1
|
Ma C, Xu A, Zuo L, Li Q, Fan F, Hu Y, Sun C. Methionine Dependency and Restriction in Cancer: Exploring the Pathogenic Function and Therapeutic Potential. Pharmaceuticals (Basel) 2025; 18:640. [PMID: 40430461 PMCID: PMC12114517 DOI: 10.3390/ph18050640] [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: 03/24/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Methionine, an essential amino acid, is obtained by dietary intake to fulfill the requirements of our bodies. Accumulating evidence indicates that methionine plays a pivotal role in various biological processes, including protein synthesis, energy metabolism, redox balance maintenance, and methylation modifications. Numerous advances underscore the heightened dependence of cancer cells on methionine, which is a significant factor in cancer pathogenesis and development. A profound comprehension of the intricate relationship between methionine metabolism and tumorigenesis is imperative for advancing the field of cancer therapeutics. Herein, we delve into the role of methionine in supporting cancer growth, the impact on epigenetic modifications, and the interaction between methionine and the tumor microenvironment. Additionally, we provide insights into the development of various methionine-targeted therapy strategies. This paper summarizes the current state of research and its translational potential, emphasizing the challenges and opportunities associated with harnessing methionine dependence as a target for innovative cancer treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chunyan Sun
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (C.M.); (A.X.); (L.Z.); (Q.L.); (F.F.); (Y.H.)
| |
Collapse
|
2
|
Gebrehiwot NT, Liu Y, Li J, Liu HM. Molecular Alterations in Gastric Intestinal Metaplasia Shed Light on Alteration of Methionine Metabolism: Insight into New Diagnostic and Treatment Approaches. Biomedicines 2025; 13:964. [PMID: 40299656 PMCID: PMC12025106 DOI: 10.3390/biomedicines13040964] [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: 02/23/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025] Open
Abstract
Gastric intestinal metaplasia (GIM) is a precancerous lesion and the key risk factor in the development of gastric cancer (GC), but early detection and treatment remain challenging. The traditional endoscopic diagnosis of metaplastic lesions is complicated by an increased rate of inappropriateness and false negativity. Although early interventions with H. pylori eradication, as well as endoscopic therapy results, were promising, there is still a significant unmet need to control GIM progression and recurrences. Molecular alterations, such as an increased DNA methylation index, have been identified as a crucial factor in the downregulation of tumor suppressor genes, such as the caudal-type homeobox (CDX2) gene, which regulates epithelial cell proliferation and GIM progression and is associated with treatment failure. CDX2 is downregulated by promoter hypermethylation in the colonic-type epithelium, in which the methylation was correlated with reduced intake of dietary folate sources. Tumor cells alter to dietary methionine sources in the biosynthesis of S-Adenosylmethionine, a universal methyl donor for transmethylation, under the conditions of limited folate and B12 availability. The gut microbiota also exhibited a shift in microbial composition, which could influence the host's dietary methionine metabolism. Meanwhile, activated oncogenic signaling via the PI3K/Akt/mTORC1/c-MYC pathway could promotes rewiring dietary methionine and cellular proliferation. Tumor methionine dependence is a metabolic phenotype that could be helpful in predictive screening of tumorigenesis and as a target for preventive therapy to enhance precision oncology. This review aimed to discuss the molecular alterations in GIM to shed light on the alteration of methionine metabolism, with insight into new diagnostic and treatment approaches and future research directions.
Collapse
Affiliation(s)
- Nigatu Tadesse Gebrehiwot
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou 450001, China;
- Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Ministry of Education, Zhengzhou 450001, China
| | - Ying Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China;
| | - Juan Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou 450001, China;
- Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Ministry of Education, Zhengzhou 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou 450001, China;
- Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Ministry of Education, Zhengzhou 450001, China
| |
Collapse
|
3
|
Church MC, Workman JL. The SWI/SNF chromatin remodeling complex: a critical regulator of metabolism. Biochem Soc Trans 2024; 52:1327-1337. [PMID: 38666605 PMCID: PMC11346436 DOI: 10.1042/bst20231141] [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: 02/06/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 06/27/2024]
Abstract
The close relationship between chromatin and metabolism has been well-studied in recent years. Many metabolites have been found to be cofactors used to modify chromatin, and these modifications can in turn affect gene transcription. One chromatin-associated factor responsible for regulating transcription is the SWI/SNF complex, an ATP-dependent chromatin remodeler conserved throughout eukaryotes. SWI/SNF was originally described in yeast as regulating genes involved in carbon source metabolism and mating type switching, and its mammalian counterpart has been extensively studied for its role in diseases such as cancer. The yeast SWI/SNF complex is closely associated with activation of stress response genes, many of which have metabolic functions. It is now recognized that this is a conserved function of the complex, and recent work has shown that mammalian SWI/SNF is also a key regulator of metabolic transcription. Emerging evidence suggests that loss of SWI/SNF introduces vulnerabilities to cells due to this metabolic influence, and that this may present opportunities for treatment of SWI/SNF-deficient cancers.
