1
|
Yang Y, Pu J, Yang Y. Glycolysis and chemoresistance in acute myeloid leukemia. Heliyon 2024; 10:e35721. [PMID: 39170140 PMCID: PMC11336864 DOI: 10.1016/j.heliyon.2024.e35721] [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: 10/17/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
While traditional high-dose chemotherapy can effectively prolong the overall survival of acute myeloid leukemia (AML) patients and contribute to better prognostic outcomes, the advent of chemoresistance is a persistent challenge to effective AML management in the clinic. The therapeutic resistance is thought to emerge owing to the heterogeneous and adaptable nature of tumor cells when exposed to exogenous stimuli. Recent studies have focused on exploring metabolic changes that may afford novel opportunities to treat AML, with a particular focus on glycolytic metabolism. The Warburg effect, a hallmark of cancer, refers to metabolism of glucose through glycolysis under normoxic conditions, which contributes to the development of chemoresistance. Despite the key significance of this metabolic process in the context of malignant transformation, the underlying molecular mechanisms linking glycolysis to chemoresistance in AML remain incompletely understood. This review offers an overview of the current status of research focused on the relationship between glycolytic metabolism and AML resistance to chemotherapy, with a particular focus on the contributions of glucose transporters, key glycolytic enzymes, signaling pathways, non-coding RNAs, and the tumor microenvironment to this relationship. Together, this article will provide a foundation for the selection of novel therapeutic targets and the formulation of new approaches to treating AML.
Collapse
Affiliation(s)
- Yan Yang
- Department of Neonatology, Zigong Maternity and Child Health Care Hospital, Zigong, Sichuan, 643000, China
| | - Jianlin Pu
- Department of Psychiatry, The Zigong Affiliated Hospital of Southwest Medical University, Zigong mental health Center, Zigong Institute of Brain Science, Zigong, Sichuan, 643000, China
| | - You Yang
- Department of Pediatrics (Children Hematological Oncology), Birth Defects and Childhood Hematological Oncology Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
- The Second Hospital, Centre for Reproductive Medicine, Advanced Medical Research Institute, Key Laboratory for Experimental Teratology of the Ministry of Education, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China
| |
Collapse
|
2
|
Kamath A, Srinivasamurthy SK, Chowta MN, Ullal SD, Daali Y, Chakradhara Rao US. Role of Drug Transporters in Elucidating Inter-Individual Variability in Pediatric Chemotherapy-Related Toxicities and Response. Pharmaceuticals (Basel) 2022; 15:990. [PMID: 36015138 PMCID: PMC9415926 DOI: 10.3390/ph15080990] [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: 07/08/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Pediatric cancer treatment has evolved significantly in recent decades. The implementation of risk stratification strategies and the selection of evidence-based chemotherapy combinations have improved survival outcomes. However, there is large interindividual variability in terms of chemotherapy-related toxicities and, sometimes, the response among this population. This variability is partly attributed to the functional variability of drug-metabolizing enzymes (DME) and drug transporters (DTS) involved in the process of absorption, distribution, metabolism and excretion (ADME). The DTS, being ubiquitous, affects drug disposition across membranes and has relevance in determining chemotherapy response in pediatric cancer patients. Among the factors affecting DTS function, ontogeny or maturation is important in the pediatric population. In this narrative review, we describe the role of drug uptake/efflux transporters in defining pediatric chemotherapy-treatment-related toxicities and responses. Developmental differences in DTS and the consequent implications are also briefly discussed for the most commonly used chemotherapeutic drugs in the pediatric population.
Collapse
Affiliation(s)
- Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Mukta N. Chowta
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Sheetal D. Ullal
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Youssef Daali
- Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Uppugunduri S. Chakradhara Rao
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
3
|
Zuo F, Yu J, He X. Single-Cell Metabolomics in Hematopoiesis and Hematological Malignancies. Front Oncol 2022; 12:931393. [PMID: 35912231 PMCID: PMC9326066 DOI: 10.3389/fonc.2022.931393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Aberrant metabolism contributes to tumor initiation, progression, metastasis, and drug resistance. Metabolic dysregulation has emerged as a hallmark of several hematologic malignancies. Decoding the molecular mechanism underlying metabolic rewiring in hematological malignancies would provide promising avenues for novel therapeutic interventions. Single-cell metabolic analysis can directly offer a meaningful readout of the cellular phenotype, allowing us to comprehensively dissect cellular states and access biological information unobtainable from bulk analysis. In this review, we first highlight the unique metabolic properties of hematologic malignancies and underscore potential metabolic vulnerabilities. We then emphasize the emerging single-cell metabolomics techniques, aiming to provide a guide to interrogating metabolism at single-cell resolution. Furthermore, we summarize recent studies demonstrating the power of single-cell metabolomics to uncover the roles of metabolic rewiring in tumor biology, cellular heterogeneity, immunometabolism, and therapeutic resistance. Meanwhile, we describe a practical view of the potential applications of single-cell metabolomics in hematopoiesis and hematological malignancies. Finally, we present the challenges and perspectives of single-cell metabolomics development.
