1
|
Ferrari I, De Grossi F, Lai G, Oliveto S, Deroma G, Biffo S, Manfrini N. CancerHubs: a systematic data mining and elaboration approach for identifying novel cancer-related protein interaction hubs. Brief Bioinform 2024; 26:bbae635. [PMID: 39657701 PMCID: PMC11631132 DOI: 10.1093/bib/bbae635] [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] [Received: 08/14/2024] [Revised: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
Conventional approaches to predict protein involvement in cancer often rely on defining either aberrant mutations at the single-gene level or correlating/anti-correlating transcript levels with patient survival. These approaches are typically conducted independently and focus on one protein at a time, overlooking nucleotide substitutions outside of coding regions or mutational co-occurrences in genes within the same interaction network. Here, we present CancerHubs, a method that integrates unbiased mutational data, clinical outcome predictions and interactomics to define novel cancer-related protein hubs. Through this approach, we identified TGOLN2 as a putative novel broad cancer tumour suppressor and EFTUD2 as a putative novel multiple myeloma oncogene.
Collapse
Affiliation(s)
- Ivan Ferrari
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Federica De Grossi
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giancarlo Lai
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Stefania Oliveto
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giorgia Deroma
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Stefano Biffo
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Nicola Manfrini
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
2
|
Qiao Z, Li Y, Li S, Liu S, Cheng Y. Hypoxia-induced SHMT2 protein lactylation facilitates glycolysis and stemness of esophageal cancer cells. Mol Cell Biochem 2024; 479:3063-3076. [PMID: 38175377 DOI: 10.1007/s11010-023-04913-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024]
Abstract
Esophageal cancer (EC) is a familiar digestive tract tumor with highly lethal. The hypoxic environment has been demonstrated to be a significant factor in modulating malignant tumor progression and is strongly associated with the abnormal energy metabolism of tumor cells. Serine hydroxymethyl transferase 2 (SHMT2) is one of the most frequently expressed metabolic enzymes in human malignancies. The study was designed to investigate the biological functions and regulation mechanisms of SHMT2 in EC under hypoxia. We conducted RT-qPCR to assess SHMT2 levels in EC tissues and cells (TE-1 and EC109). EC cells were incubated under normoxia and hypoxia, respectively, and altered SHMT2 expression was evaluated through RT-qPCR, western blot, and immunofluorescence. The biological functions of SHMT2 on EC cells were monitored by performing CCK-8, EdU, transwell, sphere formation, glucose uptake, and lactate production assays. The SHMT2 protein lactylation was measured by immunoprecipitation and western blot. In addition, SHMT2-interacting proteins were analyzed by bioinformatics and validated by rescue experiments. SHMT2 was notably upregulated in EC tissues and cells. Hypoxia elevated SHMT2 protein expression, augmenting EC cell proliferation, migration, invasion, stemness, and glycolysis. In addition, hypoxia triggered lactylation of the SHMT2 protein and enhanced its stability. SHMT2 knockdown impeded the malignant phenotype of EC cells. Further mechanistic studies disclosed that SHMT2 is involved in EC progression by interacting with MTHFD1L. Hypoxia-induced SHMT2 protein lactylation and upregulated its protein level, which in turn enhanced MTHFD1L expression and accelerated the malignant progression of EC cells.
Collapse
Affiliation(s)
- Zhe Qiao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Yu Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Shaomin Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Shiyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China
| | - Yao Cheng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
3
|
Su SW, Chen X, Wang G, Li P, Yang TX, Fang KW, Wu J, Li JM. A study on the significance of serine hydroxymethyl transferase expression and its role in bladder cancer. Sci Rep 2024; 14:8324. [PMID: 38594513 PMCID: PMC11003972 DOI: 10.1038/s41598-024-58618-2] [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] [Received: 01/07/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024] Open
Abstract
Bladder cancer (BLCA) is a common malignant tumor in urinary system all over the world. However, due to its high recurrence rate and complex causes, clinicians often have limited options for surgical and drug treatments. Recent researchs on the molecular mechanism of BLCA have reveals its biological progress and potential for early diagnosis. Serine hydroxymethyltransferase 1/2 (SHMT1/2) is a crucial enzyme in the one-carbon metabolism of tumor cells, and the expression levels of these isozymes have been found to be associated with the biological progression of various malignant tumors. However, the impact of SHMT1/2 on the biological progression of bladder cancer and its molecular regulation mechanism remain unclear. In this research utilizes BLCA clinical sample data, the TCGA database, and in vitro cell experiments to predict the expression levels of SHMT1/2 in BLCA. The findings indicate that SHMT1 remained unchanged, while SHMT2 expression is increased in BLCA, which was related to poor prognosis. Additionally, SHMT2 affects the growth, migration, and apoptosis of bladder cancer cells in vitro. It also influences the expression levels of E-cadherin and N-cadherin, ultimately impacting the malignant biological progression of bladder tumors. These results establish a correlation between SHMT2 and the malignant biological progression of BLCA, providing a theoretical basis for the early diagnosis and treatment of bladder cancer.
