexpression. In conclusion, METTL3-mediated m6A methylation of SLC38A1 stimulates cervical cancer progression. SLC38A1 inhibition is a potential therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Hai-Ting Liu
- Department of Medical Genetics, Zibo Maternal and Child Health Hospital, Zibo, 255000, China.
| | - Yun Zhao
- Department of Gynecology, Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Hong-Cai Wang
- Department of Gynecology, Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| | - Qing-Ling Liu
- Department of Clinical Laboratory, Zibo Maternal and Child Health Hospital, Zibo, 255000, China
| |
Collapse
|
3
|
Wang Y, Li H, Chen W, Huang X, Fan R, Xu M, Zou L. CircRUNX1 drives the malignant phenotypes of lung adenocarcinoma through mediating the miR-5195-3p/HMGB3 network. Gen Thorac Cardiovasc Surg 2024; 72:164-175. [PMID: 37474742 DOI: 10.1007/s11748-023-01960-5] [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: 01/06/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs) are key factors in the regulation of cancer progression. However, the role of circRUNX1 in lung adenocarcinoma (LUAD) progression is unclear. METHODS The expression levels of circRUNX1, microRNA (miR)-5195-3p, and high-mobility group protein B3 (HMGB3) were detected by quantitative real-time PCR. Cell proliferation, migration, invasion and apoptosis were analyzed by EdU staining, colony formation assay, transwell assay and flow cytometry. Protein levels were measured using western blot analysis. The interaction between miR-5195-3p and circRUNX1 or HMGB3 was verified by dual-luciferase reporter assay and RIP assay. Animal experiments were performed to investigate the role of circRUNX1 in LUAD tumorigenesis. RESULTS We found that circRUNX1 was upregulated in LUAD tumor tissues and cells. CircRUNX1 knockdown suppressed LUAD cell proliferation and metastasis, while promoted apoptosis. In terms of mechanism, we found that circRUNX1 could sponge miR-5195-3p, and miR-5195-3p inhibitor abolished the regulation of circRUNX1 knockdown on LUAD cell proliferation, metastasis and apoptosis. In addition, miR-5195-3p could target HMGB3, and HMGB3 overexpression reversed the inhibition effect of miR-5195-3p on LUAD progression. Moreover, circRUNX1 knockdown reduced LUAD tumorigenesis. CONCLUSION CircRUNX1 facilitated LUAD proliferation and metastasis by regulating the miR-5195-3p/HMGB3 axis, suggesting that it might be a possible therapeutic target for LUAD.
Collapse
Affiliation(s)
- Yan Wang
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317, Zhongshan Road, Yanping District, Nanping City, 353000, Fujian Province, People's Republic of China
| | - Hui Li
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317, Zhongshan Road, Yanping District, Nanping City, 353000, Fujian Province, People's Republic of China
| | - Wenbin Chen
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317, Zhongshan Road, Yanping District, Nanping City, 353000, Fujian Province, People's Republic of China
| | - Xiaoliang Huang
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317, Zhongshan Road, Yanping District, Nanping City, 353000, Fujian Province, People's Republic of China
| | - Renliang Fan
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317, Zhongshan Road, Yanping District, Nanping City, 353000, Fujian Province, People's Republic of China
| | - Meijia Xu
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317, Zhongshan Road, Yanping District, Nanping City, 353000, Fujian Province, People's Republic of China
| | - Li Zou
- Department of Oncology, Nanping First Hospital Affiliated to Fujian Medical University, No. 317, Zhongshan Road, Yanping District, Nanping City, 353000, Fujian Province, People's Republic of China.
