1
|
Mancini C, Menegazzi G, Peppicelli S, Versienti G, Guasti D, Pieraccini G, Rovida E, Lulli M, Papucci L, Dello Sbarba P, Biagioni A. BCR::ABL1 expression in chronic myeloid leukemia cells in low oxygen is regulated by glutamine via CD36-mediated fatty acid uptake. Cancer Cell Int 2025; 25:176. [PMID: 40369538 PMCID: PMC12080266 DOI: 10.1186/s12935-025-03805-y] [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: 01/02/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is influenced by microenvironmental nutrients, glucose (Glc), and glutamine (Gln) which regulate cell proliferation, viability, and the expression of the driver oncoprotein (BCR::ABL1). RESULTS Our study revealed that Glc, while partially supporting alone cell growth in normoxia, is essential in low oxygen conditions, whereas Gln is ineffective. Under low oxygen, Gln reduced oxidative respiratory activity while enhancing glycolysis. In these conditions, fatty acid (FA) metabolism becomes crucial, as evidenced by increased lipid droplets (LD) accumulation when Glc was absent. Gln, in particular, drives CD36-mediated FA uptake, suppressing the BCR::ABL1 oncoprotein and facilitating cell survival. By co-culturing leukemia cells with adipocytes, one of the main bone marrow (BM) cell components, we observed an enhanced FA release, suggesting a link between FA, microenvironmental BM cells, and the maintenance of leukemic stem cells (LSC). METHODS K562 and KCL22 cell lines were subjected to Glc and/or Gln deprivation under hypoxic conditions (96 h at 0.1% O2). Metabolic profiling was conducted through the Seahorse XFe96 analyzer, and the contribution of L-Glutamine-13C5 to FA de novo synthesis was determined via GC/MS. Intracellular neutral LD were measured using BODIPY 493/503 in confocal microscopy and flow cytometry, with their presence and morphology further examined via transmission electron microscopy. BCR::ABL1 as well as several FA-related markers were evaluated via Western Blotting, whilst CD36 was determined through flow cytometry. LC2 assay was used for measuring leukemia stem cell potential by inhibiting FA uptake via the usage of the Sulfo-N-Succinimidyl Oleate, a CD36 inhibitor. qPCR was exploited to detect markers of FA secretion in CML-adipocytes co-culture together with Nile Red staining to assess free FA in the media. CONCLUSIONS These findings underscore the central role of FA in the regulation of the LSC compartment of CML, highlighting the importance of Gln in facilitating CML cell survival under restrictive metabolic conditions and preparing the cell population for expansion upon the release of these restrictions.
Collapse
Affiliation(s)
- Caterina Mancini
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Giulio Menegazzi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Giampaolo Versienti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Giuseppe Pieraccini
- CISM Mass Spectrometry Center, Università degli Studi di Firenze, Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy.
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università degli Studi di Firenze, Florence, Italy.
| |
Collapse
|
2
|
Lu M, Wu Y, Xia M, Zhang Y. The role of metabolic reprogramming in liver cancer and its clinical perspectives. Front Oncol 2024; 14:1454161. [PMID: 39610917 PMCID: PMC11602425 DOI: 10.3389/fonc.2024.1454161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
Primary liver cancer (PLC), which includes hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), remains a leading cause of cancer-related death worldwide. Chronic liver diseases, such as hepatitis B and C infections and metabolic dysfunction-associated steatotic liver disease (MASLD), are key risk factors for PLC. Metabolic reprogramming, a defining feature of cancer, enables liver cancer cells to adapt to the demands of rapid proliferation and the challenging tumor microenvironment (TME). This manuscript examines the pivotal role of metabolic reprogramming in PLC, with an emphasis on the alterations in glucose, lipid, and amino acid metabolism that drive tumor progression. The Warburg effect, marked by increased glycolysis, facilitates rapid energy production and biosynthesis of cellular components in HCC. Changes in lipid metabolism, including elevated de novo fatty acid synthesis and lipid oxidation, support membrane formation and energy storage essential for cancer cell survival. Amino acid metabolism, particularly glutamine utilization, supplies critical carbon and nitrogen for nucleotide synthesis and maintains redox homeostasis. These metabolic adaptations not only enhance tumor growth and invasion but also reshape the TME, promoting immune escape. Targeting these metabolic pathways presents promising therapeutic opportunities for PLC. This review underscores the interaction between metabolic reprogramming and tumor immunity, suggesting potential metabolic targets for innovative therapeutic strategies. A comprehensive understanding of PLC's intricate metabolic landscape may lead to more effective treatments and better patient outcomes. Integrating metabolomics, genomics, and proteomics in future research will be vital for identifying precise therapeutic targets and advancing personalized therapies for liver cancer.
Collapse
Affiliation(s)
- Mengxiao Lu
- Department of Gastrointestinal Minimally Invasive Surgery, The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| | | | | | | |
Collapse
|
3
|
Zhang J, Chen M, Yang Y, Liu Z, Guo W, Xiang P, Zeng Z, Wang D, Xiong W. Amino acid metabolic reprogramming in the tumor microenvironment and its implication for cancer therapy. J Cell Physiol 2024; 239:e31349. [PMID: 38946173 DOI: 10.1002/jcp.31349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024]
Abstract
Amino acids are essential building blocks for proteins, crucial energy sources for cell survival, and key signaling molecules supporting the resistant growth of tumor cells. In tumor cells, amino acid metabolic reprogramming is characterized by the enhanced uptake of amino acids as well as their aberrant synthesis, breakdown, and transport, leading to immune evasion and malignant progression of tumor cells. This article reviews the altered amino acid metabolism in tumor cells and its impact on tumor microenvironment, and also provides an overview of the current clinical applications of amino acid metabolism. Innovative drugs targeting amino acid metabolism hold great promise for precision and personalized cancer therapy.