Collapse
Affiliation(s)
- Si-Wei Su
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian Mian Avenue, Kunming, 650101, Yunnan, People's Republic of China
| | - Xian Chen
- The Department of Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guang Wang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian Mian Avenue, Kunming, 650101, Yunnan, People's Republic of China
| | - Pei Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian Mian Avenue, Kunming, 650101, Yunnan, People's Republic of China.
| | - Tong-Xin Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian Mian Avenue, Kunming, 650101, Yunnan, People's Republic of China.
| | - Ke-Wei Fang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian Mian Avenue, Kunming, 650101, Yunnan, People's Republic of China
| | - Jing Wu
- The Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Jiong-Ming Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian Mian Avenue, Kunming, 650101, Yunnan, People's Republic of China
| |
Collapse
|
4
|
Bi X, Wang L, Li H, Ma Y, Guo R, Yue J, Kong L, Gong X, Jiao F, Chinn E, Hu J. MiR-383-5p inhibits the proliferation and migration of lung adenocarcinoma cells by targeting SHMT2. J Cancer 2024; 15:2746-2758. [PMID: 38577602 PMCID: PMC10988301 DOI: 10.7150/jca.89733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/18/2024] [Indexed: 04/06/2024] Open
Abstract
Purpose: To explore the effects of miR-383-5p and serine hydroxymethyltransferase 2 (SHMT2) on the proliferation and migration of lung adenocarcinoma cells. Methods: SHMT2 expression in lung adenocarcinoma and normal tissues was investigated using The Cancer Genome Atlas database. Immunohistochemical analysis was performed to confirm SHMT2 expression in lung adenocarcinoma and adjacent normal lung tissues. Bioinformatics analysis and luciferase reporter assays were used to analyze the relationship between miR-383-5p and SHMT2 expression. The protein expression levels of SHMT2, vimentin, N-cadherin, E-cadherin, Bcl-2, and cyclinD1 were analyzed using western blotting. The reverse transcription-quantitative polymerase chain reaction was used to detect SHMT2 knockdown efficiency, miR-383-5p overexpression, and inhibition efficiency. The proliferative ability of cells was detected using the Cell Counting Kit-8 assay. The Transwell assay was used to detect the migration ability of cells. Results: SHMT2 expression was significantly increased in patients with lung adenocarcinoma compared to that in control patients; the higher the SHMT2 expression the worse the outcomes were in patients with lung adenocarcinoma. SHMT2 knockdown inhibited the proliferation, migration, and epithelial-mesenchymal transition of lung adenocarcinoma A549 and H1299 cells. MiR-383-5p directly targeted and downregulated SHMT2 in A549 and H1299 cells. The effects of miRNA-383-5p on the proliferation and migration of these cells differed from those of SHMT2. Exogenous overexpression of SHMT2 reversed the miR-383-5p-induced proliferation and migration inhibition in A549 and H1299 cells. Conclusion: MiR-383-5p inhibits the proliferation and migration of lung adenocarcinoma cells by targeting and downregulating SHMT2.
Collapse
Affiliation(s)
- Xianxia Bi
- Peninsula Cancer Research Center of Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Luwei Wang
- Peninsula Cancer Research Center of Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Hua Li
- Yantai Environmental Sanitation Management Center, YanTai, Shandong 264000, P.R. China
| | - Ying Ma
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Ruoyu Guo
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Jicheng Yue
- Peninsula Cancer Research Center of Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Lijun Kong
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Xiangqian Gong
- Department of Gastrointestinal Surgery, Yuhuangding Hospital, YanTai, Shandong 265499, P.R. China
| | - Fei Jiao
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Eugene Chinn
- Peninsula Cancer Research Center of Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| | - Jinxia Hu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, Shandong 264003, P.R. China
| |
Collapse
|
5
|
Katinas JM, Nayeen MJ, Schneider M, Shah K, Fifer AN, Klapper LM, Sharma A, Thalluri K, Van Nieuwenhze MS, Hou Z, Gangjee A, Matherly LH, Dann CE. Structural Characterization of 5-Substituted Pyrrolo[3,2- d]pyrimidine Antifolate Inhibitors in Complex with Human Serine Hydroxymethyl Transferase 2. Biochemistry 2024:10.1021/acs.biochem.3c00613. [PMID: 38324671 PMCID: PMC11303599 DOI: 10.1021/acs.biochem.3c00613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
We previously discovered first-in-class multitargeted 5-substituted pyrrolo[3,2-d]pyrimidine antifolates that inhibit serine hydroxymethyltransferase 2 (SHMT2), resulting in potent in vitro and in vivo antitumor efficacies. In this report, we present crystallographic structures for SHMT2 in complex with an expanded series of pyrrolo[3,2-d]pyrimidine compounds with variations in bridge length (3-5 carbons) and the side chain aromatic ring (phenyl, thiophene, fluorine-substituted phenyl, and thiophene). We evaluated structural features of the inhibitor-SHMT2 complexes and correlations to inhibitor potencies (i.e., Kis), highlighting conserved polar contacts and identifying 5-carbon bridge lengths as key determinants of inhibitor potency. Based on the analysis of SHMT2 structural data, we investigated the impact of mutation of Tyr105 in SHMT2 kinetic analysis and studies with HCT116 cells with inducible expression of wild-type and Y105F SHMT2. Increased enzyme inhibition potency by the pyrrolo[3,2-d]pyrimidine inhibitors with Phe105 SHMT2 accompanied an increased growth inhibition of Phe105-expressing HCT116 cells compared to wild-type SHMT2. Pyrrolo[3,2-d]pyrimidine inhibitors with polyglutamate modifications were evaluated for potencies against SHMT2. We determined the crystal structures of SHMT2 in complex with our lead antifolate AGF347 lacking L-glutamate, or as a diglutamate and triglutamate, for comparison with parent AGF347. These data provide the first insights into the influence of antifolate polyglutamylation on SHMT2:inhibitor interactions. Collectively, our results provide new insights into the critical structural determinants of SHMT2 binding by pyrrolo[3,2-d]pyrimidine inhibitors as novel antitumor agents, as well as the first structural characterization of human SHMT2 in complex with polyglutamates of an SHMT2-targeted antifolate.