| |
Collapse
|
4
|
Ahram M, Abu Alragheb B, Abushukair H, Bawadi R, Al-Hussaini M. MicroRNAs Associated with Androgen Receptor and Metastasis in Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:665. [PMID: 38339416 PMCID: PMC10854913 DOI: 10.3390/cancers16030665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
It is crucial to identify novel molecular biomarkers and therapeutic targets for triple-negative breast cancer (TNBC). The androgen receptor (AR) is a regulator of TNBC, acting partially via microRNA molecules (miRNAs). In this study, we used PCR arrays to profile the expression of 84 miRNAs in 24 TNBC tissue samples, which were equally classified according to AR expression and/or metastasis. Several bioinformatics tools were then utilized to determine the potentially affected protein targets and signaling pathways. Seven miRNAs were found to be significantly more highly expressed in association with AR expression, including miR-328-3p and miR-489-3p. Increased expression of miR-205-3p was found to be significantly associated with metastasis. Certain miRNAs were specifically found to be differentially expressed in either metastatic or non-metastatic AR-positive tumors. A gene ontology (GO) analysis indicated biological roles in the regulation of transcription, cellular response to DNA damage, and the transforming growth factor-beta (TGF-beta) signaling pathway. The GO analysis also showed enrichment in kinase and transcription factor activities. The TGF-beta and a number of kinase-dependent pathways were also retrieved using the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. This study offers an understanding of the role of AR in TNBC and further implicates miRNAs in mediating the effects of AR on TNBC.
Collapse
Affiliation(s)
- Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | | | - Hassan Abushukair
- School of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Randa Bawadi
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Maysa Al-Hussaini
- Department of Pathology and Laboratory Medicine, King Hussein Cancer Center, Amman 11941, Jordan;
| |
Collapse
|
5
|
Feng HG, Wu CX, Zhong GC, Gong JP, Miao CM, Xiong B. Integrative analysis reveals that SLC38A1 promotes hepatocellular carcinoma development via PI3K/AKT/mTOR signaling via glutamine mediated energy metabolism. J Cancer Res Clin Oncol 2023; 149:15879-15898. [PMID: 37673823 DOI: 10.1007/s00432-023-05360-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023]
Abstract
Although hepatocellular carcinoma (HCC) is rather frequent, little is known about the molecular pathways underlying its development, progression, and prognosis. In the current study, we comprehensively analyzed the deferentially expressed metabolism-related genes (MRGs) in HCC based on TCGA datasets attempting to discover the potentially prognostic genes in HCC. The up-regulated MRGs were further subjected to analyze their prognostic values and protein expressions. Twenty-seven genes were identified because their high expressions were significant in OS, PFS, DFS, DSS, and HCC tumor samples. They were then used for GO, KEGG, methylation, genetics changes, immune infiltration analyses. Moreover, we established a prognostic model in HCC using univariate assays and LASSO regression based on these MRGs. Additionally, we also found that SLC38A1, an amino acid metabolism closely related transporter, was a potential prognostic gene in HCC, and its function in HCC was further studied using experiments. We found that the knockdown of SLC38A1 notably suppressed the growth and migration of HCC cells. Further studies revealed that SLC38A1 modulated the development of HCC cells by regulating PI3K/AKT/mTOR signaling via glutamine mediated energy metabolism. In conclusion, this study identified the potentially prognostic MRGs in HCC and uncovered that SLC38A1 regulated HCC development and progression by regulating PI3K/AKT/mTOR signaling via glutamine mediated energy metabolism, which might provide a novel marker and potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Hua-Guo Feng
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, No. 74 Linjiang Road, Chongqing, China
| | - Chuan-Xin Wu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, No. 74 Linjiang Road, Chongqing, China
| | - Guo-Chao Zhong
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, No. 74 Linjiang Road, Chongqing, China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, No. 74 Linjiang Road, Chongqing, China
| | - Chun-Mu Miao
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, No. 74 Linjiang Road, Chongqing, China
| | - Bin Xiong
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, No. 74 Linjiang Road, Chongqing, China.
