1
|
Cheng X, Jiang G, Zhou X, Wang J, Zhao Z, Zhang J, Ni T. The landscape and clinical relevance of intronic polyadenylation in human cancers. J Genet Genomics 2024; 51:1030-1039. [PMID: 38740258 DOI: 10.1016/j.jgg.2024.04.014] [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: 01/09/2024] [Revised: 04/07/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
Intronic polyadenylation (IPA) is an RNA 3' end processing event which has been reported to play important roles in cancer development. However, the comprehensive landscape of IPA events across various cancer types is lacking. Here, we apply IPAFinder to identify and quantify IPA events in 10,383 samples covering all 33 cancer types from The Cancer Genome Atlas (TCGA) project. We identify a total of 21,835 IPA events, almost half of which are ubiquitously expressed. We identify 2761 unique dynamically changed IPA events across cancer types. Furthermore, we observe 8855 non-redundant clinically relevant IPA events, which could potentially be used as prognostic indicators. Our analysis also reveals that dynamic IPA usage within cancer signaling pathways may affect drug response. Finally, we develop a user-friendly data portal, IPACancer Atlas (http://www.tingni-lab.com/Pancan_IPA/), to search and explore IPAs in cancer.
Collapse
Affiliation(s)
- Xiaomeng Cheng
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Guanghui Jiang
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaolan Zhou
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jing Wang
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Zhaozhao Zhao
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China; MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jiayu Zhang
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010070, China.
| |
Collapse
|
2
|
Binmujlli MA. Radiological and Molecular Analysis of Radioiodinated Anastrozole and Epirubicin as Innovative Radiopharmaceuticals Targeting Methylenetetrahydrofolate Dehydrogenase 2 in Solid Tumors. Pharmaceutics 2024; 16:616. [PMID: 38794278 PMCID: PMC11126143 DOI: 10.3390/pharmaceutics16050616] [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: 02/20/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic field of radiopharmaceuticals, innovating targeted agents for cancer diagnosis and therapy is crucial. Our study enriches this evolving landscape by evaluating the potential of radioiodinated anastrozole ([125I]anastrozole) and radioiodinated epirubicin ([125I]epirubicin) as targeting agents against MTHFD2-driven tumors. MTHFD2, which is pivotal in one-carbon metabolism, is notably upregulated in various cancers, presenting a novel target for radiopharmaceutical application. Through molecular docking and 200 ns molecular dynamics (MD) simulations, we assess the binding efficiency and stability of [125I]anastrozole and [125I]epirubicin with MTHFD2. Molecular docking illustrates that [125I]epirubicin has a superior binding free energy (∆Gbind) of -41.25 kJ/mol compared to -39.07 kJ/mol for [125I]anastrozole and -38.53 kJ/mol for the control ligand, suggesting that it has a higher affinity for MTHFD2. MD simulations reinforce this, showing stable binding, as evidenced by root mean square deviation (RMSD) values within a narrow range, underscoring the structural integrity of the enzyme-ligand complexes. The root mean square fluctuation (RMSF) analysis indicates consistent dynamic behavior of the MTHFD2 complex upon binding with [125I]anastrozole and [125I]epirubicin akin to the control. The radius of gyration (RG) measurements of 16.90 Å for MTHFD2-[125I]anastrozole and 16.84 Å for MTHFD2-[125I]epirubicin confirm minimal structural disruption upon binding. The hydrogen bond analysis reveals averages of two and three stable hydrogen bonds for [125I]anastrozole and [125I]epirubicin complexes, respectively, highlighting crucial stabilizing interactions. The MM-PBSA calculations further endorse the thermodynamic favorability of these interactions, with binding free energies of -48.49 ± 0.11 kJ/mol for [125I]anastrozole and -43.8 kJ/mol for MTHFD2-. The significant contribution of Van der Waals and electrostatic interactions to the binding affinities of [125I]anastrozole and [125I]epirubicin, respectively, underscores their potential efficacy for targeted tumor imaging and therapy. These computational findings lay the groundwork for the future experimental validation of [125I]anastrozole and [125I]epirubicin as MTHFD2 inhibitors, heralding a notable advancement in precision oncology tools. The data necessitate subsequent in vitro and in vivo assays to corroborate these results.