Collapse
Affiliation(s)
- Jade M Katinas
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Md Junayed Nayeen
- School of Pharmacy & Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Mathew Schneider
- Barbara Ann Karmanos Cancer Institute, Department of Oncology, Wayne State University, 4100 John R, Detroit, Michigan 48201, United States
| | - Khushbu Shah
- School of Pharmacy & Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Alexandra N Fifer
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Lily M Klapper
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Abhishekh Sharma
- School of Pharmacy & Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Kishore Thalluri
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | | | - Zhanjun Hou
- Barbara Ann Karmanos Cancer Institute, Department of Oncology, Wayne State University, 4100 John R, Detroit, Michigan 48201, United States
| | - Aleem Gangjee
- School of Pharmacy & Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Larry H Matherly
- Barbara Ann Karmanos Cancer Institute, Department of Oncology, Wayne State University, 4100 John R, Detroit, Michigan 48201, United States
| | - Charles E Dann
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
6
|
Shan Y, Liu D, Li Y, Wu C, Ye Y. The expression and clinical significance of serine hydroxymethyltransferase2 in gastric cancer. PeerJ 2024; 12:e16594. [PMID: 38188143 PMCID: PMC10771762 DOI: 10.7717/peerj.16594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024] Open
Abstract
Background Gastric cancer (GC) is one of the most common malignant tumours in the digestive system. Serine hydroxymethyltransferase 2 (SHMT2) is one of the key enzymes associated with serine metabolism. However, the prognostic role of SHMT2 in GC carcinogenesis has yet to be studied. Methods The expression of SHMT2 in human tumors and normal tissues was detected by the Assistant for Clinical Bioinformatics and Immunohistochemistry (IHC). The relationship of the expression of SHMT2 with clinical characteristics and survival data was analysed by the chi-square test, survival analysis and online databases. Finally, the correlation between SHMT2 expression and associated signalling channels, and molecules was analysed by online databases. Results SHMT2 was strongly expressed in numerous human cancers. The expression rate of SHMT2 was 56.44% in GC (P = 0.018). The survival analysis indicated that patients with high expression of SHMT2 had the worse overall survival (OS; log-rank P = 0.007). The expression of SHMT2 was correlated with tumour size (P = 0.034) and, TNM stage (P = 0.042). In particular, SHMT2, vessel invasion and M stage were independent factors for OS in GC (P = 0.044, P < 0.001, P < 0.001). The SHMT2 gene was substantially correlated with cell signalling pathways. Conclusions SHMT2 is highly expressed in GC and is associated with a poor prognosis. The exploration of its mechanism may be related to tumour proliferation, DNA repair and replication. SHMT2 is an independent prognostic risk factor and a potential biomarker for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Yiming Shan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongdong Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingze Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chu Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanwei Ye
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Zheng BS, Wang SD, Zhang JY, Ge CG. Expression, Prognostic Value, and Immune Infiltration of MTHFD Family in Bladder Cancer. Curr Cancer Drug Targets 2024; 24:178-191. [PMID: 37539926 DOI: 10.2174/1568009623666230804152603] [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: 04/18/2023] [Revised: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND The Methylenetetrahydrofolate Dehydrogenase (MTHFD) family plays an important role in the development and prognosis of a variety of tumors; however, the role of the MTHFD family in bladder cancer is unclear. METHODS R software, cBioPortal, GeneMANIA, and online sites such as String-LinkedOmics were used for bioinformatics analysis. RESULTS MTHFD1/1L/2 was significantly upregulated in bladder cancer tissues compared with normal tissues, high expression of the MTHFD family was strongly associated with poorer clinical grading and staging, and bladder cancer patients with upregulated expression of MTHFD1L/2 had a significantly worse prognosis. Gene function and PPI network analysis revealed that the MTHFD family and related genes play synergistic roles in the development of bladder cancer. 800 co-expressed genes related to the MTHFD family were used for functional enrichment analysis, and the results showed that many genes were associated with various oncogenic pathways such as cell cycle and DNA replication. More importantly, the MTHFD family was closely associated with multiple infiltrating immune lymphocytes, including Treg cells, and immune molecules such as TNFSF9, CD274, and PDCD1. CONCLUSION Our study shows that MTHFD family genes may be potential prognostic markers and therapeutic targets for patients with bladder cancer.