| |
Collapse
|
6
|
Xiong B, Huang Q, Zheng H, Lin S, Xu J. Recent advances microRNAs and metabolic reprogramming in colorectal cancer research. Front Oncol 2023; 13:1165862. [PMID: 37576895 PMCID: PMC10415904 DOI: 10.3389/fonc.2023.1165862] [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: 02/16/2023] [Accepted: 06/07/2023] [Indexed: 08/15/2023] Open
Abstract
Colorectal cancer (CRC) is a cancer with the highest incidence and mortality. Alteration of gene expression is the main pathophysiological mechanism of CRC, which results in disturbed signaling pathways and cellular metabolic processes. MicroRNAs are involved in almost all pathophysiological processes and are correlative with colorectal cancer metabolism, proliferation, and chemotherapy resistance. Metabolic reprogramming, an important feature of cancer, is strongly correlative with the development and prognosis of cancers, including colorectal cancer. MicroRNAs can target enzymes involved in metabolic processes, thus playing a regulatory role in tumor metabolism. The disorder of the signaling pathway is another characteristic of tumor, which induces the occurrence and proliferation of tumors, and is closely correlative with the prognosis and chemotherapy resistance of tumor patients. MicroRNAs can target the components of the signaling pathways to regulate their transduction. Understanding the function of microRNAs in the occurrence and proliferation of CRC provides novel insights into the optimal treatment strategies, prognosis, and development of diagnosis in CRC. This article reviews the relationship between CRC and microRNA expression and hopes to provide new options for the diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Bin Xiong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Qiaoyi Huang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Huida Zheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Jianhua Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
7
|
Wu R, Tang S, Wang Q, Kong P, Liu F. Hsa_circ_0003602 Contributes to the Progression of Colorectal Cancer by Mediating the miR-149-5p/SLC38A1 Axis. Gut Liver 2023; 17:267-279. [PMID: 36148577 PMCID: PMC10018293 DOI: 10.5009/gnl210542] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 11/04/2022] Open
Abstract
Background/Aims We aimed to investigate the role and working mechanism of Homo sapiens circular RNA_0003602 (hsa_circ_0003602) in colorectal cancer (CRC) development. Methods The expression of circ_0003602, miR-149-5p, and solute carrier family 38 member 1 (SLC38A1) was detected by quantitative real-time polymerase chain reaction. RNase R assays were conducted to determine the characteristics of circ_0003602. CCK-8 assays, flow cytometry analysis, transwell invasion assays, wound healing assays and tube formation assays were employed to evaluate cell viability, apoptosis, invasion, migration, and angiogenesis. All protein levels were examined by Western blot or immunohistochemistry assay. The glutamine metabolism was monitored by corresponding glutamine, α-ketoglutarate and glutamate assay kits. Dual-luciferase reporter assay was utilized to confirm the targeted combination between miR-149-5p and circ_0003602 or SLC38A1. A xenograft tumor model was established to analyze the role of circ_0003602 in CRC tumor growth in vivo. Results Circ_0003602 was upregulated in CRC tissues and cell lines. Circ_0003602 silencing suppressed CRC cell viability, migration, invasion, angiogenesis, and glutaminolysis; induced cell apoptosis in vitro; and blocked tumor growth in vivo. Moreover, circ_0003602 directly interacted with miR-149-5p to negatively regulate its expression, and circ_0003602 knockdown suppressed the malignant behaviors of CRC cells largely by upregulating miR-149-5p. MiR-149-5p directly bound to the 3' untranslated region of SLC38A1 to induce its degradation, and miR-149-5p overexpression reduced the malignant potential of CRC cells largely by downregulating SLC38A1. Circ_0003602 positively regulated SLC38A1 expression by sponging miR-149-5p in CRC cells. Conclusions Circ_0003602 knockdown impedes CRC development by targeting the miR-149-5p/SLC38A1 axis, which provides a novel theoretical basis and new insights for CRC treatment.