Collapse
Affiliation(s)
- Michael C. Church
- Stowers Institute of Medical Research, 1000 E 50th Street, Kansas City, MO 64118, U.S.A
| | - Jerry L. Workman
- Stowers Institute of Medical Research, 1000 E 50th Street, Kansas City, MO 64118, U.S.A
| |
Collapse
|
4
|
Austad SN, Smith JR, Hoffman JM. Amino acid restriction, aging, and longevity: an update. FRONTIERS IN AGING 2024; 5:1393216. [PMID: 38757144 PMCID: PMC11096585 DOI: 10.3389/fragi.2024.1393216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024]
Abstract
Various so-called dietary restriction paradigms have shown promise for extending health and life. All such paradigms rely on ad libitum (hereafter ad lib) feeding, something virtually never employed in animals whose long-term health we value, either as a control or, except for food restriction itself, for both control and treatment arms of the experiment. Even though the mechanism(s) remain only vaguely understood, compared to ad lib-fed animals a host of dietary manipulations, including calorie restriction, low protein, methionine, branched-chain amino acids, and even low isoleucine have demonstrable health benefits in laboratory species in a standard laboratory environment. The remaining challenge is to determine whether these health benefits remain in more realistic environments and how they interact with other health enhancing treatments such as exercise or emerging geroprotective drugs. Here we review the current state of the field of amino acid restriction on longevity of animal models and evaluate its translational potential.
Collapse
Affiliation(s)
- S. N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - J. R. Smith
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - J. M. Hoffman
- Department of Biological Sciences, Augusta University, Augusta, GA, United States
| |
Collapse
|
5
|
Shi W, Cheng Y, Zhu H, Zhao L. Metabolomics and lipidomics in non-small cell lung cancer. Clin Chim Acta 2024; 555:117823. [PMID: 38325713 DOI: 10.1016/j.cca.2024.117823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Due to its insidious nature, lung cancer remains a leading cause of cancer-related deaths worldwide. Therefore, there is an urgent need to identify sensitive/specific biomarkers for early diagnosis and monitoring. The current study was designed to provide a current metabolic profile of non-small cell lung cancer (NSCLC) by systematically reviewing and summarizing various metabolomic/ lipidomic studies based on NSCLC blood samples, attempting to find biomarkers in human blood that can predict or diagnose NSCLC, and investigating the involvement of key metabolites in the pathogenesis of NSCLC. We searched all articles on lung cancer published in Elsevier, PubMed, Web of Science and the Cochrane Library between January 2012 and December 2022. After critical selection, a total of 31 studies (including 2768 NSCLC patients and 9873 healthy individuals) met the inclusion criteria, and 22 were classified as "high quality". Forty-six metabolites related to NSCLC were repeatedly identified, involving glucose metabolism, amino acid metabolism, lipid metabolism and nucleotide metabolism. Pyruvic acid, carnitine, phenylalanine, isoleucine, kynurenine and 3-hydroxybutyrate showed upward trends in all studies, citric acid, glycine, threonine, cystine, alanine, histidine, inosine, betaine and arachidic acid showed downward trends in all studies. This review summarizes the existing metabolomic/lipidomic studies related to the identification of blood biomarkers in NSCLC, examines the role of key metabolites in the pathogenesis of NSCLC, and provides an important reference for the clinical diagnosis and treatment of NSCLC. Due to the limited size and design heterogeneity of the existing studies, there is an urgent need for standardization of future studies, while validating existing findings with more studies.