Collapse
Affiliation(s)
- Mazen Abdulrahman Binmujlli
- Department of Internal Medicine, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh 11623, Saudi Arabia
| |
Collapse
|
3
|
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
|
4
|
Li Y, Li C, Luo T, Yue T, Xiao W, Yang L, Zhang Z, Han F, Long P, Hu Y. Progress in the Treatment of High Altitude Cerebral Edema: Targeting REDOX Homeostasis. J Inflamm Res 2023; 16:2645-2660. [PMID: 37383357 PMCID: PMC10296571 DOI: 10.2147/jir.s415695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/15/2023] [Indexed: 06/30/2023] Open
Abstract
With the increasing of altitude activities from low-altitude people, the study of high altitude cerebral edema (HACE) has been revived. HACE is a severe acute mountain sickness associated with exposure to hypobaric hypoxia at high altitude, often characterized by disturbance of consciousness and ataxia. As for the pathogenesis of HACE, previous studies suggested that it might be related to the disorder of cerebral blood flow, the destruction of blood-brain barrier and the injury of brain parenchyma cells caused by inflammatory factors. In recent years, studies have confirmed that the imbalance of REDOX homeostasis is also involved in the pathogenesis of HACE, which mainly leads to abnormal activation of microglia and destruction of tight junction of vascular endothelial cells through the excessive production of mitochondrial-related reactive oxygen species. Therefore, this review summarizes the role of REDOX homeostasis and the potential of the treatment of REDOX homeostasis in HACE, which is of great significance to expand the understanding of the pathogenesis of HACE. Moreover, it will also be helpful to further study the possible therapy of HACE related to the key link of REDOX homeostasis.
Collapse
Affiliation(s)
- Yubo Li
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, People’s Republic of China
- Basic Medical Laboratory, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Chengming Li
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, People’s Republic of China
- Basic Medical Laboratory, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Tao Luo
- Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Ling Yang
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, People’s Republic of China
- Basic Medical Laboratory, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Zaiyuan Zhang
- College of Medicine, Southwest Jiaotong University, Chengdu, People’s Republic of China
| | - Fei Han
- Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Pan Long
- Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Yonghe Hu
- College of Medicine, Southwest Jiaotong University, Chengdu, People’s Republic of China
| |
Collapse
|
5
|
Geitgey DK, Lee M, Cottrill KA, Jaffe M, Pilcher W, Bhasin S, Randall J, Ross AJ, Salemi M, Castillo-Castrejon M, Kilgore MB, Brown AC, Boss JM, Johnston R, Fitzpatrick AM, Kemp ML, English R, Weaver E, Bagchi P, Walsh R, Scharer CD, Bhasin M, Chandler JD, Haynes KA, Wellberg EA, Henry CJ. The 'omics of obesity in B-cell acute lymphoblastic leukemia. J Natl Cancer Inst Monogr 2023; 2023:12-29. [PMID: 37139973 PMCID: PMC10157791 DOI: 10.1093/jncimonographs/lgad014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 05/05/2023] Open
Abstract
The obesity pandemic currently affects more than 70 million Americans and more than 650 million individuals worldwide. In addition to increasing susceptibility to pathogenic infections (eg, SARS-CoV-2), obesity promotes the development of many cancer subtypes and increases mortality rates in most cases. We and others have demonstrated that, in the context of B-cell acute lymphoblastic leukemia (B-ALL), adipocytes promote multidrug chemoresistance. Furthermore, others have demonstrated that B-ALL cells exposed to the adipocyte secretome alter their metabolic states to circumvent chemotherapy-mediated cytotoxicity. To better understand how adipocytes impact the function of human B-ALL cells, we used a multi-omic RNA-sequencing (single-cell and bulk transcriptomic) and mass spectroscopy (metabolomic and proteomic) approaches to define adipocyte-induced changes in normal and malignant B cells. These analyses revealed that the adipocyte secretome directly modulates programs in human B-ALL cells associated with metabolism, protection from oxidative stress, increased survival, B-cell development, and drivers of chemoresistance. Single-cell RNA sequencing analysis of mice on low- and high-fat diets revealed that obesity suppresses an immunologically active B-cell subpopulation and that the loss of this transcriptomic signature in patients with B-ALL is associated with poor survival outcomes. Analyses of sera and plasma samples from healthy donors and those with B-ALL revealed that obesity is associated with higher circulating levels of immunoglobulin-associated proteins, which support observations in obese mice of altered immunological homeostasis. In all, our multi-omics approach increases our understanding of pathways that may promote chemoresistance in human B-ALL and highlight a novel B-cell-specific signature in patients associated with survival outcomes.