Collapse
Affiliation(s)
- Jiarong Zhang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Mingjian Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Yuxin Yang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Ziqi Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wanni Guo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Pingjuan Xiang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Dan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| |
Collapse
|
4
|
Barrera-Adame D, Schuster S, Niedermeyer THJ. Mass Spectrometry Imaging of Coniine and Other Hemlock Alkaloids after On-Tissue Derivatization Reveals Distinct Alkaloid Distributions in the Plant. JOURNAL OF NATURAL PRODUCTS 2024; 87:2376-2383. [PMID: 38904441 PMCID: PMC11519914 DOI: 10.1021/acs.jnatprod.4c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024]
Abstract
Specialized metabolites play important roles in plants and can, for example, protect plants from predators or pathogens. Alkaloids, due to their pronounced biological activity on higher animals, are one of the most intriguing groups of specialized metabolites, and many of them are known as plant defense compounds. Poison hemlock, Conium maculatum, is well-known for its high content of piperidine alkaloids, of which coniine is the most famous. The distribution, localization, and diversity of these compounds in C. maculatum tissues have not yet been studied in detail. The hemlock alkaloids are low molecular weight compounds with relatively high volatility. They are thus difficult to analyze on-tissue by MALDI mass spectrometry imaging due to delocalization, which occurs even when using an atmospheric pressure ion source. In this manuscript, we describe an on-tissue derivatization method that allows the subsequent determination of the spatial distribution of hemlock alkaloids in different plant tissues by mass spectrometry imaging. Coniferyl aldehyde was found to be a suitable reagent for derivatization of the secondary amine alkaloids. The imaging analysis revealed that even chemically closely related hemlock alkaloids are discretely distributed in different plant tissues. Additionally, we detected a yet undescribed hemlock alkaloid in Conium maculatum seeds.
Collapse
Affiliation(s)
- Diana
A. Barrera-Adame
- Department
of Pharmaceutical Biology/Pharmacognosy, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Department
of Pharmaceutical Biology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Sabine Schuster
- Department
of Pharmaceutical Biology/Pharmacognosy, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Timo H. J. Niedermeyer
- Department
of Pharmaceutical Biology/Pharmacognosy, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Department
of Pharmaceutical Biology, Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
5
|
Xiao C, Li Y, Liu Y, Dong R, He X, Lin Q, Zang X, Wang K, Xia Y, Kong L. Overcoming Cancer Persister Cells by Stabilizing the ATF4 Promoter G-quadruplex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401748. [PMID: 38994891 PMCID: PMC11425212 DOI: 10.1002/advs.202401748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/23/2024] [Indexed: 07/13/2024]
Abstract
Persister cells (PS) selected for anticancer therapy have been recognized as a significant contributor to the development of treatment-resistant malignancies. It is found that imposing glutamine restriction induces the generation of PS, which paradoxically bestows heightened resistance to glutamine restriction treatment by activating the integrated stress response and initiating the general control nonderepressible 2-activating transcription factor 4-alanine, serine, cysteine-preferring transporter 2 (GCN2-ATF4-ASCT2) axis. Central to this phenomenon is the stress-induced ATF4 translational reprogramming. Unfortunately, directly targeting ATF4 protein has proven to be a formidable challenge because of its flat surface. Nonetheless, a G-quadruplex structure located within the promoter region of ATF4 (ATF4-G4) is uncovered and resolved, which functions as a transcriptional regulator and can be targeted by small molecules. The investigation identifies the natural compound coptisine (COP) as a potent binder that interacts with and stabilizes ATF4-G4. For the first time, the high-resolution structure of the COP-ATF4-G4 complex is determined. The formation of this stable complex disrupts the interaction between transcription factor AP-2 alpha (TFAP2A) and ATF4-G4, resulting in a substantial reduction in intracellular ATF4 levels and the eventual death of cancer cells. These seminal findings underscore the potential of targeting the ATF4-G4 structure to yield significant therapeutic advantages within the realm of persister cancer cells induced by glutamine-restricted therapy.
Collapse
Affiliation(s)
- Chengmei Xiao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Yipu Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Yushuang Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Ruifang Dong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Xiaoyu He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Qing Lin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Xin Zang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Kaibo Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| | - Yuanzheng Xia
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
- Shenzhen Research Institute of China Pharmaceutical UniversityShenzhen518057China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product ResearchSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing210009China
| |
Collapse
|
6
|
Yin H, Liu Y, Dong Q, Wang H, Yan Y, Wang X, Wan X, Yuan G, Pan Y. The mechanism of extracellular CypB promotes glioblastoma adaptation to glutamine deprivation microenvironment. Cancer Lett 2024; 597:216862. [PMID: 38582396 DOI: 10.1016/j.canlet.2024.216862] [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: 02/13/2024] [Revised: 03/21/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Glioblastoma, previously known as glioblastoma multiform (GBM), is a type of glioma with a high degree of malignancy and rapid growth rate. It is highly dependent on glutamine (Gln) metabolism during proliferation and lags in neoangiogenesis, leading to extensive Gln depletion in the core region of GBM. Gln-derived glutamate is used to synthesize the antioxidant Glutathione (GSH). We demonstrated that GSH levels are also reduced in Gln deficiency, leading to increased reactive oxygen species (ROS) levels. The ROS production induces endoplasmic reticulum (ER) stress, and the proteins in the ER are secreted into the extracellular medium. We collected GBM cell supernatants cultured with or without Gln medium; the core and peripheral regions of human GBM tumor tissues. Proteomic analysis was used to screen out the target-secreted protein CypB. We demonstrated that the extracellular CypB expression is associated with Gln deprivation. Then, we verified that GBM can promote the glycolytic pathway by activating HIF-1α to upregulate the expression of GLUT1 and LDHA. Meanwhile, the DRP1 was activated, increasing mitochondrial fission, thus inhibiting mitochondrial function. To explore the specific mechanism of its regulation, we constructed a si-CD147 knockout model and added human recombinant CypB protein to verify that extracellular CypB influenced the expression of downstream p-AKT through its cell membrane receptor CD147 binding. Moreover, we confirmed that p-AKT could upregulate HIF-1α and DRP1. Finally, we observed that extracellular CypB can bind to the CD147 receptor, activate p-AKT, upregulate HIF-1α and DRP1 in order to promote glycolysis while inhibiting mitochondrial function to adapt to the Gln-deprived microenvironment.