Collapse
Affiliation(s)
- Rong Wu
- Clinical Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyu Tang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Qiuxiao Wang
- Department of Clinical Medicine of Combination of Chinese and Western Medicine, North Sichuan Medical College, Nanchong, China
| | - Pengfei Kong
- Division of Anorectal, Department of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Fang Liu
- Department of Clinical Medicine of Combination of Chinese and Western Medicine, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
8
|
Geng Y, Wang M, Wu Z, Jia J, Yang T, Yu L. Research progress of circRNA in malignant tumour metabolic reprogramming. RNA Biol 2023; 20:641-651. [PMID: 37599427 PMCID: PMC10443989 DOI: 10.1080/15476286.2023.2247877] [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] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Cancer is a multi-factor systemic malignant disease, which has seriously threatened human health and created a heavy burden on the world economy. Metabolic reprogramming, one of the important signs of malignant tumours, provides necessary nutrition for tumorigenesis and cancer development; thus, it has recently become a research hot spot, even though the metabolic mechanism is quite intricate. Circular RNA (circRNA) affects cancer cell metabolism through various molecular mechanisms, playing an important role in promoting or suppressing cancer. Because of the structure characteristics, circRNA is quite stable, and can be utilized as biomarkers. In this review, we analysed and summarized the characteristics and biological functions of circRNA and comprehensively reviewed and discussed the important role of circRNA in cancer metabolic reprogramming. This review will provide new ideas for developing new anti-cancer therapeutic targets, mining cancer diagnostic and prognostic markers, and will provide guidance for other researchers to design circRNA-related experiments and develop anti-tumour drugs.
Collapse
Affiliation(s)
- Yikun Geng
- Graduate school, Inner Mongolia Medical University, Hohhot, China
- Clinical Medical Research Center, Inner Mongolian People’s Hospital, Hohhot, China
| | - Min Wang
- Clinical Medical Research Center, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Key Laboratory of Gene Regulation of The Metabolic Disease, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People’s Hospital, Hohhot, China
| | - Zhouying Wu
- Clinical Medical Research Center, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Key Laboratory of Gene Regulation of The Metabolic Disease, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People’s Hospital, Hohhot, China
| | - Jianchao Jia
- Clinical Medical Research Center, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Key Laboratory of Gene Regulation of The Metabolic Disease, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People’s Hospital, Hohhot, China
| | - Tingyu Yang
- Clinical Medical Research Center, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Key Laboratory of Gene Regulation of The Metabolic Disease, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People’s Hospital, Hohhot, China
| | - Lan Yu
- Clinical Medical Research Center, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Key Laboratory of Gene Regulation of The Metabolic Disease, Inner Mongolian People’s Hospital, Hohhot, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People’s Hospital, Hohhot, China
| |
Collapse
|
9
|
Ni R, Li Z, Li L, Peng D, Ming Y, Li L, Liu Y. Rethinking glutamine metabolism and the regulation of glutamine addiction by oncogenes in cancer. Front Oncol 2023; 13:1143798. [PMID: 36959802 PMCID: PMC10029103 DOI: 10.3389/fonc.2023.1143798] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
Glutamine, the most abundant non-essential amino acid in human blood, is crucial for cancer cell growth and cancer progression. Glutamine mainly functions as a carbon and nitrogen source for biosynthesis, energy metabolism, and redox homeostasis maintenance in cancer cells. Dysregulated glutamine metabolism is a notable metabolic characteristic of cancer cells. Some carcinogen-driven cancers exhibit a marked dependence on glutamine, also known as glutamine addiction, which has rendered the glutamine metabolic pathway a breakpoint in cancer therapeutics. However, some cancer cells can adapt to the glutamine unavailability by reprogramming metabolism, thus limiting the success of this therapeutic approach. Given the complexity of metabolic networks and the limited impact of inhibiting glutamine metabolism alone, the combination of glutamine metabolism inhibition and other therapeutic methods may outperform corresponding monotherapies in the treatment of cancers. This review summarizes the uptake, transport, and metabolic characteristics of glutamine, as well as the regulation of glutamine dependence by some important oncogenes in various cancers to emphasize the therapeutic potential of targeting glutamine metabolism. Furthermore, we discuss a glutamine metabolic pathway, the glutaminase II pathway, that has been substantially overlooked. Finally, we discuss the applicability of polytherapeutic strategies targeting glutamine metabolism to provide a new perspective on cancer therapeutics.