Collapse
Affiliation(s)
- Wei Shi
- Shenyang Pharmaceutical University, 103 Wenhua Road Shenhe District, 110016 Shenyang, Liaoning Province, PR China
| | - Yizhen Cheng
- Shenyang Pharmaceutical University, 103 Wenhua Road Shenhe District, 110016 Shenyang, Liaoning Province, PR China
| | - Haihua Zhu
- Betta Pharmaceuticals Co., Ltd, 24 Wuzhou Road Yuhang Economic and Technological Development Area, Hangzhou, Zhejiang Province, PR China
| | - Longshan Zhao
- Shenyang Pharmaceutical University, 103 Wenhua Road Shenhe District, 110016 Shenyang, Liaoning Province, PR China.
| |
Collapse
|
6
|
Church MC, Price A, Li H, Workman JL. The Swi-Snf chromatin remodeling complex mediates gene repression through metabolic control. Nucleic Acids Res 2023; 51:10278-10291. [PMID: 37650639 PMCID: PMC10602859 DOI: 10.1093/nar/gkad711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 08/02/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
In eukaryotes, ATP-dependent chromatin remodelers regulate gene expression in response to nutritional and metabolic stimuli. However, altered transcription of metabolic genes may have significant indirect consequences which are currently poorly understood. In this study, we use genetic and molecular approaches to uncover a role for the remodeler Swi-Snf as a critical regulator of metabolism. We find that snfΔ mutants display a cysteine-deficient phenotype, despite growth in nutrient-rich media. This correlates with widespread perturbations in sulfur metabolic gene transcription, including global redistribution of the sulfur-sensing transcription factor Met4. Our findings show how a chromatin remodeler can have a significant impact on a whole metabolic pathway by directly regulating an important gene subset and demonstrate an emerging role for chromatin remodeling complexes as decisive factors in metabolic control.
Collapse
Affiliation(s)
- Michael C Church
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Andrew Price
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Jerry L Workman
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| |
Collapse
|
7
|
Alapati S, Fortuna G, Ramage G, Delaney C. Evaluation of Metabolomics as Diagnostic Targets in Oral Squamous Cell Carcinoma: A Systematic Review. Metabolites 2023; 13:890. [PMID: 37623834 PMCID: PMC10456490 DOI: 10.3390/metabo13080890] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
In recent years, high-throughput technologies have facilitated the widespread use of metabolomics to identify biomarkers and targets for oral squamous cell carcinoma (OSCC). As a result, the primary goal of this systematic review is to identify and evaluate metabolite biomarkers and their pathways for OSCC that featured consistently across studies despite methodological variations. Six electronic databases (Medline, Cochrane, Web of Science, CINAHL, ProQuest, and Embase) were reviewed for the longitudinal studies involving OSCC patients and metabolic marker analysis (in accordance with PRISMA 2020). The studies included ranged from the inception of metabolomics in OSCC (i.e., 1 January 2007) to 30 April 2023. The included studies were then assessed for their quality using the modified version of NIH quality assessment tool and QUADOMICS. Thirteen studies were included after screening 2285 studies. The majority of the studies were from South Asian regions, and metabolites were most frequently derived from saliva. Amino acids accounted for more than quarter of the detected metabolites, with glutamate and methionine being the most prominent. The top dysregulated metabolites indicated dysregulation of six significantly enriched pathways including aminoacyl-tRNA biosynthesis, glutathione metabolism and arginine biosynthesis with the false discovery rate (FDR) <0.05. Finally, this review highlights the potential of metabolomics for early diagnosis and therapeutic targeting of OSCC. However, larger studies and standardized protocols are needed to validate these findings and make them a clinical reality.