Collapse
Affiliation(s)
- Hang Yin
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Yang Liu
- Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China; Neurological Diseases Clinical Medical Research Center of Gansu Province, Lanzhou, China
| | - Qiang Dong
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Hongyu Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Yunji Yan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaoqing Wang
- Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China; Neurological Diseases Clinical Medical Research Center of Gansu Province, Lanzhou, China
| | - Xiaoyu Wan
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore, Singapore; School of Basic Medicine, Henan University, Kaifeng, China
| | - Guoqiang Yuan
- Laboratory of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China; Neurological Diseases Clinical Medical Research Center of Gansu Province, Lanzhou, China.
| | - Yawen Pan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
7
|
Zhang Q, Zhou J, Zhai D, Jiang Q, Yang M, Zhou M. Gut microbiota regulates the ALK5/NOX1 axis by altering glutamine metabolism to inhibit ferroptosis of intrahepatic cholangiocarcinoma cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167152. [PMID: 38582012 DOI: 10.1016/j.bbadis.2024.167152] [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: 04/14/2023] [Revised: 03/14/2024] [Accepted: 03/27/2024] [Indexed: 04/08/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a kind of hepatobiliary tumor that is increasing in incidence and mortality. The gut microbiota plays a role in the onset and progression of cancer, however, the specific mechanism by which the gut microbiota acts on ICC remains unclear. In this study, feces and plasma from healthy controls and ICC patients were collected for 16S rRNA sequencing or metabolomics analysis. Gut microbiota analysis showed that gut microbiota abundance and biodiversity were altered in ICC patients compared with controls. Plasma metabolism analysis showed that the metabolite glutamine content of the ICC patient was significantly higher than that of the controls. KEGG pathway analysis showed that glutamine plays a vital role in ICC. In addition, the use of antibiotics in ICC animals further confirmed that changes in gut microbiota affect changes in glutamine. Further experiments showed that supplementation with glutamine inhibited ferroptosis and downregulated ALK5 and NOX1 expression in HuCCT1 cells. ALK5 overexpression or NOX1 overexpression increased NOX1, p53, PTGS2, ACSL4, LPCAT3, ROS, MDA and Fe2+ and decreased FTH1, SLC7A11 and GSH. Knockdown of NOX1 suppressed FIN56-induced ferroptosis. In vivo, supplementation with glutamine promoted tumor growth. Overexpression of ALK5 repressed tumor growth and induced ferroptosis in nude mice, which could be reversed by the addition of glutamine. Our results suggested that the gut microbiota altered glutamine metabolism to inhibit ferroptosis in ICC by regulating the ALK5/NOX1 axis.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Xiangya Hospital Central South University, Changsha 410008, China; International Joint Research Center of Minimally Invasive Endoscopic Technology Equipment & Standards, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jixiang Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Xiangya Hospital Central South University, Changsha 410008, China
| | - Denggao Zhai
- Department of Hepatobiliary and Pancreatic Surgery, Xiangya Hospital Central South University, Changsha 410008, China
| | - Qin Jiang
- Department of Ultrasonography, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, NHC Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Maojun Zhou
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
8
|
Duncan KD, Pětrošová H, Lum JJ, Goodlett DR. Mass spectrometry imaging methods for visualizing tumor heterogeneity. Curr Opin Biotechnol 2024; 86:103068. [PMID: 38310648 PMCID: PMC11520788 DOI: 10.1016/j.copbio.2024.103068] [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: 08/28/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
Profiling spatial distributions of lipids, metabolites, and proteins in tumors can reveal unique cellular microenvironments and provide molecular evidence for cancer cell dysfunction and proliferation. Mass spectrometry imaging (MSI) is a label-free technique that can be used to map biomolecules in tumors in situ. Here, we discuss current progress in applying MSI to uncover molecular heterogeneity in tumors. First, the analytical strategies to profile small molecules and proteins are outlined, and current methods for multimodal imaging to maximize biological information are highlighted. Second, we present and summarize biological insights obtained by MSI of tumor tissue. Finally, we discuss important considerations for designing MSI experiments and several current analytical challenges.
Collapse
Affiliation(s)
- Kyle D Duncan
- Department of Chemistry, Vancouver Island University, Nanaimo, British Columbia, Canada; Department of Chemistry, University of Victoria, Victoria, British Columbia, Canada.
| | - Helena Pětrošová
- University of Victoria Genome British Columbia Proteomics Center, University of Victoria, Victoria, British Columbia, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada.
| | - Julian J Lum
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada; Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - David R Goodlett
- University of Victoria Genome British Columbia Proteomics Center, University of Victoria, Victoria, British Columbia, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
9
|
Yang F, Hilakivi-Clarke L, Shaha A, Wang Y, Wang X, Deng Y, Lai J, Kang N. Metabolic reprogramming and its clinical implication for liver cancer. Hepatology 2023; 78:1602-1624. [PMID: 36626639 PMCID: PMC10315435 DOI: 10.1097/hep.0000000000000005] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023]
Abstract
Cancer cells often encounter hypoxic and hypo-nutrient conditions, which force them to make adaptive changes to meet their high demands for energy and various biomaterials for biomass synthesis. As a result, enhanced catabolism (breakdown of macromolecules for energy production) and anabolism (macromolecule synthesis from bio-precursors) are induced in cancer. This phenomenon is called "metabolic reprogramming," a cancer hallmark contributing to cancer development, metastasis, and drug resistance. HCC and cholangiocarcinoma (CCA) are 2 different liver cancers with high intertumoral heterogeneity in terms of etiologies, mutational landscapes, transcriptomes, and histological representations. In agreement, metabolism in HCC or CCA is remarkably heterogeneous, although changes in the glycolytic pathways and an increase in the generation of lactate (the Warburg effect) have been frequently detected in those tumors. For example, HCC tumors with activated β-catenin are addicted to fatty acid catabolism, whereas HCC tumors derived from fatty liver avoid using fatty acids. In this review, we describe common metabolic alterations in HCC and CCA as well as metabolic features unique for their subsets. We discuss metabolism of NAFLD as well, because NAFLD will likely become a leading etiology of liver cancer in the coming years due to the obesity epidemic in the Western world. Furthermore, we outline the clinical implication of liver cancer metabolism and highlight the computation and systems biology approaches, such as genome-wide metabolic models, as a valuable tool allowing us to identify therapeutic targets and develop personalized treatments for liver cancer patients.