Collapse
Affiliation(s)
- Delaney K Geitgey
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
| | - Miyoung Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
| | - Kirsten A Cottrill
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Maya Jaffe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - William Pilcher
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Swati Bhasin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Jessica Randall
- Emory Integrated Computational Core, Emory University, Atlanta, GA, USA
| | - Anthony J Ross
- Riley Children’s Health, Indiana University Health, Indianapolis, IN, USA
| | - Michelle Salemi
- Proteomics Core Facility, University of California Davis Genome Center, Davis, 95616, CA
| | - Marisol Castillo-Castrejon
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew B Kilgore
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ayjha C Brown
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Rich Johnston
- Emory Integrated Computational Core, Emory University, Atlanta, GA, USA
| | - Anne M Fitzpatrick
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Melissa L Kemp
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Emory Integrated Proteomics Core, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Eric Weaver
- Shimadzu Scientific Instruments, Columbia, MD, USA
| | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan Walsh
- Shimadzu Scientific Instruments, Columbia, MD, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Manoj Bhasin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Joshua D Chandler
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Karmella A Haynes
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Curtis J Henry
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Atlanta, GA, USA
- Children’s Healthcare of Atlanta, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
| |
Collapse
|
6
|
Tian H, Liu Z, Liu J, Zong Z, Chen Y, Zhang Z, Li H. Application of machine learning algorithm in predicting distant metastasis of T1 gastric cancer. Sci Rep 2023; 13:5741. [PMID: 37029221 PMCID: PMC10082185 DOI: 10.1038/s41598-023-31880-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/20/2023] [Indexed: 04/09/2023] Open
Abstract
Distant metastasis (DM) is relatively uncommon in T1 stage gastric cancer (GC). The aim of this study was to develop and validate a predictive model for DM in stage T1 GC using machine learning (ML) algorithms. Patients with stage T1 GC from 2010 to 2017 were screened from the public Surveillance, Epidemiology and End Results (SEER) database. Meanwhile, we collected patients with stage T1 GC admitted to the Department of Gastrointestinal Surgery of the Second Affiliated Hospital of Nanchang University from 2015 to 2017. We applied seven ML algorithms: logistic regression, random forest (RF), LASSO, support vector machine, k-Nearest Neighbor, Naive Bayesian Model, Artificial Neural Network. Finally, a RF model for DM of T1 GC was developed. The AUC, sensitivity, specificity, F1-score and accuracy were used to evaluate and compare the predictive performance of the RF model with other models. Finally, we performed a prognostic analysis of patients who developed distant metastases. Independent risk factors for prognosis were analysed by univariate and multifactorial regression. K-M curves were used to express differences in survival prognosis for each variable and subvariable. A total of 2698 cases were included in the SEER dataset, 314 with DM, and 107 hospital patients were included, 14 with DM. Age, T-stage, N-stage, tumour size, grade and tumour location were independent risk factors for the development of DM in stage T1 GC. A combined analysis of seven ML algorithms in the training and test sets found that the RF prediction model had the best prediction performance (AUC: 0.941, Accuracy: 0.917, Recall: 0.841, Specificity: 0.927, F1-score: 0.877). The external validation set ROCAUC was 0.750. Meanwhile, survival prognostic analysis showed that surgery (HR = 3.620, 95% CI 2.164-6.065) and adjuvant chemotherapy (HR = 2.637, 95% CI 2.067-3.365) were independent risk factors for survival prognosis in patients with DM from stage T1 GC. Age, T-stage, N-stage, tumour size, grade and tumour location were independent risk factors for the development of DM in stage T1 GC. ML algorithms had shown that RF prediction models had the best predictive efficacy to accurately screen at-risk populations for further clinical screening for metastases. At the same time, aggressive surgery and adjuvant chemotherapy can improve the survival rate of patients with DM.