Collapse
Affiliation(s)
- Susanth Alapati
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, University of Glasgow, 378 Sauchiehall Street, Glasgow G2 3JZ, UK; (S.A.)
| | - Giulio Fortuna
- Department of Oral Medicine, Glasgow Dental School, School of Medicine, Dentistry and Nursing, University of Glasgow, 378 Sauchiehall Street, Glasgow G2 3JZ, UK
| | - Gordon Ramage
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, University of Glasgow, 378 Sauchiehall Street, Glasgow G2 3JZ, UK; (S.A.)
| | - Christopher Delaney
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, University of Glasgow, 378 Sauchiehall Street, Glasgow G2 3JZ, UK; (S.A.)
| |
Collapse
|
8
|
Bort A, G. Sánchez B, León C, Nozal L, Mora-Rodríguez JM, Castro F, Crego AL, Díaz-Laviada I. Metabolic fingerprinting of chemotherapy-resistant prostate cancer stem cells. An untargeted metabolomic approach by liquid chromatography-mass spectrometry. Front Cell Dev Biol 2022; 10:1005675. [PMID: 36325358 PMCID: PMC9618794 DOI: 10.3389/fcell.2022.1005675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Chemoresistance is one of the most important challenges in cancer therapy. The presence of cancer stem cells within the tumor may contribute to chemotherapy resistance since these cells express high levels of extrusion pumps and xenobiotic metabolizing enzymes that inactivate the therapeutic drug. Despite the recent advances in cancer cell metabolism adaptations, little is known about the metabolic adaptations of the cancer stem cells resistant to chemotherapy. In this study, we have undertaken an untargeted metabolomic analysis by liquid chromatography–high-resolution spectrometry combined with cytotoxicity assay, western blot, quantitative real-time polymerase chain reaction (qPCR), and fatty acid oxidation in a prostate cancer cell line resistant to the antiandrogen 2-hydroxiflutamide with features of cancer stem cells, compared to its parental androgen-sensitive cell line. Metabolic fingerprinting revealed 106 out of the 850 metabolites in ESI+ and 67 out of 446 in ESI- with significant differences between the sensitive and the resistant cell lines. Pathway analysis performed with the unequivocally identified metabolites, revealed changes in pathways involved in energy metabolism as well as posttranscriptional regulation. Validation by enzyme expression analysis indicated that the chemotherapy-resistant prostate cancer stem cells were metabolically dormant with decreased fatty acid oxidation, methionine metabolism and ADP-ribosylation. Our results shed light on the pathways underlying the entry of cancer cells into dormancy that might contribute to the mechanisms of drug resistance.
Collapse
Affiliation(s)
- Alicia Bort
- Yale University School of Medicine, Vascular Biology and Therapeutics Program, New Haven, CT, United states
| | - Belén G. Sánchez
- Alcala University, School of Medicine, Department of Systems Biology and Research Institute in Chemistry “Andrés M. Del Río” (IQAR), Madrid, Spain
| | - Carlos León
- Carlos III University, Department of Bioengineering and Aerospatial Engineering, Madrid, Spain
| | - Leonor Nozal
- Alcala University and General Foundation of Alcalá University, Center of Applied Chemistry and Biotechnology, Madrid, Spain
| | - José M. Mora-Rodríguez
- Alcala University, School of Medicine, Department of Systems Biology and Research Institute in Chemistry “Andrés M. Del Río” (IQAR), Madrid, Spain
| | - Florentina Castro
- Alcala University and General Foundation of Alcalá University, Center of Applied Chemistry and Biotechnology, Madrid, Spain
| | - Antonio L. Crego
- Alcala University, Department of Analytical Chemistry, Physical Chemistry and Chemical Engineering, Madrid, Spain
- *Correspondence: Antonio L. Crego, ; Inés Díaz-Laviada,
| | - Inés Díaz-Laviada
- Alcala University, School of Medicine, Department of Systems Biology and Research Institute in Chemistry “Andrés M. Del Río” (IQAR), Madrid, Spain
- *Correspondence: Antonio L. Crego, ; Inés Díaz-Laviada,
| |
Collapse
|
9
|
Guo R, Liang JH, Zhang Y, Lutchenkov M, Li Z, Wang Y, Trujillo-Alonso V, Puri R, Giulino-Roth L, Gewurz BE. Methionine metabolism controls the B cell EBV epigenome and viral latency. Cell Metab 2022; 34:1280-1297.e9. [PMID: 36070681 PMCID: PMC9482757 DOI: 10.1016/j.cmet.2022.08.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022]
Abstract
Epstein-Barr virus (EBV) subverts host epigenetic pathways to switch between viral latency programs, colonize the B cell compartment, and reactivate. Within memory B cells, the reservoir for lifelong infection, EBV genomic DNA and histone methylation marks restrict gene expression. But this epigenetic strategy also enables EBV-infected tumors, including Burkitt lymphomas, to evade immune detection. Little is known about host cell metabolic pathways that support EBV epigenome landscapes. We therefore used amino acid restriction, metabolomic, and CRISPR approaches to identify that an abundant methionine supply and interconnecting methionine and folate cycles maintain Burkitt EBV gene silencing. Methionine restriction, or methionine cycle perturbation, hypomethylated EBV genomes and de-repressed latent membrane protein and lytic gene expression. Methionine metabolism also shaped EBV latency gene regulation required for B cell immortalization. Dietary methionine restriction altered murine Burkitt xenograft metabolomes and de-repressed EBV immunogens in vivo. These results highlight epigenetic/immunometabolism crosstalk supporting the EBV B cell life cycle and suggest therapeutic approaches.