Collapse
|
4
|
Kansal R. Fructose Metabolism and Acute Myeloid Leukemia. EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2022; 7:25-38. [DOI: 10.14218/erhm.2021.00042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
5
|
Song ZB, Yu Y, Zhang GP, Li SQ. Genomic Instability of Mutation-Derived Gene Prognostic Signatures for Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:728574. [PMID: 34676211 PMCID: PMC8523793 DOI: 10.3389/fcell.2021.728574] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the major cancer-related deaths worldwide. Genomic instability is correlated with the prognosis of cancers. A biomarker associated with genomic instability might be effective to predict the prognosis of HCC. In the present study, data of HCC patients from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases were used. A total of 370 HCC patients from the TCGA database were randomly classified into a training set and a test set. A prognostic signature of the training set based on nine overall survival (OS)–related genomic instability–derived genes (SLCO2A1, RPS6KA2, EPHB6, SLC2A5, PDZD4, CST2, MARVELD1, MAGEA6, and SEMA6A) was constructed, which was validated in the test and TCGA and ICGC sets. This prognostic signature showed more accurate prediction for prognosis of HCC compared with tumor grade, pathological stage, and four published signatures. Cox multivariate analysis revealed that the risk score could be an independent prognostic factor of HCC. A nomogram that combines pathological stage and risk score performed well compared with an ideal model. Ultimately, paired differential expression profiles of genes in the prognostic signature were validated at mRNA and protein level using HCC and paratumor tissues obtained from our institute. Taken together, we constructed and validated a genomic instability–derived gene prognostic signature, which can help to predict the OS of HCC and help us to explore the potential therapeutic targets of HCC.
Collapse
Affiliation(s)
- Ze-Bing Song
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Yu
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guo-Pei Zhang
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shao-Qiang Li
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
SLC1A1 mediated glutamine addiction and contributed to natural killer T-cell lymphoma progression with immunotherapeutic potential. EBioMedicine 2021; 72:103614. [PMID: 34628354 PMCID: PMC8511843 DOI: 10.1016/j.ebiom.2021.103614] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Metabolic reprogramming plays an essential role on lymphoma progression. Dysregulation of glutamine metabolism is implicated in natural-killer T-cell lymphoma (NKTCL) and tumor cell response to asparaginase-based anti-metabolic treatment. METHODS To understand the metabolomic alterations and determine the potential therapeutic target of asparaginase, we assessed metabolomic profile using liquid chromatography-mass spectrometry in serum samples of 36 NKTCL patients, and integrated targeted metabolic analysis and RNA sequencing in tumor samples of 102 NKTCL patients. The biological function of solute carrier family 1 member 1 (SLC1A1) on metabolic flux, lymphoma cell growth, and drug sensitivity was further examined in vitro in NK-lymphoma cell line NK-92 and SNK-6, and in vivo in zebrafish xenograft models. FINDINGS In NKTCL patients, serum metabolomic profile was characterized by aberrant glutamine metabolism and SLC1A1 was identified as a central regulator of altered glutaminolysis. Both in vitro and in vivo, ectopic expression of SLC1A1 increased cellular glutamine uptake, enhanced glutathione metabolic flux, and induced glutamine addiction, leading to acceleration of cell proliferation and tumor growth. Of note, SLC1A1 overexpression was significantly associated with PD-L1 downregulation and reduced cytotoxic CD3+/CD8+ T cell activity when co-cultured with peripheral blood mononuclear cells. Asparaginase treatment counteracted SLC1A1-mediated glutamine addiction, restored SLC1A1-induced impaired T-cell immunity. Clinically, high EAAT3 (SLC1A1-encoded protein) expression independently predicted superior progression-free and overall survival in 90 NKTCL patients treated with asparaginase-based regimens. INTERPRETATION SLC1A1 functioned as an extracellular glutamine transporter, promoted tumor growth through reprogramming glutamine metabolism of NKTCL, while rendered tumor cells sensitive to asparaginase treatment. Moreover, SLC1A1-mediated modulation of PD-L1 expression might provide clinical rationale of co-targeting metabolic vulnerability and immunosuppressive microenvironment in NKTCL. FUNDING This study was supported, in part, by research funding from the National Natural Science Foundation of China (82130004, 81830007 and 81900192), Chang Jiang Scholars Program, Shanghai Municipal Education Commission Gaofeng Clinical Medicine Grant Support (20152206 and 20152208), Clinical Research Plan of SHDC (2020CR1032B), Multicenter Clinical Research Project by Shanghai Jiao Tong University School of Medicine (DLY201601), Shanghai Chenguang Program (19CG15), Shanghai Sailing Program (19YF1430800), Medical-Engineering Cross Foundation of Shanghai Jiao Tong University (ZH2018QNA46), and Shanghai Yi Yuan Xin Xing Program.