Collapse
Affiliation(s)
- Flora Yang
- BA/MD Joint Admission Scholars Program, University of Minnesota, Minneapolis, Minnesota
| | - Leena Hilakivi-Clarke
- Food Science and Nutrition Section, The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Aurpita Shaha
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yuanguo Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Xianghu Wang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yibin Deng
- Department of Urology, Masonic Cancer Center, The University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jinping Lai
- Department of Pathology and Laboratory Medicine, Kaiser Permanente Sacramento Medical Center, Sacramento, California
| | - Ningling Kang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, Minnesota
| |
Collapse
|
10
|
Zhang X, Han S, Jiang X, Duan S, Gao Y, Ding J, Li X, Sun B, Hu X, Zhang X, Zhang W. Comparative analysis of bile metabolic profile in patients with biliary obstruction complicated by Clonorchis sinensis infection. Front Cell Infect Microbiol 2023; 13:1254016. [PMID: 37868349 PMCID: PMC10585366 DOI: 10.3389/fcimb.2023.1254016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/21/2023] [Indexed: 10/24/2023] Open
Abstract
Background Clonorchiasis is an important foodborne parasitic disease. However, eggs of Clonorchis sinensis (C. sinensis) cannot be detected in feces during biliary obstruction. Moreover, many diseases can cause biliary obstruction, such as gallstones, adenocarcinoma, cholangiocarcinoma and Ascaris lumbricoides infection. Therefore, it is of great significance to distinguish between patients of biliary obstruction and biliary obstruction with C. sinensis infection. Methods A total of 48 biliary obstruction patients were enrolled, including 23 infected with C. sinensis (C. sinensis) (OB+C.s) and 25 non-infected subjects (OB). The bile samples were collected by endoscopic retrograde cholangiopancreatography and analyzed using ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-QTOF MS). Additionally, multivariate statistical analysis methods were employed to identify differential metabolites. Next, bile amino acid levels were determined by targeted metabolomics analysis. Result A total of 146 and 132 significant metabolites were identified in electrospray ionization (ESI)+ and ESI- modes, respectively. The levels of amino acids (asparagine, glutamate, ornithine) and polyamines (spermidine and spermine) were significantly changed. Targeted analysis showed that the levels of amino acids (such as L-arginine, L-glutamine, L-lysine, L-propionic, and L-tyrosine) were lower in OB+C.s patients compared to those in OB patients. Marked metabolic pathways were involved in "Glutathione metabolism", "Caffeine metabolism", "Alanine, aspartate and glutamate metabolism", "Arginine and proline metabolism", "Purine metabolism", "Beta-Alanine metabolism", and "D-glutamine and D-glutamate metabolism". Conclusion These results show that there were significant differences between OB+C.s and OB patients, especially in amino acids. The metabolic signature and perturbations in metabolic pathways may help to better distinguish OB+C.s and OB patients.
Collapse
Affiliation(s)
- Xueli Zhang
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Su Han
- Department of Parasitology, Harbin Medical University, Harbin, China
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xu Jiang
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Shanshan Duan
- Beijing Obstetrics and Gynecology Hospital Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yannan Gao
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian Ding
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Xiang Li
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Beibei Sun
- Clinical Laboratory, Zhuhai Maternal and Child Health Hospital, Zhuhai, China
| | - Xinyi Hu
- Department of Stomatology, Laixi People's Hospital, Shandong, China
| | - Xiaoli Zhang
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Weizhe Zhang
- Department of Parasitology, Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Yang Y, Zhang X. An overview of extrahepatic cholangiocarcinoma: from here to where? Front Oncol 2023; 13:1171098. [PMID: 37197436 PMCID: PMC10183586 DOI: 10.3389/fonc.2023.1171098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/11/2023] [Indexed: 05/19/2023] Open
Abstract
Extrahepatic cholangiocarcinoma (eCCA) contains perihilar cholangiocarcinoma and distal cholangiocarcinoma both of which can arise at any point of the biliary tree and originate from disparate anatomical sites. Generally, the incidence of eCCA is increasing globally. Though surgical resection is the principal treatment of choice for the early stages of eCCA, optimal survival remains restricted by the high risk of recurrence when most patients are present with unresectable disease or distant metastasis. Furthermore, both intra- and intertumoral heterogeneity make it laborious to determine molecularly targeted therapies. In this review, we mainly focused on current findings in the field of eCCA, mostly including epidemiology, genomic abnormalities, molecular pathogenesis, tumor microenvironment, and other details while a summary of the biological mechanisms driving eCCA may shed light on intricate tumorigenesis and feasible treatment strategies.
Collapse
|
12
|
Xu Y, Xia Z, Sun X, Wei B, Fu Y, Shi D, Zhu Y. Identification of a glutamine metabolism reprogramming signature for predicting prognosis, immunotherapy efficacy, and drug candidates in bladder cancer. Front Immunol 2023; 14:1111319. [PMID: 36911676 PMCID: PMC9995899 DOI: 10.3389/fimmu.2023.1111319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Background Bladder cancer is the most common malignancy of the urinary system. However, patient prognosis and treatment outcomes in bladder cancer are difficult to predict owing to high tumor heterogeneity. Given that abnormal glutamine metabolism has been identified as a key factor driving the progression of bladder cancer, it is necessary to assess the prognosis and therapeutic efficacy of bladder cancer treatments based on an analysis of glutamine metabolism-related genes. Methods We used bladder cancer sample data downloaded from The Cancer Genome Atlas to identify glutamine metabolism-related genes as prognostic markers, and established a novel Glutamine Metabolism Immunity Index (GMII) based on univariate and multivariate COX regression analyses. On the basis of GMII values, bladder cancer patients were divided into high- and low-risk groups, and systematic analysis was conducted for clinical features, somatic mutations, immune cell infiltration, chemotherapeutic response, and immunotherapeutic efficacy. Candidate small-molecule drugs targeting the GMII core target proteins were identified based on molecular docking analysis. Results The GMII consisting of eight independent prognostic genes was established to be an excellent tool for predicting the survival in patients with bladder cancer and was validated using multiple datasets. Compared with patients in the high-risk group, those in the low-risk group had significantly better responses to gemcitabine and immune checkpoint blockade. In addition, we predicted 12 potential small-molecule drugs that could bind to three of the GMII core target proteins. Conclusions The GMII can be used to accurately predict the prognosis and immunotherapeutic response of bladder cancer patients, as well as candidate small-molecule drugs. Furthermore, the novel "Glutamine Metabolism-related Gene"-guided strategy for predicting survival and chemo-immunotherapeutic efficacy may also be applicable for cancers other than bladder cancer.