Collapse
Affiliation(s)
- Rui Guo
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Jin Hua Liang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Yuchen Zhang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Michael Lutchenkov
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Zhixuan Li
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Yin Wang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Vicenta Trujillo-Alonso
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Rishi Puri
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Lisa Giulino-Roth
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Benjamin E Gewurz
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Harvard Program in Virology, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Junior RP, Sonehara NM, Jardim-Perassi BV, Pal A, Asad Y, Almeida Chuffa LG, Chammas R, Raynaud FI, Zuccari DAPC. Presence of human breast cancer xenograft changes the diurnal profile of amino acids in mice. Sci Rep 2022; 12:1008. [PMID: 35046467 PMCID: PMC8770691 DOI: 10.1038/s41598-022-04994-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 01/04/2022] [Indexed: 12/25/2022] Open
Abstract
Human xenografts are extremely useful models to study the biology of human cancers and the effects of novel potential therapies. Deregulation of metabolism, including changes in amino acids (AAs), is a common characteristic of many human neoplasms. Plasma AAs undergo daily variations, driven by circadian endogenous and exogenous factors. We compared AAs concentration in triple negative breast cancer MDA-MB-231 cells and MCF10A non-tumorigenic immortalized breast epithelial cells. We also measured plasma AAs in mice bearing xenograft MDA-MB-231 and compared their levels with non-tumor-bearing control animals over 24 h. In vitro studies revealed that most of AAs were significantly different in MDA-MB-231 cells when compared with MCF10A. Plasma concentrations of 15 AAs were higher in cancer cells, two were lower and four were observed to shift across 24 h. In the in vivo setting, analysis showed that 12 out of 20 AAs varied significantly between tumor-bearing and non-tumor bearing mice. Noticeably, these metabolites peaked in the dark phase in non-tumor bearing mice, which corresponds to the active time of these animals. Conversely, in tumor-bearing mice, the peak time occurred during the light phase. In the early period of the light phase, these AAs were significantly higher in tumor-bearing animals, yet significantly lower in the middle of the light phase when compared with controls. This pilot study highlights the importance of well controlled experiments in studies involving plasma AAs in human breast cancer xenografts, in addition to emphasizing the need for more precise examination of exometabolomic changes using multiple time points.
Collapse
Affiliation(s)
- Rubens Paula Junior
- Faculdade de Medicina de São José Do Rio Preto, São José do Rio Preto, Brazil.