Collapse
Affiliation(s)
- Bai Shu Zheng
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Shun De Wang
- Department of Urology, The ChenJiaqiao Hospital of ShaPingba District of Chongqing City, Chongqing, 401331, China
| | - Jun Yong Zhang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Cheng Guo Ge
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
8
|
Yue SY, Niu D, Liu XH, Li WY, Ding K, Fang HY, Wu XD, Li C, Guan Y, Du HX. BLCA prognostic model creation and validation based on immune gene-metabolic gene combination. Discov Oncol 2023; 14:232. [PMID: 38103068 PMCID: PMC10725402 DOI: 10.1007/s12672-023-00853-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/14/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Bladder cancer (BLCA) is a prevalent urinary system malignancy. Understanding the interplay of immunological and metabolic genes in BLCA is crucial for prognosis and treatment. METHODS Immune/metabolism genes were extracted, their expression profiles analyzed. NMF clustering found prognostic genes. Immunocyte infiltration and tumor microenvironment were examined. Risk prognostic signature using Cox/LASSO methods was developed. Immunological Microenvironment and functional enrichment analysis explored. Immunotherapy response and somatic mutations evaluated. RT-qPCR validated gene expression. RESULTS We investigated these genes in 614 BLCA samples, identifying relevant prognostic genes. We developed a predictive feature and signature comprising 7 genes (POLE2, AHNAK, SHMT2, NR2F1, TFRC, OAS1, CHKB). This immune and metabolism-related gene (IMRG) signature showed superior predictive performance across multiple datasets and was independent of clinical indicators. Immunotherapy response and immune cell infiltration correlated with the risk score. Functional enrichment analysis revealed distinct biological pathways between low- and high-risk groups. The signature demonstrated higher prediction accuracy than other signatures. qRT-PCR confirmed differential gene expression and immunotherapy response. CONCLUSIONS The model in our work is a novel assessment tool to measure immunotherapy's effectiveness and anticipate BLCA patients' prognosis, offering new avenues for immunological biomarkers and targeted treatments.
Collapse
Affiliation(s)
- Shao-Yu Yue
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Di Niu
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xian-Hong Liu
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Wei-Yi Li
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ke Ding
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Hong-Ye Fang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xin-Dong Wu
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Chun Li
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Yu Guan
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - He-Xi Du
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, People's Republic of China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China.
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| |
Collapse
|
9
|
Qiao Z, Li Y, Cheng Y, Li S, Liu S. SHMT2 regulates esophageal cancer cell progression and immune Escape by mediating m6A modification of c-myc. Cell Biosci 2023; 13:203. [PMID: 37932821 PMCID: PMC10629073 DOI: 10.1186/s13578-023-01148-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND In recent years, the role of altered cellular metabolism in tumor progression has attracted widespread attention. Related metabolic enzymes have also been considered as potential cancer therapeutic targets. Serine hydroxymethyltransferase 2 (SHMT2) has been reported to be upregulated in several cancers and associated with poor prognosis. However, there are few studies of SHMT2 in esophageal cancer (EC), and the related functions and mechanisms also need to be further explored. METHODS In this study, we first analyzed SHMT2 expression in EC by online database and clinical samples. Then, the biological functions of SHMT2 in EC were investigated by cell and animal experiments. The intracellular m6A methylation modification levels were also evaluated by MeRIP. Linked genes and mechanisms of SHMT2 were analyzed by bioinformatics and rescue experiments. RESULTS We found that SHMT2 expression was abnormally upregulated in EC and associated with poor prognosis. Functionally, SHMT2 silencing suppressed c-myc expression in an m6A-dependent manner, thereby blocking the proliferation, migration, invasion and immune escape abilities of EC cells. Mechanistically, SHMT2 encouraged the accumulation of methyl donor SAM through a one-carbon metabolic network, thereby regulating the m6A modification and stability of c-myc mRNA in a METTL3/FTO/ALKBH5/IGF2BP2-dependent way. In vivo animal experiments also demonstrated that SHMT2 mediated MYC expression by m6A-methylation modification, thus boosting EC tumorigenesis. CONCLUSION In conclusion, our data illustrated that SHMT2 regulated malignant progression and immune escape of EC cell through c-myc m6A modification. These revealed mechanisms related to SHMT2 in EC and maybe offer promise for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Zhe Qiao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, 710004, Xi'an, Shaanxi, China
| | - Yu Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, 710004, Xi'an, Shaanxi, China
| | - Yao Cheng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, 710004, Xi'an, Shaanxi, China
| | - Shaomin Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, 710004, Xi'an, Shaanxi, China
| | - Shiyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West 5th Road, 710004, Xi'an, Shaanxi, China.