Collapse
Affiliation(s)
- HuaKai Tian
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zitao Liu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiang Liu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - YanMei Chen
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zuo Zhang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanchang University, 1 MinDe Road, Nanchang, 330006, China.
| | - Hui Li
- Department of Rheumatology and Immunology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
7
|
Islam A, Shaukat Z, Hussain R, Gregory SL. One-Carbon and Polyamine Metabolism as Cancer Therapy Targets. Biomolecules 2022; 12:biom12121902. [PMID: 36551330 PMCID: PMC9775183 DOI: 10.3390/biom12121902] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer metabolic reprogramming is essential for maintaining cancer cell survival and rapid replication. A common target of this metabolic reprogramming is one-carbon metabolism which is notable for its function in DNA synthesis, protein and DNA methylation, and antioxidant production. Polyamines are a key output of one-carbon metabolism with widespread effects on gene expression and signaling. As a result of these functions, one-carbon and polyamine metabolism have recently drawn a lot of interest for their part in cancer malignancy. Therapeutic inhibitors that target one-carbon and polyamine metabolism have thus been trialed as anticancer medications. The significance and future possibilities of one-carbon and polyamine metabolism as a target in cancer therapy are discussed in this review.
Collapse
Affiliation(s)
- Anowarul Islam
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Zeeshan Shaukat
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Rashid Hussain
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Stephen L. Gregory
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Correspondence: ; Tel.: +61-0466987583
| |
Collapse
|
8
|
Wang X, Lv Z, Xia H, Guo X, Wang J, Wang J, Liu M. Biochemical recurrence related metabolic novel signature associates with immunity and ADT treatment responses in prostate cancer. Cancer Med 2022; 12:862-878. [PMID: 35681277 PMCID: PMC9844602 DOI: 10.1002/cam4.4856] [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: 11/17/2021] [Revised: 05/11/2022] [Accepted: 05/15/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a unique cancer from a metabolic perspective. Androgen receptor assumes a vital part in normal and malignant prostate cells regarding almost all aspects of cell metabolism, such as glucose, fat, amino acids, nucleotides, and so on. METHODS We used The Cancer Genome Atlas database as training set, Memorial Sloan-Kettering Cancer Center cohort as validation set, and Gene Expression Omnibus database (GSE70769) as test set to identify the optimal prognostic signature. We evaluated the signature in terms of biochemical progression-free survival (bPFS), ROC curve, clinicopathological features, independent prognostic indicators, tumor microenvironment, and infiltrating immune cells. Nomogram was built dependent on the results of cox regression analyses. GSEA algorithm was used to evaluate differences in metabolism. The signature's prediction of androgen deprivation therapy (ADT) response was validated based on two groups of basic cytological experiments treat with ADT (GSE143408 and GSE120343) and the transcriptional information of pre-ADT/post-ADT of six local PCa patients. RESULTS We finally input four screened genes into the stepwise regression model to construct metabolism-related signature. The signature shows good prediction performance in training set, verification set, and test set. A nomogram based on the PSA, Gleason score, T staging, and the signature risk score could predict 1-, 3-, and 5-year bPFS with the high area under curve values. Based on gene-set enrichment analysis, the characteristics of four genes signature could influence some important metabolic biological processes of PCa and were serendipitously found to be significantly related to androgen response. Subsequently, two cytological experimental data sets and our local patient sequencing data set verified that the signature may be helpful to evaluate the therapeutic response of PCa to ADT. CONCLUSIONS Our systematic study definite a metabolism-related gene signature to foresee prognosis of PCa patients which might add to individual prevention and treatment.