| | | | | | - Akos Pal
- The Institute of Cancer Research, London, UK
| | - Yasmin Asad
- The Institute of Cancer Research, London, UK
| | | | - Roger Chammas
- Instituto Do Câncer Do Estado de São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
11
|
The CBL-LSD1-CXCL8 axis regulates methionine metabolism in glioma. Cytokine 2022; 151:155789. [PMID: 34998158 DOI: 10.1016/j.cyto.2021.155789] [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: 08/10/2021] [Revised: 11/24/2021] [Accepted: 12/20/2021] [Indexed: 11/23/2022]
Abstract
Gliomas are the most frequent type of brain tumors, with a high mortality rate and a lack of efficient targeted therapy. Methionine is an essential amino acid, and restriction of methionine in the diet has been found to prevent metabolic diseases and aging, inhibit cancer growth and improve cancer treatment. However, mechanisms of action by which methionine metabolism affects gliomas remain largely unclear. The present study found that methionine starvation of glioma cells significantly increased the expression of CXCL8. Mechanistically, E3 ubiquitin ligase was found to mediate the ubiquitinated degradation of the histone demethylase LSD1 via CBL, reducing LSD1 protein stability and, enhancing H3K4me1 modification of the CXCL8 gene. CXCL8 was found to be involved in regulating the reprogramming of glycerophospholipid metabolism, enabling it to respond to a methionine-deprived environment. CXCL8 expression was significantly higher in glioma than in normal brain tissue samples, with elevated CXCL8 being associated with poor prognosis. In summary, CBL-mediated degradation of LSD1 acts as an anti-braking system and serves as a quick adaptive mechanism for re-remodeling epigenetic modifications. This, in turn, promotes cell proliferation, even in a methionine-restricted environment. Taken together, these findings indicate that the CBL/LSD1/CXCL8 axis is a novel mechanistic connection linking between methionine metabolism, histone methylation and glycerophospholipid reprogramming in the tumor microenvironment.
Collapse
|
12
|
Zhao J, Dong B, Zhao Y, Yang J, Pei P, Ji X, Quan Y. High-throughput and high-sensitivity capillary electrophoresis–mass spectrometry method for sulfur-containing amino acids. J Anal Sci Technol 2021. [DOI: 10.1186/s40543-021-00295-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
AbstractBiological thiol amino acids have been suggested as biomarkers for pathological changes because they are reactive chemicals that participate in various physiological processes. In this study, multisegmented injection capillary electrophoresis–mass spectrometry with online sample preconcentration was used for analysis of thiol amino acids and intermediates of sulfur metabolism in human glioma cell line U-251 with high accuracy, throughput, and sensitivity. This was achieved using multiple, large-volume injections for online sample preconcentration. The 16 intermediates of sulfur metabolism had a good linear correlation coefficient range of 0.984–1 and the limit of detection range was 1.4–203.9 ng/mL. The recovery ranges of most amino acids were 88.1–114.5%, 89.0–104.3%, and 76.9–104.5% at 0.3, 0.75, and 1.5 μg/mL, respectively. The relative standard deviation ranges for the inter- and intra-day precision were 1.8–10.7% and 4.3–18.8%, respectively. Compared with the traditional injection method, the analytical time for compounds in sulfur metabolism was reduced to 4 min/sample, the method throughput was enhanced five times, and the sensitivity was increased 14.4–33.1 times. Customized injection sequences were applied in experimental optimization. The developed method simplified the experimental optimization to one injection and is suitable for the analysis of sulfur metabolites in biological samples and has high sensitivity, throughput, speed, and accuracy.
Collapse
|
13
|
Firdous S, Abid R, Nawaz Z, Bukhari F, Anwer A, Cheng LL, Sadaf S. Dysregulated Alanine as a Potential Predictive Marker of Glioma-An Insight from Untargeted HRMAS-NMR and Machine Learning Data. Metabolites 2021; 11:507. [PMID: 34436448 PMCID: PMC8402070 DOI: 10.3390/metabo11080507] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 01/04/2023] Open
Abstract
Metabolic alterations play a crucial role in glioma development and progression and can be detected even before the appearance of the fatal phenotype. We have compared the circulating metabolic fingerprints of glioma patients versus healthy controls, for the first time, in a quest to identify a panel of small, dysregulated metabolites with potential to serve as a predictive and/or diagnostic marker in the clinical settings. High-resolution magic angle spinning nuclear magnetic resonance spectroscopy (HRMAS-NMR) was used for untargeted metabolomics and data acquisition followed by a machine learning (ML) approach for the analyses of large metabolic datasets. Cross-validation of ML predicted NMR spectral features was done by statistical methods (Wilcoxon-test) using JMP-pro16 software. Alanine was identified as the most critical metabolite with potential to detect glioma with precision of 1.0, recall of 0.96, and F1 measure of 0.98. The top 10 metabolites identified for glioma detection included alanine, glutamine, valine, methionine, N-acetylaspartate (NAA), γ-aminobutyric acid (GABA), serine, α-glucose, lactate, and arginine. We achieved 100% accuracy for the detection of glioma using ML algorithms, extra tree classifier, and random forest, and 98% accuracy with logistic regression. Classification of glioma in low and high grades was done with 86% accuracy using logistic regression model, and with 83% and 79% accuracy using extra tree classifier and random forest, respectively. The predictive accuracy of our ML model is superior to any of the previously reported algorithms, used in tissue- or liquid biopsy-based metabolic studies. The identified top metabolites can be targeted to develop early diagnostic methods as well as to plan personalized treatment strategies.