Collapse
Affiliation(s)
- Rui Ni
- Department of pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Ziwei Li
- Department of pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Li Li
- Department of pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Dan Peng
- Department of pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yue Ming
- Department of pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin Li
- Department of pharmacy, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
- *Correspondence: Lin Li, ; Yao Liu,
| | - Yao Liu
- Department of pharmacy, Daping Hospital, Army Medical University, Chongqing, China
- *Correspondence: Lin Li, ; Yao Liu,
| |
Collapse
|
10
|
Wang C, Zhou M, Zhu P, Ju C, Sheng J, Du D, Wan J, Yin H, Xing Y, Li H, He J, He F. IGF2BP2-induced circRUNX1 facilitates the growth and metastasis of esophageal squamous cell carcinoma through miR-449b-5p/FOXP3 axis. J Exp Clin Cancer Res 2022; 41:347. [PMID: 36522683 PMCID: PMC9753396 DOI: 10.1186/s13046-022-02550-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common digestive malignancies with relatively high morbidity and mortality. Emerging evidence suggests circular RNAs (circRNAs) play critical roles in tumor cell malignancy. However, the biological function and clinical significance of many circRNAs in ESCC remain elusive. METHODS The expression level and clinical implication of circRUNX1 in ESCC tissues were evaluated using qRT-PCR. In vitro and in vivo functional studies were conducted to investigate the underlying biological effects of circRUNX1 on ESCC cell growth and metastasis. Moreover, bioinformatics analysis, RNA sequencing (RNA-seq), RNA immunoprecipitation (RIP) assays, dual-luciferase reporter assays, and rescue experiments were performed to explore the relationships between circRUNX1, miR-449b-5p, Forkhead box protein P3 (FOXP3), and insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2). RESULTS CircRUNX1 was found to be significantly up-regulated in ESCC tissues and associated with TNM stage and differentiation grade. Functionally, circRUNX1 promoted ESCC cell proliferation and metastasis in vitro and in vivo. CircRUNX1 enhanced FOXP3 expression by competitively sponging miR-449b-5p. Notably, both miR-449b-5p mimics and FOXP3 knockdown restored the effects of circRUNX1 overexpression on cell proliferation and metastasis. Furthermore, IGF2BP2 binding to circRUNX1 prevented its degradation. CONCLUSIONS IGF2BP2 mediated circRUNX1 functions as an oncogenic factor to facilitate ESCC progression through the miR-449b-5p/FOXP3 axis, implying that circRUNX1 has the potential to be a promising diagnostic marker and therapeutic target for ESCC patients.
Collapse
Affiliation(s)
- Chang Wang
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mingxia Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Peiyu Zhu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Chenxi Ju
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jinxiu Sheng
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dan Du
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junhu Wan
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Huiqing Yin
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yurong Xing
- Center of Health Examination, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongle Li
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China.
| | - Jing He
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Fucheng He
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
11
|
Yi C, Yu AM. MicroRNAs in the Regulation of Solute Carrier Proteins Behind Xenobiotic and Nutrient Transport in Cells. Front Mol Biosci 2022; 9:893846. [PMID: 35755805 PMCID: PMC9220936 DOI: 10.3389/fmolb.2022.893846] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
Altered metabolism, such as aerobic glycolysis or the Warburg effect, has been recognized as characteristics of tumor cells for almost a century. Since then, there is accumulating evidence to demonstrate the metabolic reprogramming of tumor cells, addiction to excessive uptake and metabolism of key nutrients, to support rapid proliferation and invasion under tumor microenvironment. The solute carrier (SLC) superfamily transporters are responsible for influx or efflux of a wide variety of xenobiotic and metabolites that are needed for the cells to function, as well as some medications. To meet the increased demand for nutrients and energy, SLC transporters are frequently dysregulated in cancer cells. The SLCs responsible for the transport of key nutrients for cancer metabolism and energetics, such as glucose and amino acids, are of particular interest for their roles in tumor progression and metastasis. Meanwhile, rewired metabolism is accompanied by the dysregulation of microRNAs (miRNAs or miRs) that are small, noncoding RNAs governing posttranscriptional gene regulation. Studies have shown that many miRNAs directly regulate the expression of specific SLC transporters in normal or diseased cells. Changes of SLC transporter expression and function can subsequently alter the uptake of nutrients or therapeutics. Given the important role for miRNAs in regulating disease progression, there is growing interest in developing miRNA-based therapies, beyond serving as potential diagnostic or prognostic biomarkers. In this article, we discuss how miRNAs regulate the expression of SLC transporters and highlight potential influence on the supply of essential nutrients for cell metabolism and drug exposure toward desired efficacy.