Collapse
Affiliation(s)
- Xuan Wang
- Department of UrologyBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingPeople's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Zhengtong Lv
- Department of UrologyBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingPeople's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Haoran Xia
- Department of UrologyBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingPeople's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Xiaoxiao Guo
- Department of UrologyBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingPeople's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jianye Wang
- Department of UrologyBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingPeople's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jianlong Wang
- Department of UrologyBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingPeople's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Ming Liu
- Department of UrologyBeijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingPeople's Republic of China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical SciencesBeijingPeople's Republic of China
| |
Collapse
|
9
|
Zhu DD, Yu XB, Jiang W, Zhu Y. Pathological stage-associated non-coding RNA long intergenic non-protein coding RNA 1234 (LINC01234) participation in cell cycle regulation in adrenocortical carcinoma through bromodomain-containing protein 4 (BRD4) expression mediation via sponging microRNA (miR)-140-3p. Bioengineered 2022; 13:13607-13621. [PMID: 35765893 PMCID: PMC9275903 DOI: 10.1080/21655979.2022.2081464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Many researches indicated that long non-coding RNAs (lncRNAs) were involved in the malignant progression of tumors, including Adrenocortical Carcinoma (ACC). However, as for most lncRNAs, their biological behaviors and molecular mechanism remain unclear in ACC. In the present research, weighted gene co-expression network analysis (WGCNA) was used to identify pathologically relevant gene, including lncRNAs. By comparing their expressions in GSE61359 tumors and normal controls, long intergenic non-protein coding RNA 1234 (LINC01234) was selected to investigate the clinical significance, biological function, and mechanism in ACC. Data mining revealed that LINC01234 expression was significantly up-regulated in ACC patients, and a shorter survival time presents in patients with higher LINC01234 expression compared to that in patients with lower LINC01234 expression. Further, LINC01234 silencing resulted in cells growth arrest in vitro and in vivo. Mechanism studies suggested that LINC01234 silencing induced cell cycle arrest, and bromodomain-containing protein 4 (BRD4) overexpression could restore this phenomenon. Further research showed that LINC01234 could mediate BRD4 expression through competitively sequestering microRNA (miR)-140-3p, as evidenced by the positive correlation of LINC01234 with BRD4 and inverse correlation with miR-140-3p expression. Luciferase activity assay also verified the targeting relationship between LINC01234, BRD4 and miR-140-3p. And up-regulated LINC01234 in ACC cells significantly reversed the degradation of BRD4 by miR-140-3p. Collectively, we deduce that LINC01234 functions as a ceRNA to regulate BRD4 expression by sponging miR-140-3p in ACC progress. Our findings have the potential to provide a new target for the diagnosis and treatment of ACC.
Collapse
Affiliation(s)
- Dan-Dan Zhu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Huangpu, Shanghai, China
| | - Xin-Bo Yu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Huangpu, Shanghai, China
| | - Wen Jiang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Huangpu, Shanghai, China
| | - Yu Zhu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Huangpu, Shanghai, China
| |
Collapse
|
10
|
Sial N, Rehman J, Saeed S, Ahmad M, Hameed Y, Atif M, Rehman A, Asif R, Ahmed H, Hussain M, Khan M, Ambreen A, Ambreen A. Integrative analysis reveals methylenetetrahydrofolate dehydrogenase 1-like as an independent shared diagnostic and prognostic biomarker in five different human cancers. Biosci Rep 2022; 42:BSR20211783. [PMID: 34908119 PMCID: PMC8738869 DOI: 10.1042/bsr20211783] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/12/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Defects in methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) expression have earlier been examined in only a few human cancers. OBJECTIVES Multi-omics profiling of MTHFD1L as a shared biomarker in distinct subtypes of human cancers. METHODS In the current study, for the multi-omics analysis of MTHFD1L in 24 major subtypes of human cancers, a comprehensive in silico approach was adopted to mine different open access online databases including UALCAN, Kaplan-Meier (KM) plotter, LOGpc, GEPIA, Human Protein Atlas (HPA), Gene Expression across Normal and Tumor tissue (GENT2), MEXPRESS, cBioportal, STRING, DAVID, TIMER, and Comparative Toxicogenomics Database (CTD). RESULTS We noticed that the expression of MTHFD1L was significantly higher in all the analyzed 24 subtypes of human cancers as compared with the normal controls. Moreover, MTHDF1L overexpression was also found to be significantly associated with the reduced overall survival (OS) duration of Bladder urothelial cancer (BLCA), Head and neck cancer (HNSC), Kidney renal papillary cell carcinoma (KIRP), Lung adenocarcinoma (LUAD), and Uterine corpus endometrial carcinoma (UCEC). This implies that MTHFD1L plays a significant role in the development and progression of these cancers. We further noticed that MTHFD1L was also overexpressed in BLCA, HNSC, KIRP, LUAD, and UCEC patients of different clinicopathological features. Pathways enrichment analysis revealed the involvement of MTHFD1L-associated genes in five diverse pathways. We also explored few interesting correlations between MTHFD1L expression and its promoter methylation, genetic alterations, CNVs, and between CD8+ T immune cells level. CONCLUSION In conclusion, our results elucidated that MTHFD1L can serve as a shared diagnostic and prognostic biomarker in BLCA, HNSC, KIRP, LUAD, and UCEC patients of different clinicopathological features.