Collapse
Affiliation(s)
- Yan Xu
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Zhixiu Xia
- Colorectal Tumor Surgery Ward, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoyu Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Baojun Wei
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Yang Fu
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Du Shi
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Yuyan Zhu
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Glutamine Transporter SLC1A5 Regulates Ionizing Radiation-Derived Oxidative Damage and Ferroptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3403009. [PMID: 36262284 PMCID: PMC9576409 DOI: 10.1155/2022/3403009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 12/08/2022]
Abstract
Ionizing radiation-derived oxidative stress and ferroptosis are one of the most important biological effects on destroying the liver tumor, whereas radioresistance of liver tumor remains a leading cause of radiotherapy (RT) failure mainly because of the protective antiferroptosis, in which oxidative stress and subsequent lipid peroxidation are the key initiators. Thus, it is of great importance to overcome ferroptosis resistance to improve the therapeutic efficacy of RT in liver tumor patients. Irradiation-resistant HepG2 cells (HepG2-IRR) were established by long-term exposure to X-ray (2 to 8 Gy), and targeted metabolomics analysis revealed an obvious increase in intracellular amino acids in HepG2-IRR cells upon ferroptosis stress. Among these amino acids with obvious changes, N-acetylglutamine, a derivative of glutamine, is essential for the redox homeostasis and progression of tumor cells. Interestingly, the treatment of glutamine starvation could promote the ferroptosis effect significantly, whereas glutamine supplementation reversed the ferroptosis effect completely. Consistent with the changes in amino acids pattern, the glutamine transporter SLC1A5 was verified in liver tumor samples from TCGA training and validation cohorts as an independent prognostic amino acid-ferroptosis gene (AFG). A risk score for screening prognosis based on the SLC1A5, SLC7A11, ASNS, and TXNRD1 demonstrated that a high-risk score was correlated with poor survival. In vitro studies had shown that the knockdown of SLC1A5 resulted in a significant decrease in cell viability and promoted lipid peroxidation and oxidative damage introduced by irradiation (10 Gy). Collectively, our findings indicated that SLC1A5 may act as a suppressor gene against ferroptosis and can be a potential target for ionizing radiation mediated effects.
Collapse
|
14
|
Bhardwaj V, He J, Jain A. Glutamine stabilizes myc via alpha-ketoglutarate and regulates paclitaxel sensitivity. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:227. [PMID: 36175776 DOI: 10.1007/s12032-022-01834-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022]
Abstract
Metabolic reprogramming wherein the cancer cells exhibit altered energetics is a hallmark of cancer. Although recent discoveries have enhanced our understanding of tumor metabolism, the therapeutic utility of targeting tumor metabolism is not yet realized. Glutamine, a non-essential amino acid, plays a critical role in regulating tumor metabolism and provides an alternative tumor energy source. In this study, we investigate the molecular mechanism regulated by glutamine and elucidate if targeting glutamine metabolism would enhance the efficacy of cancer chemotherapy. Using clonogenic and cell cycle analysis, we found that deprivation of glutamine suppress the growth of cancer cells. Mechanistically we demonstrate that glutamine stabilizes myc by preventing its ubiquitination through alpha-ketoglutarate. Inhibition of glutamine metabolism enhanced the sensitivity of tumor cells to chemotherapeutic agent paclitaxel. Our results delineate the mechanism behind glutamine-induced myc stabilization, and they provide a viable strategy to target cancer with a glutamine metabolism inhibitor in the clinic.
Collapse
Affiliation(s)
- Vikas Bhardwaj
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Jun He
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aditi Jain
- The Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
15
|
Chen Y, Xie Y, Li L, Wang Z, Yang L. Advances in mass spectrometry imaging for toxicological analysis and safety evaluation of pharmaceuticals. MASS SPECTROMETRY REVIEWS 2022:e21807. [PMID: 36146929 DOI: 10.1002/mas.21807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 06/16/2023]
Abstract
Safety issues caused by pharmaceuticals have frequently occurred worldwide, posing a tremendous threat to human health. As an essential part of drug development, the toxicological analysis and safety evaluation is of great significance. In addition, the risk of pharmaceuticals accumulation in the environment and the monitoring of the toxicity from natural medicines have also received ongoing concerns. Due to a lack of spatial distribution information provided by common analytical methods, analyses that provide spatial dimensions could serve as complementary safety evaluation methods for better prediction and evaluation of drug toxicity. With advances in technical solutions and software algorithms, mass spectrometry imaging (MSI) has received increasing attention as a popular analytical tool that enables the simultaneous implementation of qualitative, quantitative, and localization without complex sample pretreatment and labeling steps. In recent years, MSI has become more attractive, powerful, and sensitive and has been applied in several scientific fields that can meet the safety assessment requirements. This review aims to cover a detailed summary of the various MSI technologies utilized in the biomedical and pharmaceutical area, including technical principles, advantages, current status, and future trends. Representative applications and developments in the safety-related issues of different pharmaceuticals and natural medicines are also described to provide a reference for pharmaceutical research, improve rational clinical medicine use, and ensure public safety.
Collapse
Affiliation(s)
- Yilin Chen
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqiao Xie
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linnan Li
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengtao Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Yang
- The MOE Key Laboratory of Standardization of Chinese Medicines, the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Raggi C, Taddei ML, Rae C, Braconi C, Marra F. Metabolic reprogramming in cholangiocarcinoma. J Hepatol 2022; 77:849-864. [PMID: 35594992 DOI: 10.1016/j.jhep.2022.04.038] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/16/2022] [Accepted: 04/28/2022] [Indexed: 12/25/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer and allows tumour cells to meet the increased energy demands required for rapid proliferation, invasion, and metastasis. Indeed, many tumour cells acquire distinctive metabolic and bioenergetic features that enable them to survive in resource-limited conditions, mainly by harnessing alternative nutrients. Several recent studies have explored the metabolic plasticity of cancer cells with the aim of identifying new druggable targets, while therapeutic strategies to limit the access to nutrients have been successfully applied to the treatment of some tumours. Cholangiocarcinoma (CCA), a highly heterogeneous tumour, is the second most common form of primary liver cancer. It is characterised by resistance to chemotherapy and poor prognosis, with 5-year survival rates of below 20%. Deregulation of metabolic pathways have been described during the onset and progression of CCA. Increased aerobic glycolysis and glutamine anaplerosis provide CCA cells with the ability to generate biosynthetic intermediates. Other metabolic alterations involving carbohydrates, amino acids and lipids have been shown to sustain cancer cell growth and dissemination. In this review, we discuss the complex metabolic rewiring that occurs during CCA development and leads to unique nutrient addiction. The possible role of therapeutic interventions based on metabolic changes is also thoroughly discussed.