| |
Collapse
|
10
|
Vit O, Talacko P, Musil Z, Hartmann I, Pacak K, Petrak J. Identification of potential molecular targets for the treatment of cluster 1 human pheochromocytoma and paraganglioma via comprehensive proteomic characterization. Clin Proteomics 2023; 20:39. [PMID: 37749499 PMCID: PMC10518975 DOI: 10.1186/s12014-023-09428-7] [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: 03/02/2023] [Accepted: 08/21/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors. New drug targets and proteins that would assist sensitive PPGL imagining could improve therapy and quality of life of patients with PPGL, namely those with recurrent or metastatic disease. Using a combined proteomic strategy, we looked for such clinically relevant targets among integral membrane proteins (IMPs) upregulated on the surface of tumor cells and non-membrane druggable enzymes in PPGL. METHODS We conducted a detailed proteomic analysis of 22 well-characterized human PPGL samples and normal chromaffin tissue from adrenal medulla. A standard quantitative proteomic analysis of tumor lysate, which provides information largely on non-membrane proteins, was accompanied by specific membrane proteome-aimed methods, namely glycopeptide enrichment using lectin-affinity, glycopeptide capture by hydrazide chemistry, and enrichment of membrane-embedded hydrophobic transmembrane segments. RESULTS The study identified 67 cell surface integral membrane proteins strongly upregulated in PPGL compared to control chromaffin tissue. We prioritized the proteins based on their already documented direct role in cancer cell growth or progression. Increased expression of the seven most promising drug targets (CD146, CD171, ANO1, CD39, ATP8A1, ACE and SLC7A1) were confirmed using specific antibodies. Our experimental strategy also provided expression data for soluble proteins. Among the druggable non-membrane enzymes upregulated in PPGL, we identified three potential drug targets (SHMT2, ARG2 and autotaxin) and verified their upregulated expression. CONCLUSIONS Application of a combined proteomic strategy recently presented as "Pitchfork" enabled quantitative analysis of both, membrane and non-membrane proteome, and resulted in identification of 10 potential drug targets in human PPGL. Seven membrane proteins localized on the cell surface and three non-membrane druggable enzymes proteins were identified and verified as significantly upregulated in PPGL. All the proteins have been previously shown to be upregulated in several human cancers, and play direct role in cancer progression. Marked upregulation of these proteins along with their localization and established direct roles in tumor progression make these molecules promising candidates as drug targets or proteins for sensitive PPGL imaging.
Collapse
Affiliation(s)
- Ondrej Vit
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, 25250, Czech Republic
| | - Pavel Talacko
- Proteomics Core Facility, Faculty of Science, BIOCEV, Charles University, Vestec, 25250, Czech Republic
| | - Zdenek Musil
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital, Prague, 12800, Czech Republic
| | - Igor Hartmann
- Department of Urology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, 77900, Czech Republic
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, 20892, USA
| | - Jiri Petrak
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, 25250, Czech Republic.
| |
Collapse
|
11
|
Nayeen MJ, Katinas JM, Magdum T, Shah K, Wong JE, O’Connor CE, Fifer AN, Wallace-Povirk A, Hou Z, Matherly LH, Dann CE, Gangjee A. Structure-Based Design of Transport-Specific Multitargeted One-Carbon Metabolism Inhibitors in Cytosol and Mitochondria. J Med Chem 2023; 66:11294-11323. [PMID: 37582241 PMCID: PMC10461232 DOI: 10.1021/acs.jmedchem.3c00763] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Indexed: 08/17/2023]
Abstract
Multitargeted agents provide tumor selectivity with reduced drug resistance and dose-limiting toxicities. We previously described the multitargeted 6-substituted pyrrolo[3,2-d]pyrimidine antifolate 1 with activity against early- and late-stage pancreatic tumors with limited tumor selectivity. Structure-based design with our human serine hydroxymethyl transferase (SHMT) 2 and glycinamide ribonucleotide formyltransferase (GARFTase) structures, and published X-ray crystal structures of 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase (ATIC), SHMT1, and folate receptor (FR) α and β afforded 11 analogues. Multitargeted inhibition and selective tumor transport were designed by providing promiscuous conformational flexibility in the molecules. Metabolite rescue identified mitochondrial C1 metabolism along with de novo purine biosynthesis as the targeted pathways. We identified analogues with tumor-selective transport via FRs and increased SHMT2, SHMT1, and GARFTase inhibition (28-, 21-, and 11-fold, respectively) compared to 1. These multitargeted agents represent an exciting new structural motif for targeted cancer therapy with substantial advantages of selectivity and potency over clinically used antifolates.