Collapse
Affiliation(s)
- Nuzhat Sial
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Jalil Ur Rehman
- Department of Eastern Medicine, Qarshi University, Lahore, Pakistan
- University College of Conventional Medicine, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Saba Saeed
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Mukhtiar Ahmad
- Department of Biochemistry and Biotechnology, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Yasir Hameed
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Atif
- University College of Conventional Medicine, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Abdul Rehman
- Department of Eastern Medicine, Qarshi University, Lahore, Pakistan
| | - Rizwan Asif
- Department of Microbiology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Hamad Ahmed
- Department of Eastern Medicine, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Safdar Hussain
- Department of Biochemistry and Biotechnology, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Rashid Khan
- University College of Eastern Medicine, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Atifa Ambreen
- Allied Department, The Sahara College, Narowal, Pakistan
| | - Ayesha Ambreen
- Allied Department, The Sahara College, Narowal, Pakistan
| |
Collapse
|
11
|
Quevedo-Ocampo J, Escobedo-Calvario A, Souza-Arroyo V, Miranda-Labra RU, Bucio-Ortiz L, Gutiérrez-Ruiz MC, Chávez-Rodríguez L, Gomez-Quiroz LE. Folate Metabolism in Hepatocellular Carcinoma. What Do We Know So Far? Technol Cancer Res Treat 2022; 21:15330338221144446. [PMID: 36503290 DOI: 10.1177/15330338221144446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer cells are characterized by accelerated proliferation and an outstanding adaptation of their metabolic pathways to meet energy demands. The folate cycle, also known as folate metabolism or one-carbon metabolism, through enzymatic interconversions, provides metabolites necessary for nucleotide synthesis, methylation, and reduction power, helping to maintain the high rate of proliferation; therefore, the study of this metabolic pathway is of great importance in the study of cancer. Moreover, multiple enzymes involved in this cycle have been implicated in different types of cancer, corroborating the cell's adaptations under this pathology. During the last decade, nonalcoholic fatty liver disease has emerged as the leading etiology related to the rise in the incidence and deaths of hepatocellular carcinoma. Specifically, cholesterol accumulation has been a determinant promoter of tumor formation, with solid evidence that an enriched-cholesterol diet plays a crucial role in accelerating the development of an aggressive subtype of hepatocellular carcinoma compared to other models. In this review, we will discuss the most recent findings to understand the contribution of folate metabolism to cancer cells and tumor microenvironment while creating a link between the dynamics given by cholesterol and methylenetetrahydrofolate dehydrogenase 1-like, a key enzyme of the cycle located in the mitochondrial compartment.
Collapse
Affiliation(s)
- Jaqueline Quevedo-Ocampo
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Alejandro Escobedo-Calvario
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Verónica Souza-Arroyo
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Roxana U Miranda-Labra
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Leticia Bucio-Ortiz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - María C Gutiérrez-Ruiz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Lisette Chávez-Rodríguez
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Luis E Gomez-Quiroz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
12
|
Cui L, Zhao X, Jin Z, Wang H, Yang SF, Hu S. Melatonin modulates metabolic remodeling in HNSCC by suppressing MTHFD1L-formate axis. J Pineal Res 2021; 71:e12767. [PMID: 34533844 DOI: 10.1111/jpi.12767] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/05/2021] [Accepted: 09/14/2021] [Indexed: 12/26/2022]
Abstract
Metabolic remodeling is now widely recognized as a hallmark of cancer, yet its role in head and neck squamous cell carcinoma (HNSCC) remains largely unknown. In this study, metabolomic analysis of melatonin-treated HNSCC cell lines revealed that exogenous melatonin inhibited many important metabolic pathways including folate cycle in HNSCC cells. Methylenetetrahydrofolate dehydrogenase 1 like (MTHFD1L), a metabolic enzyme of the folate cycle regulating the production of formate, was identified as a downstream target of melatonin. MTHFD1L was found to be markedly upregulated in HNSCC, and MTHFD1L overexpression was significantly associated with unfavorable clinical outcome of HNSCC patients. In addition, MTHFD1L promoted HNSCC progression in vitro and in vivo and reversed the oncostatic effects of exogenous melatonin. More importantly, the malignant phenotypes suppressed by knockdown of MTHFD1L or exogenous melatonin could be partially rescued by formate. Furthermore, we found that melatonin inhibited the expression of MTHFD1L in HNSCC cells through the downregulation of cyclic AMP-responsive element-binding protein 1 (CREB1) phosphorylation. Lastly, this novel regulatory axis of melatonin-p-CREB1-MTHFD1L-formate was also verified in HNSCC tissues. Collectively, our findings have demonstrated that MTHFD1L-formate axis promotes HNSCC progression and melatonin inhibits HNSCC progression through CREB1-mediated downregulation of MTHFD1L and formate. These findings have revealed new metabolic mechanisms in HNSCC and may provide novel insights on the therapeutic intervention of HNSCC.