Collapse
Affiliation(s)
- Chiara Raggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Colin Rae
- Institute of Cancer Sciences, The University of Glasgow, Glasgow, United Kingdom
| | - Chiara Braconi
- Institute of Cancer Sciences, The University of Glasgow, Glasgow, United Kingdom; Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
17
|
Hypoxia-driven metabolic heterogeneity and immune evasive behaviour of gastrointestinal cancers: Elements of a recipe for disaster. Cytokine 2022; 156:155917. [PMID: 35660715 DOI: 10.1016/j.cyto.2022.155917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/28/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
Gastrointestinal (GI) cancers refer to a group of malignancies associated with the GI tract (GIT). Like other solid tumors, hypoxic regions consistently feature inside the GI tumor microenvironment (TME) and contribute towards metabolic reprogramming of tumor-resident cells by modulating hypoxia-induced factors. We highlight here how the metabolic crosstalk between cancer cells and immune cells generate immunosuppressive environment inside hypoxic tumors. Given the fluctuating nature of tumor hypoxia, the metabolic fluxes between immune cells and cancer cells change dynamically. These changes alter cellular phenotypes and functions, resulting in the acceleration of cancer progression. These evolved properties of hypoxic tumors make metabolism-targeting monotherapy approaches or immunotherapy-measures unsuccessful. The current review highlights the advantages of combined immunometabolic treatment strategies to target hypoxic GI cancers and also identifies research areas to develop better combinational therapeutics for future.
Collapse
|
18
|
Zhang X, Li Q, Du A, Li Y, Shi Q, Chen Y, Zhao Y, Wang B, Pan F. Adipocytic Glutamine Synthetase Upregulation via Altered Histone Methylation Promotes 5FU Chemoresistance in Peritoneal Carcinomatosis of Colorectal Cancer. Front Oncol 2021; 11:748730. [PMID: 34712612 PMCID: PMC8547656 DOI: 10.3389/fonc.2021.748730] [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] [Received: 07/28/2021] [Accepted: 09/22/2021] [Indexed: 11/18/2022] Open
Abstract
The development of resistance to 5-fluorouracil (5FU) chemotherapy is a major handicap for sustained effective treatment in peritoneal carcinomatosis (PC) of colorectal cancer (CRC). Metabolic reprogramming of adipocytes, a component of the tumor microenvironment and the main composition of peritoneum, plays a significant role in drug resistance of PC, with the mechanisms being not fully understood. By performing metabolomics analysis, we identified glutamine (Gln), an important amino acid, inducing resistance to 5FU-triggered tumor suppression of CRC-PC through activating mTOR pathway. Noteworthily, genetic overexpression of glutamine synthetase (GS) in adipocytes increased chemoresistance to 5FU in vitro and in vivo while this effect was reversed by pharmacological blockage of GS. Next, we showed that methionine metabolism were enhanced in amino acid omitted from CRC-PC of GS transgenic (TgGS) mice, increasing intracellular levels of S-carboxymethy-L-cys. Moreover, loss of dimethylation at lysine 4 of histone H3 (H3k4me2) was found in adipocytes in vitro, which may lead to increased expression of GS. Furthermore, biochemical inhibition of lysine specific demethylase 1 (LSD1) restored H3k4me2, thereby reducing GS-induced chemoresistance to 5FU. Our findings indicate that GS upregulation-induced excessive of Gln in adipocytes via altered histone methylation is potential mediator of resistance to 5FU chemotherapy in patients with CRC-PC.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Oncology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qing Li
- Department of Science and Education, The People's Hospital of Tongliang District, Chongqing, China
| | - Aibei Du
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yifei Li
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Shi
- Department of Respiratory Medicine, The People's Hospital of Tongliang District, Chongqing, China
| | - Yanrong Chen
- Department of Oncology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yang Zhao
- Department of Oncology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bin Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Feng Pan
- Department of Oncology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
19
|
Merdas M, Lagarrigue M, Vanbellingen Q, Umbdenstock T, Da Violante G, Pineau C. On-tissue chemical derivatization reagents for matrix-assisted laser desorption/ionization mass spectrometry imaging. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4731. [PMID: 34080257 DOI: 10.1002/jms.4731] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/26/2021] [Accepted: 04/13/2021] [Indexed: 05/27/2023]
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) is a key tool for the analysis of biological tissues. It provides spatial and quantitative information about different types of analytes within tissue sections. Despite the increasing improvements of this technique, the low detection sensitivity of some compounds remains an important challenge to overcome. Poor sensitivity is related to weak ionization efficiency, low abundance of analytes and matrix ions, or endogenous interferences. On-tissue chemical derivatization (OTCD) has proven to be an important solution to these issues and is increasingly employed in MALDI MSI studies. OTCD reagents, synthesized or commercially available, have been essentially used for the detection of small exogenous or endogenous molecules within tissues. Optimally, an OTCD reaction is performed in mild conditions, in an acceptable range of time, preserves the integrity of the tissues, and prevents the delocalization. In addition to their reactivity with a targeted chemical function, some OTCD reagents can also be used as a matrix, which simplifies the sample preparation procedure. In this review, we present an exhaustive overview of OTCD reagents and methods used in MALDI MSI studies.
Collapse
Affiliation(s)
- Mira Merdas
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, F-35042, France
- Protim, Univ Rennes, Rennes, F-35042, France
- DMPK Department, Technologie Servier, Orléans, 45007, France
| | - Mélanie Lagarrigue
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, F-35042, France
- Protim, Univ Rennes, Rennes, F-35042, France
| | | | | | | | - Charles Pineau
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, F-35042, France
- Protim, Univ Rennes, Rennes, F-35042, France
| |
Collapse
|
20
|
Poteti M, Menegazzi G, Peppicelli S, Tusa I, Cheloni G, Silvano A, Mancini C, Biagioni A, Tubita A, Mazure NM, Lulli M, Rovida E, Dello Sbarba P. Glutamine Availability Controls BCR/Abl Protein Expression and Functional Phenotype of Chronic Myeloid Leukemia Cells Endowed with Stem/Progenitor Cell Potential. Cancers (Basel) 2021; 13:cancers13174372. [PMID: 34503182 PMCID: PMC8430815 DOI: 10.3390/cancers13174372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary In chronic myeloid leukemia (CML), a neoplasm brilliantly taken care of by a molecularly targeted therapeutic approach, the achievement of cure is nevertheless prevented by the maintenance of a small subset of treatment-resistant leukemia stem cells (LSCs), sustaining the so-called minimal residual disease of CML. The phenotypical and functional characterization of this LSC subset is, therefore, crucial to aim at the eradication of disease. Such a characterization includes the acquisition of information relative to the metabolic profile of treatment-resistant LSCs, which is functional to their maintenance in bone marrow. A number of metabolic features of LSCs were shown to determine their sensitivity or resistance to therapy. Glutamine metabolism emerged from this study as a potential target to overcome the persistence of therapy-resistant LSCs. Abstract This study was directed to characterize the role of glutamine in the modulation of the response of chronic myeloid leukemia (CML) cells to low oxygen, a main condition of hematopoietic stem cell niches of bone marrow. Cells were incubated in atmosphere at 0.2% oxygen in the absence or the presence of glutamine. The absence of glutamine markedly delayed glucose consumption, which had previously been shown to drive the suppression of BCR/Abl oncoprotein (but not of the fusion oncogene BCR/abl) in low oxygen. Glutamine availability thus emerged as a key regulator of the balance between the pools of BCR/Abl protein-expressing and -negative CML cells endowed with stem/progenitor cell potential and capable to stand extremely low oxygen. These findings were confirmed by the effects of the inhibitors of glucose or glutamine metabolism. The BCR/Abl-negative cell phenotype is the best candidate to sustain the treatment-resistant minimal residual disease (MRD) of CML because these cells are devoid of the molecular target of the BCR/Abl-active tyrosine kinase inhibitors (TKi) used for CML therapy. Therefore, the treatments capable of interfering with glutamine action may result in the reduction in the BCR/Abl-negative cell subset sustaining MRD and in the concomitant rescue of the TKi sensitivity of CML stem cell potential. The data obtained with glutaminase inhibitors seem to confirm this perspective.
