1
|
Zhang YH, Liu XS, Gao Y, Yuan LL, Huang ZM, Zhang Y, Liu ZY, Yang Y, Liu XY, Ke CB, Pei ZJ. SFXN1 as a potential diagnostic and prognostic biomarker of LUAD is associated with 18F-FDG metabolic parameters. Lung Cancer 2024; 188:107449. [PMID: 38184958 DOI: 10.1016/j.lungcan.2023.107449] [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: 10/28/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Sideroflexin 1 (SFXN1) has been discovered as a novel tumor marker for lung adenocarcinoma, but data on its importance in the development of lung adenocarcinoma is still limited. This study evaluated the correlation between SFXN1 and parameters related to 18F-flurodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT), and further explored the role of SFXN1 in the value-added and glycolytic processes of LUAD. METHOD The expression and prognostic value of SFXN1 mRNA in LUAD were analyzed using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) data base. Retrospective analysis of 18F-FDG PET imaging and metabolic parameters in 42 patients to explore the relationship between the expression of SFXN1 and glucose metabolism levels in lung adenocarcinoma and its clinical significance. H1975 cells were selected as the in vitro research object, and the biological effects of SFXN1 on LUAD were further elucidated through Edu proliferation assay, CCK8 activity assay, wound healing experiment, and cell flow cytometry. RESULT SFXN1 is highly expressed in various tumors, including LUAD, and its high expression can serve as an independent predictor of overall survival in lung adenocarcinoma. In addition, the expression of SFXN1 in LUAD was significantly correlated with 18F-FDG PET/CT parameters: maximum and average standardized uptake values (SUVmax and SUVmean), as well as total lesion glycolysis (TLG) (rho = 0.574, 0.589, and 0.338, p < 0.05), which can predict the expression of SFXN1 with an accuracy of 0.934. In vitro functional experiments have shown that knocking down SFXN1 inhibits the proliferation and migration of LUAD cells, promotes cell apoptosis, and may inhibit tumor activity by regulating the expression of glycolytic related genes SLC2A1, HK2, GPI, ALDOA, GAPDH, ENO1, PKM, and LDHA. CONCLUSION The overexpression of SFXN1 is closely related to FDG uptake, and SFXN1, as a promising prognostic biomarker, may mediate the development of LUAD through the glycolytic pathway.
Collapse
Affiliation(s)
- Yao-Hua Zhang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xu-Sheng Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan 442000, Hubei, China
| | - Yan Gao
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Ling-Ling Yuan
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Zhong-Min Huang
- Department of Medical Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yu Zhang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Zi-Yue Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yi Yang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xiao-Yu Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Chang-Bin Ke
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China.
| | - Zhi-Jun Pei
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan 442000, Hubei, China.
| |
Collapse
|
2
|
Alam S, Doherty E, Ortega-Prieto P, Arizanova J, Fets L. Membrane transporters in cell physiology, cancer metabolism and drug response. Dis Model Mech 2023; 16:dmm050404. [PMID: 38037877 PMCID: PMC10695176 DOI: 10.1242/dmm.050404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
By controlling the passage of small molecules across lipid bilayers, membrane transporters influence not only the uptake and efflux of nutrients, but also the metabolic state of the cell. With more than 450 members, the Solute Carriers (SLCs) are the largest transporter super-family, clustering into families with different substrate specificities and regulatory properties. Cells of different types are, therefore, able to tailor their transporter expression signatures depending on their metabolic requirements, and the physiological importance of these proteins is illustrated by their mis-regulation in a number of disease states. In cancer, transporter expression is heterogeneous, and the SLC family has been shown to facilitate the accumulation of biomass, influence redox homeostasis, and also mediate metabolic crosstalk with other cell types within the tumour microenvironment. This Review explores the roles of membrane transporters in physiological and malignant settings, and how these roles can affect drug response, through either indirect modulation of sensitivity or the direct transport of small-molecule therapeutic compounds into cells.
Collapse
Affiliation(s)
- Sara Alam
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Emily Doherty
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Paula Ortega-Prieto
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Julia Arizanova
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Louise Fets
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
3
|
Tang YC, Yang CS, Liang MX, Zhang Y, Liu Y, Zou SH, Shi SF. Development and evaluation of an adenosine-to-inosine RNA editing-based prognostic model for survival prediction of bladder cancer patients. Medicine (Baltimore) 2023; 102:e33719. [PMID: 37171335 PMCID: PMC10174396 DOI: 10.1097/md.0000000000033719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/13/2023] Open
Abstract
Adenosine-to-inosine RNA editing (ATIRE) is a common form of ribonucleic acid (RNA) editing, which has highlighted the importance of ATIRE in tumors. However, its role in bladder cancer (BLCA) remains poorly understood. To study ATIRE impact on BLCA patient prognosis, we obtained ATIRE, gene expression, and clinical data from the Cancer Genome Atlas (TCGA) database for 251 patients, randomly dividing them into training and testing groups. Univariate proportional hazards model (COX) regression identified prognosis-associated ATIRE loci, while the least absolute shrinkage and selection operator (LASSO) selected final loci to construct prognostic models and generate ATIRE scores. We developed a nomogram to predict BLCA patients' overall survival (OS) and analyzed the effect of ATIRE editing levels on host gene expression. We also compared immune cell infiltration and drug treatment between patients with high and low ATIRE scores. The ATIRE prognostic prediction model was constructed using ten ATIRE loci that are closely associated with BLCA survival. Patients with high ATIRE scores showed significantly worse OS than those with low ATIRE scores. Furthermore, the nomogram, which incorporates the ATIRE score, can better predict the prognosis of patients. Multiple functional and pathway changes associated with immune responses, as well as significant differences in immune cell infiltration levels and response to drug therapy were observed between patients with high and low ATIRE scores. This study represented the first comprehensive analysis of the role of ATIRE events in BLCA patient prognosis and provided new insights into potential prognostic markers for BLCA research.