Collapse
Affiliation(s)
- Colleen Yi
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
12
|
Circ_0001093 promotes glutamine metabolism and cancer progression of esophageal squamous cell carcinoma by targeting miR-579-3p/glutaminase axis. J Bioenerg Biomembr 2022; 54:119-134. [PMID: 35322289 DOI: 10.1007/s10863-022-09935-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/01/2022] [Indexed: 12/23/2022]
Abstract
Increasing studies indicate that circular RNAs (circRNAs) play critical roles in tumor metabolism of multiple cancers. However, the contribution of circRNAs in glutamine metabolism of esophageal squamous cell carcinoma (ESCC) remains elusive. The objective of this research was to investigate the role and mechanism of circRNA hsa_circ_0001093 (circ_0001093) in the glutamine metabolism and tumorigenesis of ESCC. Circ_0001093, microRNA-579-3p (miR-579-3p) and glutaminase (GLS) expressions in ESCC tissues and cell lines were measured by qRT-PCR, tissue array or Western blot. Cell proliferation, invasion and migration were assessed by CCK-8 or transwell assays. Glutamine consumption, glutamate and ATP production were detected by indicated assay kits. The relationships between circ_0001093 and miR-579-3p or GLS mRNA were investigated by bioinformatics analysis, RNA pull-down, luciferase reporter and RNA immunoprecipitation (RIP) assays. Here, we found that circ_0001093 expression was up-regulated in ESCC tissues and cell lines. Increased circ_0001093 expression predicted an unfavourable prognosis, and was associated with the lymph node metastasis, TNM staging and tumor size in ESCC tissues. Circ_0001093 knockdown suppressed cell proliferation, invasion, migration and glutamine metabolism of ESCC cells, while circ_0001093 over-expression showed the opposite effects. Mechanistically, circ_0001093 acted as a competing endogenous RNA (ceRNA) by sponging miR-579-3p, thereby increasing GLS expression. Furthermore, the inhibitory effects of circ_0001093 knockdown on the invasion, migration and glutamine metabolism were partly rescued by miR-579-3p inhibition or GLS over-expression in ESCC cells. Additionally, miR-579-3p expression was down-regulated in ESCC tissues, while GLS expression was up-regulated. In conclusion, this study first provides evidence that the circ_0001093/miR-579-3p/GLS regulatory network can affect glutamine metabolism and malignant phenotype of ESCC, which can further impact ESCC progression.
Collapse
|
13
|
Zhang M, Wang S. Roles of circular RNAs in colorectal cancer. Oncol Lett 2021; 22:602. [PMID: 34188704 PMCID: PMC8227629 DOI: 10.3892/ol.2021.12863] [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: 01/25/2021] [Accepted: 05/12/2021] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of malignant cancer worldwide and poses a significant burden on both the individual and healthcare systems. Despite advances in treatment options, advanced-stage CRC has a high mortality rate due to its heterogeneity, metastatic potential and/or delay in diagnosis. In recent years, an increasing number of studies have indicated that circular RNAs (circRNAs) serve important roles in several types of cancer, including CRC. Recent studies have revealed that circRNAs are aberrantly expressed in CRC tissues and function as oncogenic or tumor suppressive regulators of CRC carcinogenesis and development. Numerous circRNAs have been associated with the clinicopathological features of patients with CRC and have been considered as potential biomarkers for the diagnosis and prognosis of CRC, as well as targets for treatment. However, a deeper understanding of their potential function is required. In the present review, the current body of knowledge on the biogenesis and functions of CRC-associated circRNAs, and their potential value in clinical applications, such as in CRC diagnosis, prognosis and treatment, is discussed and summarized.
Collapse
Affiliation(s)
- Mingying Zhang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
- Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Shubin Wang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute of Shenzhen-PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
- Correspondence to: Professor Shubin Wang, Department of Oncology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian, Shenzhen, Guangdong 518036, P.R. China, E-mail:
| |
Collapse
|