Collapse
Affiliation(s)
- Martina Poteti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Giulio Menegazzi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Giulia Cheloni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
- Beth Israel Deaconess Medical Center, Department of Medicine, Division of Genetics, Harvard University Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Angela Silvano
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Caterina Mancini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Nathalie M. Mazure
- Mediterranean Centre for Molecular Medicine-INSERM U1065, University of Nice-Sophia-Antipolis, 151 Route Saint Antoine de Ginestière, 06204 Nice, France;
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
- Correspondence: (E.R.); (P.D.S.)
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy; (M.P.); (G.M.); (S.P.); (I.T.); (G.C.); (A.S.); (C.M.); (A.B.); (A.T.); (M.L.)
- Correspondence: (E.R.); (P.D.S.)
| |
Collapse
|
21
|
Shuvalov O, Daks A, Fedorova O, Petukhov A, Barlev N. Linking Metabolic Reprogramming, Plasticity and Tumor Progression. Cancers (Basel) 2021; 13:cancers13040762. [PMID: 33673109 PMCID: PMC7917602 DOI: 10.3390/cancers13040762] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/03/2021] [Accepted: 02/07/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In the present review, we discuss the role of metabolic reprogramming which occurs in malignant cells. The process of metabolic reprogramming is also known as one of the “hallmarks of cancer”. Due to several reasons, including the origin of cancer, tumor microenvironment, and the tumor progression stage, metabolic reprogramming can be heterogeneous and dynamic. In this review, we provide evidence that the usage of metabolic drugs is a promising approach to treat cancer. However, because these drugs can damage not only malignant cells but also normal rapidly dividing cells, it is important to understand the exact metabolic changes which are elicited by particular drivers in concrete tissue and are specific for each stage of cancer development, including metastases. Finally, the review highlights new promising targets for the development of new metabolic drugs. Abstract The specific molecular features of cancer cells that distinguish them from the normal ones are denoted as “hallmarks of cancer”. One of the critical hallmarks of cancer is an altered metabolism which provides tumor cells with energy and structural resources necessary for rapid proliferation. The key feature of a cancer-reprogrammed metabolism is its plasticity, allowing cancer cells to better adapt to various conditions and to oppose different therapies. Furthermore, the alterations of metabolic pathways in malignant cells are heterogeneous and are defined by several factors including the tissue of origin, driving mutations, and microenvironment. In the present review, we discuss the key features of metabolic reprogramming and plasticity associated with different stages of tumor, from primary tumors to metastases. We also provide evidence of the successful usage of metabolic drugs in anticancer therapy. Finally, we highlight new promising targets for the development of new metabolic drugs.
Collapse
Affiliation(s)
- Oleg Shuvalov
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Alexandra Daks
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Olga Fedorova
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
| | - Alexey Petukhov
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
- Almazov National Medical Research Center, 197341 St-Petersburg, Russia
| | - Nickolai Barlev
- Institute of Cytology RAS, 194064 St-Petersburg, Russia; (O.S.); (A.D.); (O.F.); (A.P.)
- MIPT, 141701 Dolgoprudny, Moscow Region, Russia
- Orekhovich IBMC, 119435 Moscow, Russia
- Correspondence: ; Tel.: +7-812-297-4519
| |
Collapse
|
22
|
Possible Nutrition-Related Mechanisms of Metabolic Management in Cancer Treatment. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2021. [DOI: 10.5812/ijcm.107678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Context: Somatic mutation theory has been considered as a potential cause for cancer. However, major inconsistencies with the gene theory have necessitated serious reconsideration of this assumption. According to these inconsistencies, cancer may be considered as a metabolic disorder. According to the mitochondrial metabolic theory, substrate-level phosphorylation has been suggested to be superior to oxidative phosphorylation in cancer cells. Cancer metabolic therapies such as ketogenic diets (KD) and limitation in glutamine and calorie can be beneficial and are in line with this theory. In this study, we have reviewed the potential effects of KD as well as glutamine and calorie restriction in various types/stages of cancer with a focus on possible mechanisms. Evidence Acquisition: A comprehensive electronic search of different databases was performed using “cancer”, “ketogenic diet”, and “metabolic” as the main keywords. A comprehensive electronic search of different databases was performed using “cancer”, “ketogenic diet”, and “metabolic” as the main keywords. Results: Emerging evidence has indicated that KD can affect tumor cells by reducing glucose availability and simultaneous elevation of ketone bodies as non-fermentable metabolic fuels. KD has been suggested to be more effective as a non-toxic therapeutic measure in combination with glutamine targeting agents, chloroquine for lysosomal targeting, hyperbaric oxygen therapy, and calorie restriction. Conclusions: This metabolic approach can be considered as a promising non-toxic strategy for cancer management.