Collapse
Affiliation(s)
- Md. Junayed Nayeen
- Division
of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Jade M. Katinas
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47408, United States
| | - Tejashree Magdum
- Division
of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Khushbu Shah
- Division
of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Jennifer E. Wong
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47408, United States
| | - Carrie E. O’Connor
- Department
of Oncology, Wayne State University School
of Medicine, Detroit, Michigan 48201, United States
| | - Alexandra N. Fifer
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47408, United States
| | - Adrianne Wallace-Povirk
- Department
of Oncology, Wayne State University School
of Medicine, Detroit, Michigan 48201, United States
| | - Zhanjun Hou
- Department
of Oncology, Wayne State University School
of Medicine, Detroit, Michigan 48201, United States
- Molecular
Therapeutics Program, Barbara Ann Karmanos
Cancer Institute, 4100 John R, Detroit, Michigan 48201, United States
| | - Larry H. Matherly
- Department
of Oncology, Wayne State University School
of Medicine, Detroit, Michigan 48201, United States
- Molecular
Therapeutics Program, Barbara Ann Karmanos
Cancer Institute, 4100 John R, Detroit, Michigan 48201, United States
| | - Charles E. Dann
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47408, United States
| | - Aleem Gangjee
- Division
of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| |
Collapse
|
12
|
Shuvalov O, Kirdeeva Y, Fefilova E, Netsvetay S, Zorin M, Vlasova Y, Fedorova O, Daks A, Parfenyev S, Barlev N. 20-Hydroxyecdysone Confers Antioxidant and Antineoplastic Properties in Human Non-Small Cell Lung Cancer Cells. Metabolites 2023; 13:metabo13050656. [PMID: 37233697 DOI: 10.3390/metabo13050656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
20-Hydroxyecdysone (20E) is an arthropod hormone which is synthesized by some plants as part of their defense mechanism. In humans, 20E has no hormonal activity but possesses a number of beneficial pharmacological properties including anabolic, adaptogenic, hypoglycemic, and antioxidant properties, as well as cardio-, hepato-, and neuroprotective features. Recent studies have shown that 20E may also possess antineoplastic activity. In the present study, we reveal the anticancer properties of 20E in Non-Small Cell Lung Cancer (NSCLC) cell lines. 20E displayed significant antioxidant capacities and induced the expression of antioxidative stress response genes. The RNA-seq analysis of 20E-treated lung cancer cells revealed the attenuation of genes involved in different metabolic processes. Indeed, 20E suppressed several enzymes of glycolysis and one-carbon metabolism, as well as their key transcriptional regulators-c-Myc and ATF4, respectively. Accordingly, using the SeaHorse energy profiling approach, we observed the inhibition of glycolysis and respiration mediated by 20E treatment. Furthermore, 20E sensibilized lung cancer cells to metabolic inhibitors and markedly suppressed the expression of Cancer Stem Cells (CSCs) markers. Thus, in addition to the known beneficial pharmacological activities of 20E, our data uncovered novel antineoplastic properties of 20E in NSCLC cells.
Collapse
Affiliation(s)
- Oleg Shuvalov
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Yulia Kirdeeva
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Elizaveta Fefilova
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Sofia Netsvetay
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Mark Zorin
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Yulia Vlasova
- Almazov National Medical Research Center Russia, 197341 St. Petersburg, Russia
| | - Olga Fedorova
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Alexandra Daks
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Sergey Parfenyev
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Nickolai Barlev
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
- School of Medicine, Nazarbayev University, 001000 Astana, Kazakhstan
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
13
|
Wang W, Wang M, Du T, Hou Z, You S, Zhang S, Ji M, Xue N, Chen X. SHMT2 Promotes Gastric Cancer Development through Regulation of HIF1α/VEGF/STAT3 Signaling. Int J Mol Sci 2023; 24:ijms24087150. [PMID: 37108312 PMCID: PMC10138966 DOI: 10.3390/ijms24087150] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
The metabolic enzymes involved in one-carbon metabolism are closely associated with tumor progression and could be potential targets for cancer therapy. Recent studies showed that serine hydroxymethyltransferase 2 (SHMT2), a crucial enzyme in the one-carbon metabolic pathway, plays a key role in tumor proliferation and development. However, the precise role and function of SHMT2 in gastric cancer (GC) remain poorly understood. In this study, we presented evidence that SHMT2 was necessary for hypoxia-inducible factor-1α (HIF1α) stability and contributed to GC cells' hypoxic adaptation. The analysis of datasets retrieved from The Cancer Genome Atlas and the experimentation with human cell lines revealed a marked increase in SHMT2 expression in GC. The SHMT2 knockdown in MGC803, SGC7901, and HGC27 cell lines inhibited cell proliferation, colony formation, invasion, and migration. Notably, SHMT2 depletion disrupted redox homeostasis and caused glycolytic function loss in GC cells under hypoxic circumstances. Mechanistically, we discovered SHMT2 modulated HIF1α stability, which acted as a master regulator of hypoxia-inducible genes under hypoxic conditions. This, in turn, regulated the downstream VEGF and STAT3 pathways. The in vivo xenograft experiments showed that SHMT2 knockdown markedly reduced GC growth. Our results elucidate the novel function of SHMT2 in stabilizing HIF1α under hypoxic conditions, thus providing a potential therapeutic strategy for GC treatment.
Collapse
Affiliation(s)
- Weida Wang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Mingjin Wang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Tingting Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Zhenyan Hou
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Shen You
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Sen Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Ming Ji
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Nina Xue
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| | - Xiaoguang Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China
| |
Collapse
|
14
|
Zhou X, Tian C, Cao Y, Zhao M, Wang K. The role of serine metabolism in lung cancer: From oncogenesis to tumor treatment. Front Genet 2023; 13:1084609. [PMID: 36699468 PMCID: PMC9868472 DOI: 10.3389/fgene.2022.1084609] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Metabolic reprogramming is an important hallmark of malignant tumors. Serine is a non-essential amino acid involved in cell proliferation. Serine metabolism, especially the de novo serine synthesis pathway, forms a metabolic network with glycolysis, folate cycle, and one-carbon metabolism, which is essential for rapidly proliferating cells. Owing to the rapid development in metabolomics, abnormal serine metabolism may serve as a biomarker for the early diagnosis and pathological typing of tumors. Targeting serine metabolism also plays an essential role in precision and personalized cancer therapy. This article is a systematic review of de novo serine biosynthesis and the link between serine and folate metabolism in tumorigenesis, particularly in lung cancer. In addition, we discuss the potential of serine metabolism to improve tumor treatment.