Collapse
Affiliation(s)
- Li Cui
- School of Dentistry, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| | - Xinyuan Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Zhenning Jin
- School of Dentistry, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shen Hu
- School of Dentistry, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|
13
|
Wang J, Wang K, Liang J, Jin J, Wang X, Yan S. Chitosan-tripolyphosphate nanoparticles-mediated co-delivery of MTHFD1L shRNA and 5-aminolevulinic acid for combination photodynamic-gene therapy in oral cancer. Photodiagnosis Photodyn Ther 2021; 36:102581. [PMID: 34648994 DOI: 10.1016/j.pdpdt.2021.102581] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/29/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Rationally designed nanostructured materials can produce improved drug carriers that play an increasingly important role in cancer treatment. In comparison with conventional drug combination approaches, using co-delivery systems of multiple drugs achieves sophisticated targeting strategies and multifunctionality. METHODS First, a nano-co-delivery of chitosan/tripolyphosphate (CS-TPP) was synthesized and characterized combining 5-aminolevulinic acid photodynamic therapy (ALA-PDT) with methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) shRNA. In this report, we investigated the efficacy of the simultaneous delivery of shRNA/photosensitizer on the gene expression of oral squamous cell carcinoma (OSCC) cells. The efficacy of CS-TPP-(shMTHFD1L-ALA)-PDT in inducing apoptosis and in generating of reactive oxygen species (ROS) in vitro was then assessed by Annexin V-PI and DCFH-DA assays respectively. In vivo therapeutic experiments were conducted in well-established orthotopic animal models of HNSCC. RESULTS The results showed that the CS-TPP-(shMTHFD1L-ALA) nanoparticles (NPs) were approximately 145 nm in size. The cytotoxicity of OSCC cells was significantly increased by co-delivery of MTHFD1L shRNA and ALA-PDT compared with other groups. Furthermore, individual and combined therapies revealed remarkable pro-apoptotic, ROS and anti-tumorigenesis effects, and CS-TPP-(shMTHFD1L-ALA)-PDT had additive effects in vitro and in vivo. CONCLUSION These observations indicate that CS-TPP-(shMTHFD1L-ALA) NPs may be an ideal candidate for gene/photosensitizer delivery.
Collapse
Affiliation(s)
- Jian Wang
- Department of Stomatology, PLA Strategic Support Force Medical Center, Beijing, 100101, China
| | - Ke Wang
- Department of Stomatology, PLA Strategic Support Force Medical Center, Beijing, 100101, China
| | - Jin Liang
- Department of Stomatology, PLA Strategic Support Force Medical Center, Beijing, 100101, China
| | - Jianqiu Jin
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100100, China
| | - Xing Wang
- Foshan (Southern China) Institute for New Materials, Foshan, 528220, China.
| | - Shu Yan
- Department of Stomatology, PLA Strategic Support Force Medical Center, Beijing, 100101, China; PLA 306 Clinical College of Anhui Medical University, Hefei, 230001, China.
| |
Collapse
|
14
|
Integrated bioinformatics analysis identified MTHFD1L as a potential biomarker and correlated with immune infiltrates in hepatocellular carcinoma. Biosci Rep 2021; 41:227681. [PMID: 33605411 PMCID: PMC7897918 DOI: 10.1042/bsr20202063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
Liver hepatocellular carcinoma (LIHC) is one of the most frequently occurring primary malignant liver tumors and seriously harms people’s health in the world. Methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) has been shown to be associated with colon cancer cell proliferation, colony formation and invasion. In the present study, a total of 370 LIHC and 51 normal samples data were downloaded from The Cancer Genome Atlas (TCGA) database. Bioinformatics and immunohistochemistry (IHC) analysis showed that MTHFD1L is highly expressed in liver tumors. Correlation analysis suggested the differences of vital status between high- and low-expression MTHFD1L groups of LIHC. Univariate and multivariate Cox proportional hazards regression were performed to identify the relationship between clinical characteristics and overall survival (OS). In addition, to explore whether MTHFD1L has an effect on the immune infiltration of LIHC. The correlation between MTHFD1L expression and 24 immune cells were analyzed by ImmuneCellAI database. Furthermore, we combined three databases CIBERSORT, TIMER and ImmuneCellAI to do a comprehensive validation and determined that dendritic cells (DCs) resting, macrophage M0 and macrophage M2 closely related to the expression of MTHFD1L. The results showed that MTHFD1L was a potential prognostic biomarker for LIHC, and could help to elucidate that how the immune microenvironment promotes liver cancer development.