Collapse
Affiliation(s)
- Yin-Chao Tang
- Clinical Laboratory, The First People’s Hospital of Huaihua, Huaihua, Hunan, China
| | - Chang-Shun Yang
- Clinical Laboratory, The First People’s Hospital of Huaihua, Huaihua, Hunan, China
| | - Ming-Xing Liang
- Clinical Laboratory, The First People’s Hospital of Huaihua, Huaihua, Hunan, China
| | - Yong Zhang
- Clinical Laboratory, The First People’s Hospital of Huaihua, Huaihua, Hunan, China
| | - Yuan Liu
- Clinical Laboratory, The First People’s Hospital of Huaihua, Huaihua, Hunan, China
| | - Shao-Hui Zou
- Clinical Laboratory, The First People’s Hospital of Huaihua, Huaihua, Hunan, China
| | - Shu-Fan Shi
- Clinical Laboratory, The First People’s Hospital of Huaihua, Huaihua, Hunan, China
| |
Collapse
|
4
|
Hou C, Wu M, Zhang H, Yang Z. The specific phagocytosis regulators could predict recurrence and therapeutic effect in thyroid cancer: A study based on bioinformatics analysis. Medicine (Baltimore) 2023; 102:e33290. [PMID: 36930113 PMCID: PMC10019206 DOI: 10.1097/md.0000000000033290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Thyroid cancer (TC) is one of the growing cancers and is prone to recurrence. Meanwhile, in immunotherapy, antibody-dependent cellular phagocytosis (ADCP) phagocytosis related regulators (PRs) play an important role. This study aims to investigate the prognostic value of specific PRs in TC. METHODS The purpose of this study was to identify specific PRs in TC patients by retrieving RNA-seq and Clustered Regularly Interspaced Short Palindromic Repeats-cas9 data and an algorithm based on LASSO was used to construct the PRs-signature. Subsequently, prognosis value of PRs-signature for recurrence-free survival (RFS) was explored through various statistical analysis, including Cox regression analysis, Kaplan-Meier analysis, and receiver operating characteristic curve. Additionally, an analysis of immune cell content by risk group was conducted using CIBERSORT, single sample gene set enrichment analysis and MCP-counter algorithms, with a particular focus on the correlation between macrophages and specific PRs. RESULTS We identified 36 specific PRs, and a PRs-signature was constructed using 5-prognostic PRs (CAPN6, MUC21, PRDM1, SEL1L3, and CPQ). Receiver operating characteristic analysis showed that predictive power of PRs-signature was decent, and the PRs risk score as an independent prognostic factor was found to be correlated with RFS showed by multivariate cox regression analysis. Meanwhile, a lower RFS was observed in the high-risk group than in the low-risk group. The results of the 3 algorithms suggested that our PRs-signature may have certain significance for macrophage content and ADCP. Interestingly, the low-risk group had higher levels of mRNA expression than the high-risk group at PDCD1, CTLA4, and pro-inflammatory factors from macrophage. CONCLUSION For the purpose of prognostic management, this study developed a prediction model. And the cross-talk between certain PRs and TC patients was revealed in this study. Besides, the PRs-signature can predict the immunotherapy response, macrophage content, and ADCP status. TC patients will benefit from these developments by gaining insight into novel therapeutic strategies.
Collapse
Affiliation(s)
- Changran Hou
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Mengmeng Wu
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| | - Haojie Zhang
- Binzhou Medical University, Yantai, Shandong, P.R. China
| | - Zhenlin Yang
- Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, P.R. China
| |
Collapse
|
5
|
Overexpression of SFXN1 indicates poor prognosis and promotes tumor progression in lung adenocarcinoma. Pathol Res Pract 2022; 237:154031. [PMID: 35878532 DOI: 10.1016/j.prp.2022.154031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 11/20/2022]
Abstract
Sideroflexin 1 (SFXN1) functions as a mitochondrial serine transporter in one-carbon metabolism. The association between SFXN1 and tumorigenesis remains to be elucidated. This study illustrated the functional role of SFXN1 in lung adenocarcinoma (LUAD). SFXN1 expression in LUAD specimens was examined using western blotting and quantitative real-time PCR (qRT-PCR), and the prognostic value between SFXN1 and clinicopathological parameters was investigated. Subsequently, the effects of SFXN1 on cellular proliferation, migration, and apoptosis were assessed by using Transwell assays and flow cytometry in A549 and H1299 cell lines. Western blotting was also employed to explore the mechanism of tumor progression. SFXN1 was significantly elevated in the LUAD samples compared with the para-carcinoma tissues. Furthermore, SFXN1 expression was an independent prognostic predictor for patients with LUAD. The expression of SFXN1 was altered in A549 and H1299 cell lines and this showed that SFXN1 promoted cell proliferation, migration, and invasion and inhibited apoptosis. SFXN1, at least partially, influenced LUAD progression via the mTOR signaling pathway. Collectively, the findings from this study demonstrated that SFXN1 promotes LUAD progression via the mTOR pathway and that SFXN1 expression is associated with clinicopathological features of LUAD. SFXN1 significantly contributes to the development of LUAD and might have potential, not only as an independent prognostic marker of LAUD but also as a promising target for LUAD therapy.
Collapse
|