Collapse
|
7
|
Touzart A, Mayakonda A, Smith C, Hey J, Toth R, Cieslak A, Andrieu GP, Tran Quang C, Latiri M, Ghysdael J, Spicuglia S, Dombret H, Ifrah N, Macintyre E, Lutsik P, Boissel N, Plass C, Asnafi V. Epigenetic analysis of patients with T-ALL identifies poor outcomes and a hypomethylating agent-responsive subgroup. Sci Transl Med 2021; 13:13/595/eabc4834. [PMID: 34039737 DOI: 10.1126/scitranslmed.abc4834] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/10/2020] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
Adult "T cell" acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that is associated with poor outcomes, requiring additional therapeutic options. The DNA methylation landscapes of adult T-ALL remain undercharacterized. Here, we systematically analyzed the DNA methylation profiles of normal thymic-sorted T cell subpopulations and 143 primary adult T-ALLs as part of the French GRAALL 2003-2005 trial. Our results indicated that T-ALL is epigenetically heterogeneous consisting of five subtypes (C1-C5), which were either associated with co-occurring DNA methyltransferase 3 alpha (DNMT3A)/isocitrate dehydrogenase [NADP(+)] 2 (IDH2) mutations (C1), TAL bHLH transcription factor 1, erythroid differentiation factor (TAL1) deregulation (C2), T cell leukemia homeobox 3 (TLX3) (C3), TLX1/in cis-homeobox A9 (HOXA9) (C4), or in trans-HOXA9 overexpression (C5). Integrative analysis of DNA methylation and gene expression identified potential cluster-specific oncogenes and tumor suppressor genes. In addition to an aggressive hypomethylated subgroup (C1), our data identified an unexpected subset of hypermethylated T-ALL (C5) associated with poor outcome and primary therapeutic response. Using mouse xenografts, we demonstrated that hypermethylated T-ALL samples exhibited therapeutic responses to the DNA hypomethylating agent 5-azacytidine, which significantly (survival probability; P = 0.001 for C3, 0.01 for C4, and 0.0253 for C5) delayed tumor progression. These findings suggest that epigenetic-based therapies may provide an alternative treatment option in hypermethylated T-ALL.
Collapse
Affiliation(s)
- Aurore Touzart
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Université de Paris (Descartes), Institut Necker -Enfants Malades (INEM), Institut national de la santé et de la recherche médicale (Inserm) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, 75743 Paris, France
| | - Anand Mayakonda
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Charlotte Smith
- Université de Paris (Descartes), Institut Necker -Enfants Malades (INEM), Institut national de la santé et de la recherche médicale (Inserm) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, 75743 Paris, France
| | - Joschka Hey
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.,Germany-Israeli Helmholtz Research School in Cancer Biology, 69120 Heidelberg, Germany
| | - Reka Toth
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Agata Cieslak
- Université de Paris (Descartes), Institut Necker -Enfants Malades (INEM), Institut national de la santé et de la recherche médicale (Inserm) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, 75743 Paris, France
| | - Guillaume P Andrieu
- Université de Paris (Descartes), Institut Necker -Enfants Malades (INEM), Institut national de la santé et de la recherche médicale (Inserm) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, 75743 Paris, France
| | - Christine Tran Quang
- Institut Curie, Orsay, France.,CNRS UMR3348, Institut Curie, Orsay, France.,INSERM 1278, Centre Universitaire, Orsay, France.,PSL Research University, Paris, France.,Paris-Saclay, 91400 Orsay, France
| | - Mehdi Latiri
- Université de Paris (Descartes), Institut Necker -Enfants Malades (INEM), Institut national de la santé et de la recherche médicale (Inserm) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, 75743 Paris, France
| | - Jacques Ghysdael
- Institut Curie, Orsay, France.,CNRS UMR3348, Institut Curie, Orsay, France.,INSERM 1278, Centre Universitaire, Orsay, France.,PSL Research University, Paris, France.,Paris-Saclay, 91400 Orsay, France
| | - Salvatore Spicuglia
- Aix-Marseille University, Inserm, Theories and Approaches of Genomic Complexity (TAGC), Equipe labellisée Ligue, UMR1090, 13288 Marseille, France
| | - Hervé Dombret
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, 75010 Paris, France
| | - Norbert Ifrah
- PRES LUNAM, CHU Angers service des Maladies du Sang et INSERM U 892, 49933 Angers, France
| | - Elizabeth Macintyre
- Université de Paris (Descartes), Institut Necker -Enfants Malades (INEM), Institut national de la santé et de la recherche médicale (Inserm) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, 75743 Paris, France
| | - Pavlo Lutsik
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,German Cancer Research Consortium (DKTK), 69120 Heidelberg, Germany
| | - Nicolas Boissel
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, 75010 Paris, France
| | - Christoph Plass
- Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany. .,German Cancer Research Consortium (DKTK), 69120 Heidelberg, Germany
| | - Vahid Asnafi
- Université de Paris (Descartes), Institut Necker -Enfants Malades (INEM), Institut national de la santé et de la recherche médicale (Inserm) U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, 75743 Paris, France.