Collapse
|
15
|
Zhang J, Ma C, Qin H, Wang Z, Zhu C, Liu X, Hao X, Liu J, Li L, Cai Z. Construction and validation of a metabolic-related genes prognostic model for oral squamous cell carcinoma based on bioinformatics. BMC Med Genomics 2022; 15:269. [PMID: 36566175 PMCID: PMC9789624 DOI: 10.1186/s12920-022-01417-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) accounts for a frequently-occurring head and neck cancer, which is characterized by high rates of morbidity and mortality. Metabolism-related genes (MRGs) show close association with OSCC development, metastasis and progression, so we constructed an MRGs-based OSCC prognosis model for evaluating OSCC prognostic outcome. METHODS This work obtained gene expression profile as well as the relevant clinical information from the The Cancer Genome Atlas (TCGA) database, determined the MRGs related to OSCC by difference analysis, screened the prognosis-related MRGs by performing univariate Cox analysis, and used such identified MRGs for constructing the OSCC prognosis prediction model through Lasso-Cox regression. Besides, we validated the model with the GSE41613 dataset based on Gene Expression Omnibus (GEO) database. RESULTS The present work screened 317 differentially expressed MRGs from the database, identified 12 OSCC prognostic MRGs through univariate Cox regression, and then established a clinical prognostic model composed of 11 MRGs by Lasso-Cox analysis. Based on the optimal risk score threshold, cases were classified as low- or high-risk group. As suggested by Kaplan-Meier (KM) analysis, survival rate was obviously different between the two groups in the TCGA training set (P < 0.001). According to subsequent univariate and multivariate Cox regression, risk score served as the factor to predict prognosis relative to additional clinical features (P < 0.001). Besides, area under ROC curve (AUC) values for patient survival at 1, 3 and 5 years were determined as 0.63, 0.70, and 0.76, separately, indicating that the prognostic model has good predictive accuracy. Then, we validated this clinical prognostic model using GSE41613. To enhance our model prediction accuracy, age, gender, risk score together with TNM stage were incorporated in a nomogram. As indicated by results of ROC curve and calibration curve analyses, the as-constructed nomogram had enhanced prediction accuracy compared with clinicopathological features alone, besides, combining clinicopathological characteristics with risk score contributed to predicting patient prognosis and guiding clinical decision-making. CONCLUSION In this study, 11 MRGs prognostic models based on TCGA database showed superior predictive performance and had a certain clinical application prospect in guiding individualized.
Collapse
Affiliation(s)
- Jingfei Zhang
- grid.440653.00000 0000 9588 091XDepartment of Stomatology, Binzhou Medical University, Yantai, 264000 Shandong China
| | - Chenxi Ma
- grid.27255.370000 0004 1761 1174Department of Human Microbiome, School and Hospital of Stomatology, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong University, Jinan, 250000 Shandong China
| | - Han Qin
- grid.440653.00000 0000 9588 091XDepartment of Stomatology, Binzhou Medical University, Yantai, 264000 Shandong China
| | - Zhi Wang
- grid.415946.b0000 0004 7434 8069Department of Stomatology, Linyi People’s Hospital, Linyi, 276000 Shandong China
| | - Chao Zhu
- grid.415946.b0000 0004 7434 8069Department of Stomatology, Linyi People’s Hospital, Linyi, 276000 Shandong China
| | - Xiujuan Liu
- grid.415946.b0000 0004 7434 8069Department of Stomatology, Linyi People’s Hospital, Linyi, 276000 Shandong China
| | - Xiuyan Hao
- grid.415946.b0000 0004 7434 8069Department of Stomatology, Linyi People’s Hospital, Linyi, 276000 Shandong China
| | - Jinghua Liu
- grid.415946.b0000 0004 7434 8069Department of Hepatobiliary Surgery and Minimally Invasive Institute of Digestive Surgery and Prof. Cai’s Laboratory, Linyi People’s Hospital, Shandong University, Linyi, 264000 Shandong China
| | - Ling Li
- grid.415946.b0000 0004 7434 8069Department of Stomatology, Linyi People’s Hospital, Linyi, 276000 Shandong China
| | - Zhen Cai
- grid.415946.b0000 0004 7434 8069Department of Stomatology, Linyi People’s Hospital, Linyi, 276000 Shandong China
| |
Collapse
|
16
|
Mao Y, Zhang T. Knockdown of SHMT2 enhances the sensitivity of gastric cancer cells to radiotherapy through the Wnt/β-catenin pathway. Open Life Sci 2022; 17:1249-1255. [PMID: 36213384 PMCID: PMC9490860 DOI: 10.1515/biol-2022-0480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/20/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors. The mechanism of GC radioresistance and new radiosensitizers must be revealed and developed to treat GC. Serine hydroxymethyltransferase 2 (SHMT2) is responsible for encoding the mitochondrial form of the pyridoxal phosphate-dependent enzyme. SHMT2 plays a critical role in several types of cancers, while its possible effect on the radiological resistance in GC is still unclear. In this study, we investigated the role of SHMT2 in the radiological resistance of GC. Our data confirmed that SHMT2 was highly expressed in radiation-resistant GC cells. SHMT2 reduced the radiosensitivity of GC cells. In addition, SHMT2 is involved in radiation-induced GC cell apoptosis. Further, SHMT2 regulated the Wnt/β-catenin pathway, therefore reducing the radiosensitivity of GC cells in vivo. In conclusion, we revealed that depletion of SHMT2 enhanced the sensitivity of GC cells to interventional radiotherapy through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yu Mao