Collapse
|
15
|
Yang C, Zhang J, Liao M, Yang Y, Wang Y, Yuan Y, Ouyang L. Folate-mediated one-carbon metabolism: a targeting strategy in cancer therapy. Drug Discov Today 2020; 26:817-825. [PMID: 33316375 DOI: 10.1016/j.drudis.2020.12.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/22/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023]
Abstract
Folate-mediated one-carbon metabolism (FOCM) supports vital events for the growth and survival of proliferating cells. Nucleotide synthesis and DNA methylation are the biochemical bases of cancers that are highly dependent on FOCM. Recent studies revealed that FOCM is connected with redox homeostasis and epigenetics in cancer. Furthermore, folate-metabolizing enzymes, such as serine hydroxymethyltransferase 2 (SHMT2) and methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), are associated with the development of cancers, including breast cancer, highlighting their potential application in tumor-targeted therapy. Therefore, targeting metabolizing enzymes, especially SHMT2 and MTHFD2, provides a novel strategy for cancer treatment. In this review, we outline current understanding of the functions of SHMT2 and MTHFD2, discussing their expression, potential functions, and regulatory mechanism in cancers. Furthermore, we discuss examples of inhibitors of SHMT2 and MTHFD2.
Collapse
Affiliation(s)
- Chengcan Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, China
| | - Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, China
| | - Yushang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, China; Department of Thoracic Surgery, West China Hospital, Sichuan University, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, China; Department of Respiratory and Critical Care Medicine, West China Medical School/West China Hospital, Sichuan University, China.
| | - Yong Yuan
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, China; Department of Thoracic Surgery, West China Hospital, Sichuan University, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, China; The Research Units of West China, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
16
|
Wang L, Yang Y, Wang XM, Wang CQ, Zhang YM, Li BL. MTHFD1L as a folate cycle enzyme correlates with prognostic outcome and its knockdown impairs cell invasive behaviors in osteosarcoma via mediating the AKT/mTOR pathway. J Recept Signal Transduct Res 2020; 40:584-590. [PMID: 32456526 DOI: 10.1080/10799893.2020.1769658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Osteosarcoma (OS) is the most frequent primary malignancy initially in bone with multiple genomic aberrations. Methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) is linked with the progression of diverse tumors. However, its function in OS is not understood completely. The expression pattern and prognostic significance of MTHFD1L in OS tissues were analyzed based on GEO database. The expression level of MTHFD1L in OS cell lines was explored by qRT-PCR. The cell proliferation, colony formation ability, invasion as well as migration in OS cells after MTHFD1L knockdown were determined using cell counting kit 8 (CCK-8) assay, colony formation and transwell methods. GSEA analysis was performed to predict the underlying mechanisms of MTHFD1L in OS development. Furthermore, the western blot was utilized to study the influence of MTHFD1L on AKT/mTOR pathway. Our results indicated that MTHFD1L expression was significantly up-regulated in OS tissues and cells compared with normal tissues and cells. High expression of MTHFD1L could lead to poor prognosis of OS patients. Cell proliferation, colony formation ability, migration and invasion were blocked because of reduced MTHFD1L in vitro. Moreover, cell cycle and AKT/mTOR pathway were all associated with MTHFD1L expression. In conclusion, the findings revealed that MTHFD1L might promote the development of OS via mediating cell cycle and AKT/mTOR pathway, indicating that MTHFD1L might act as a promising therapeutic target for OS treatment.
Collapse
Affiliation(s)
- Lei Wang
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Ya Yang
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Xiu-Mei Wang
- Electroencephalogram Room, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Cheng-Qun Wang
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Yuan-Min Zhang
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, PR China
| | - Bing-Liang Li
- Department of Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, PR China
| |
Collapse
|