Collapse
|
23
|
Zhou Q, Fülöp A, Hopf C. Recent developments of novel matrices and on-tissue chemical derivatization reagents for MALDI-MSI. Anal Bioanal Chem 2020; 413:2599-2617. [PMID: 33215311 PMCID: PMC8007514 DOI: 10.1007/s00216-020-03023-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023]
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) is a fast-growing technique for visualization of the spatial distribution of the small molecular and macromolecular biomolecules in tissue sections. Challenges in MALDI-MSI, such as poor sensitivity for some classes of molecules or limited specificity, for instance resulting from the presence of isobaric molecules or limited resolving power of the instrument, have encouraged the MSI scientific community to improve MALDI-MSI sample preparation workflows with innovations in chemistry. Recent developments of novel small organic MALDI matrices play a part in the improvement of image quality and the expansion of the application areas of MALDI-MSI. This includes rationally designed/synthesized as well as commercially available small organic molecules whose superior matrix properties in comparison with common matrices have only recently been discovered. Furthermore, on-tissue chemical derivatization (OTCD) processes get more focused attention, because of their advantages for localization of poorly ionizable metabolites and their‚ in several cases‚ more specific imaging of metabolites in tissue sections. This review will provide an overview about the latest developments of novel small organic matrices and on-tissue chemical derivatization reagents for MALDI-MSI. Graphical abstract ![]()
Collapse
Affiliation(s)
- Qiuqin Zhou
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163, Mannheim, Germany
| | - Annabelle Fülöp
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163, Mannheim, Germany
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163, Mannheim, Germany.
| |
Collapse
|
24
|
Yan L, Zhao Z, Wang X, Lyu T, Li J, Qi Y, Wang X, Guo X. Short-term in vitro glutamine restriction differentially impacts the chromosomal stability of transformed and non-transformed cells. Mutagenesis 2020; 35:geaa026. [PMID: 33043986 DOI: 10.1093/mutage/geaa026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/10/2020] [Indexed: 11/13/2022] Open
Abstract
Glutamine (Gln) is a non-essential amino acid central for generating building blocks and cellular energy in tumours and rapidly proliferating non-transformed cells. However, the influence of Gln on regulating chromosomal stability of transformed and non-transformed cells remain poorly understand. We hypothesised that Gln is required for maintaining a homeostatic level of chromosomal stability. To this end, transformed cells HeLa and A375 and non-transformed cells NCM460 and HUVEC cells were intervened with varying concentrations of Gln (10, 1, 0.1 and 0.01 mM), with or without cisplatin (0.1 µg/ml), for 24 h. The cytokinesis-block micronucleus (MN) assay was used to determine chromosomal instability (CIN), the extent of which is reflected by the frequency of MN, nucleoplasmic bridge (NPB) and nuclear bud (NB). We demonstrated an unexpected decrease in the spontaneous rate of MN, but not NPB and NB, after Gln restriction in HeLa and A375 cells. Gln restriction reduced cisplatin-induced MN, but not NPB and NB, in HeLa and A375 cells. We further revealed that Gln restriction suppressed the proliferation of HeLa cells with high CIN induced by nocodazole, partially explaining why Gln restriction decreased the frequency of spontaneous and cisplatin-induced MN in transformed cells. In contrast, Gln restriction increased MN and NB, but not NPB, in NCM460 cells. In HUVEC cells, Gln restriction increased MN, NPB and NB. Meanwhile, Gln restriction sensitised NCM460 cells to cisplatin-induced genotoxicity. A similar but more pronounced pattern was observed in HUVEC cells. Collectively, these results suggest that the in vitro influences of Gln metabolism on CIN depend on cellular contexts: Transformed cells require high Gln to fine tune their CIN in an optimal rate to maximise genomic heterogeneity and fitness, whereas non-transformed cells need high Gln to prevent CIN.
Collapse
Affiliation(s)
- Ling Yan
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
| | - Ziru Zhao
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
| | - Xiaoran Wang
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
| | - Ting Lyu
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
| | - Jianfei Li
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
| | - Yanmei Qi
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
| | - Xu Wang
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Chenggong District, Kunming, Yunnan, China
- Yunnan Environmental Society, Chenggong District, Kunming, Yunnan, China
| | - Xihan Guo
- School of Life Sciences, Yunnan Normal University, Chenggong District, Kunming, Yunnan, China
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Chenggong District, Kunming, Yunnan, China
- Yunnan Environmental Society, Chenggong District, Kunming, Yunnan, China
| |
Collapse
|
25
|
Yoo HC, Yu YC, Sung Y, Han JM. Glutamine reliance in cell metabolism. Exp Mol Med 2020; 52:1496-1516. [PMID: 32943735 PMCID: PMC8080614 DOI: 10.1038/s12276-020-00504-8] [Citation(s) in RCA: 542] [Impact Index Per Article: 108.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022] Open
Abstract
As knowledge of cell metabolism has advanced, glutamine has been considered an important amino acid that supplies carbon and nitrogen to fuel biosynthesis. A recent study provided a new perspective on mitochondrial glutamine metabolism, offering mechanistic insights into metabolic adaptation during tumor hypoxia, the emergence of drug resistance, and glutaminolysis-induced metabolic reprogramming and presenting metabolic strategies to target glutamine metabolism in cancer cells. In this review, we introduce the various biosynthetic and bioenergetic roles of glutamine based on the compartmentalization of glutamine metabolism to explain why cells exhibit metabolic reliance on glutamine. Additionally, we examined whether glutamine derivatives contribute to epigenetic regulation associated with tumorigenesis. In addition, in discussing glutamine transporters, we propose a metabolic target for therapeutic intervention in cancer.
Collapse
Affiliation(s)
- Hee Chan Yoo
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, South Korea
| | - Ya Chun Yu
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, South Korea
| | - Yulseung Sung
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, South Korea
| | - Jung Min Han
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, South Korea.
- Department of Integrated OMICS for Biomedical Science, Yonsei University, Seoul, 03722, South Korea.
| |
Collapse
|
26
|
Multifaceted Aspects of Metabolic Plasticity in Human Cholangiocarcinoma: An Overview of Current Perspectives. Cells 2020; 9:cells9030596. [PMID: 32138158 PMCID: PMC7140515 DOI: 10.3390/cells9030596] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a deadly tumor without an effective therapy. Unique metabolic and bioenergetics features are important hallmarks of tumor cells. Metabolic plasticity allows cancer cells to survive in poor nutrient environments and maximize cell growth by sustaining survival, proliferation, and metastasis. In recent years, an increasing number of studies have shown that specific signaling networks contribute to malignant tumor onset by reprogramming metabolic traits. Several evidences demonstrate that numerous metabolic mediators represent key-players of CCA progression by regulating many signaling pathways. Besides the well-known Warburg effect, several other different pathways involving carbohydrates, proteins, lipids, and nucleic acids metabolism are altered in CCA. The goal of this review is to highlight the main metabolic processes involved in the cholangio-carcinogeneis that might be considered as potential novel druggable candidates for this disease.
Collapse
|