- Department of General Surgery Unit 1, Yuhuan People’s Hospital , Taizhou , Zhejiang Province, 317699 , China
| | - Tiyong Zhang
- Department of Radiology, Qianjiang Central Hospital of Chongqing Municipality , No. 63, West Ninth Road , Qianjiang District City , Chongqing, 409000 , China
| |
Collapse
|
17
|
Jin Y, Jung SN, Lim MA, Oh C, Piao Y, Kim HJ, Nguyena Q, Kang YE, Chang JW, Won HR, Koo BS. SHMT2 Induces Stemness and Progression of Head and Neck Cancer. Int J Mol Sci 2022; 23:ijms23179714. [PMID: 36077112 PMCID: PMC9456418 DOI: 10.3390/ijms23179714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Various enzymes in the one-carbon metabolic pathway are closely related to the development of tumors, and they can all be potential targets for cancer therapy. Serine hydroxymethyltransferase2 (SHMT2), a key metabolic enzyme, is very important for the proliferation and growth of cancer cells. However, the function and mechanism of SHMT2 in head and neck cancer (HNC) are not clear. An analysis of The Cancer Genome Atlas (TCGA) data showed that the expression of SHMT2 was higher in tumor tissue than in normal tissue, and its expression was significantly associated with male sex, aggressive histological grade, lymph node metastasis, distant metastasis, advanced TNM stage, and lymphovascular invasion in HNC. SHMT2 knockdown in FADU and SNU1041 cell lines significantly inhibited cell proliferation, colony formation, migration, and invasion. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses using TCGA data revealed that SHMT2 was closely related to cancer stem cell regulation and maintenance. Furthermore, we found that silencing SHMT2 inhibited the expression of stemness markers and tumor spheroid formation compared with a control group. On the contrary, stemness markers were significantly increased after SHMT2 overexpression in HEP-2 cells. Interestingly, we found that knocking down SHMT2 reduced the expression of genes related to the Notch and Wnt pathways. Finally, silencing SHMT2 significantly reduced tumor growth and decreased stemness markers in a xenograft model. Taken together, our study suggests that targeting SHMT2 may play an important role in inhibiting HNC progression.
Collapse
Affiliation(s)
- Yanli Jin
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Seung-Nam Jung
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Mi Ae Lim
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Chan Oh
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Yudan Piao
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Hae Jong Kim
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - QuocKhanh Nguyena
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Jae Won Chang
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Ho-Ryun Won
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Bon Seok Koo
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Correspondence: ; Tel.: +82-42-280-7690
| |
Collapse
|
18
|
Zhang H, Che Y, Xuan B, Wu X, Li H. Serine hydroxymethyltransferase 2 (SHMT2) potentiates the aggressive process of oral squamous cell carcinoma by binding to interleukin enhancer-binding factor 2 (ILF2). Bioengineered 2022; 13:8785-8797. [PMID: 35333683 PMCID: PMC9161932 DOI: 10.1080/21655979.2022.2051886] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a frequent threatening head and neck malignancy. Serine hydroxymethyltransferase 2 (SHMT2) was identified to be upregulated in OSCC and its high expression was associated with poor patient prognosis. This paper set out to assess the influence of SHMT2 on OSCC progression and the potential mechanisms related to interleukin enhancer-binding factor 2 (ILF2). First of all, reverse transcription-quantitative PCR (RT-qPCR) and western blot examined the expression of SHMT2 and ILF2 in OSCC cells. Cell Counting Kit-8 (CCK-8) and colony formation assays appraised cell proliferation. Terminal-deoxynucleotidyl Transferase Mediated Nick End Labeling (TUNEL) staining was to estimate the apoptotic rate of cells. Further, wound healing and transwell assays verified the migration and invasion of cells. Western blot was adopted to detect the expression of factors related to apoptosis, migration, and epithelial–mesenchymal transition (EMT). The possible interaction of SHMT2 and ILF2 was predicted by a Molecular INTeraction (MINT) and BioGRID databases and determined using co-immunoprecipitation (IP) assay. Subsequently, ILF2 was overexpressed to investigate whether SHMT2 regulated OSCC progression by binding to ILF2. Results implied that SHMT2 possessed increased expression in OSCC cells, and OSCC cell viability, migration, invasion, EMT were inhibited and apoptosis was potentiated after its silencing. ILF2 bound to SHMT2 and ILF2 expression was downregulated after SHMT2 silencing in OSCC cells. Importantly, ILF2 overexpression abolished the suppressive role of SHMT2 interference in the progression of OSCC. Collectively, SHMT2 could promote the progression of OSCC by binding to ILF2.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Stomatology, Aerospace Center Hospital, Beijing, China
| | - Yilei Che
- Department of Stomatology, Aerospace Center Hospital, Beijing, China
| | - Bin Xuan
- Department of Stomatology, Aerospace Center Hospital, Beijing, China
| | - Xiaozhen Wu
- Department of Stomatology, Aerospace Center Hospital, Beijing, China
| | - Hui Li
- Department of Stomatology, Aerospace Center Hospital, Beijing, China
| |
Collapse
|