Collapse
Affiliation(s)
- Safia Firdous
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore 54590, Pakistan; (S.F.); (R.A.)
- Riphah College of Rehabilitation and Allied Health Sciences, Riphah International University, Lahore 54770, Pakistan
| | - Rizwan Abid
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore 54590, Pakistan; (S.F.); (R.A.)
| | - Zubair Nawaz
- Department of Data Science, Punjab University College of Information Technology, University of the Punjab, Lahore 54590, Pakistan; (Z.N.); (F.B.)
| | - Faisal Bukhari
- Department of Data Science, Punjab University College of Information Technology, University of the Punjab, Lahore 54590, Pakistan; (Z.N.); (F.B.)
| | - Ammar Anwer
- Punjab Institute of Neurosciences (PINS), Lahore General Hospital, Lahore 54000, Pakistan;
| | - Leo L. Cheng
- Departments of Radiology and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA;
| | - Saima Sadaf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore 54590, Pakistan; (S.F.); (R.A.)
| |
Collapse
|
14
|
Nasim MJ, Zuraik MM, Abdin AY, Ney Y, Jacob C. Selenomethionine: A Pink Trojan Redox Horse with Implications in Aging and Various Age-Related Diseases. Antioxidants (Basel) 2021; 10:antiox10060882. [PMID: 34072794 PMCID: PMC8229699 DOI: 10.3390/antiox10060882] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Accepted: 05/27/2021] [Indexed: 01/15/2023] Open
Abstract
Selenium is an essential trace element. Although this chalcogen forms a wide variety of compounds, there are surprisingly few small-molecule organic selenium compounds (OSeCs) in biology. Besides its more prominent relative selenocysteine (SeCys), the amino acid selenomethionine (SeMet) is one example. SeMet is synthesized in plants and some fungi and, via nutrition, finds its way into mammalian cells. In contrast to its sulfur analog methionine (Met), SeMet is extraordinarily redox active under physiological conditions and via its catalytic selenide (RSeR')/selenoxide (RSe(O)R') couple provides protection against reactive oxygen species (ROS) and other possibly harmful oxidants. In contrast to SeCys, which is incorporated via an eloquent ribosomal mechanism, SeMet can enter such biomolecules by simply replacing proteinogenic Met. Interestingly, eukaryotes, such as yeast and mammals, also metabolize SeMet to a small family of reactive selenium species (RSeS). Together, SeMet, proteins containing SeMet and metabolites of SeMet form a powerful triad of redox-active metabolites with a plethora of biological implications. In any case, SeMet and its family of natural RSeS provide plenty of opportunities for studies in the fields of nutrition, aging, health and redox biology.
Collapse
Affiliation(s)
- Muhammad Jawad Nasim
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, D-66123 Saarbruecken, Germany; (M.J.N.); (M.M.Z.); (A.Y.A.); (Y.N.)
| | - Mhd Mouayad Zuraik
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, D-66123 Saarbruecken, Germany; (M.J.N.); (M.M.Z.); (A.Y.A.); (Y.N.)
| | - Ahmad Yaman Abdin
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, D-66123 Saarbruecken, Germany; (M.J.N.); (M.M.Z.); (A.Y.A.); (Y.N.)
- University Lille, CNRS, Centrale Lille, University Artois, UMR 8181–UCCS–Unité de Catalyse et Chimie du Solide, F-59000 Lille, France
| | - Yannick Ney
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, D-66123 Saarbruecken, Germany; (M.J.N.); (M.M.Z.); (A.Y.A.); (Y.N.)
| | - Claus Jacob
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, D-66123 Saarbruecken, Germany; (M.J.N.); (M.M.Z.); (A.Y.A.); (Y.N.)
- Correspondence: ; Tel.: +49-681-302-3129
| |
Collapse
|