| |
Collapse
|
8
|
Lai Y, Sheng L, Wang J, Zhou M, OuYang G. A Novel 85-Gene Expression Signature Predicts Unfavorable Prognosis in Acute Myeloid Leukemia. Technol Cancer Res Treat 2021; 20:15330338211004933. [PMID: 33784904 PMCID: PMC8020099 DOI: 10.1177/15330338211004933] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: Acute myeloid leukemia (AML) is a heterogeneous disorder with complex genetic
basis and adverse prognosis. Cytogenetics risk, somatic mutations and gene
expression profiles are important prognostic factors for AML patients.
However, accurate stratification of patient prognosis remains an unsolved
problem in AML. This study was to to develop a novel gene profile to
accurately classify AML patients into subgroups with different survival
probabilities. Methods: Survival-related genes were determined by Kaplan–Meier survival analysis and
multivariate analysis using the expression and clinical data of 405 AML
patients from Oregon Health & Science University (OHSU) dataset and
validated in The Cancer Genome Atlas (TCGA) database. Feature selection was
performed by using the Least Absolute Shrinkage and Selection Operator
(LASSO) method. With the LASSO model, a prognostic 85-gene score was
established and compared with 2 known gene-expression risk scores. The
stratification of AML patients was performed by unsupervised hierarchical
clustering of 85 gene expression levels to identify clusters of AML patients
with different survival probabilities. Results: The LASSO model comprising 85 genes was considered as the optimal model based
on relatively high area under curve value (0.83) and the minimum mean
squared error. The 85-gene score was associated with increased mortality in
AML patients. Hierarchical clustering analysis of the 85 genes revealed 3
subgroups of AML patients in the OHSU dataset. The cluster1 AML patients
were associated with more female cases, higher percent of bone marrow blast
cells, 85-gene score, cytogenetics risk, more frequent FLT3-ITD,
DNMT3A, NP1 mutations, less frequent
TP53, RUNX1 mutations, poorer overall
survival than cluster2 tumors. The 85-gene score had higher AUC (0.75) than
the 5-gene risk score and LSC17 score (0.74 and 0.65). Conclusions: The 85-gene score is superior to the 2 established prognostic gene signatures
in the prediction of prognosis of AML patients.
Collapse
Affiliation(s)
- Yanli Lai
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Lixia Sheng
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Jiaping Wang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Miao Zhou
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Guifang OuYang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| |
Collapse
|
9
|
Liang RJ, Taylor S, Nahiyaan N, Song J, Murphy CJ, Dantas E, Cheng S, Hsu TW, Ramsamooj S, Grover R, Hwang SK, Ngo B, Cantley LC, Rhee KY, Goncalves MD. GLUT5 (SLC2A5) enables fructose-mediated proliferation independent of ketohexokinase. Cancer Metab 2021; 9:12. [PMID: 33762003 PMCID: PMC7992954 DOI: 10.1186/s40170-021-00246-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/08/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Fructose is an abundant source of carbon and energy for cells to use for metabolism, but only certain cell types use fructose to proliferate. Tumor cells that acquire the ability to metabolize fructose have a fitness advantage over their neighboring cells, but the proteins that mediate fructose metabolism in this context are unknown. Here, we investigated the determinants of fructose-mediated cell proliferation. METHODS Live cell imaging and crystal violet assays were used to characterize the ability of several cell lines (RKO, H508, HepG2, Huh7, HEK293T (293T), A172, U118-MG, U87, MCF-7, MDA-MB-468, PC3, DLD1 HCT116, and 22RV1) to proliferate in fructose (i.e., the fructolytic ability). Fructose metabolism gene expression was determined by RT-qPCR and western blot for each cell line. A positive selection approach was used to "train" non-fructolytic PC3 cells to utilize fructose for proliferation. RNA-seq was performed on parental and trained PC3 cells to find key transcripts associated with fructolytic ability. A CRISPR-cas9 plasmid containing KHK-specific sgRNA was transfected in 293T cells to generate KHK-/- cells. Lentiviral transduction was used to overexpress empty vector, KHK, or GLUT5 in cells. Metabolic profiling was done with seahorse metabolic flux analysis as well as LC/MS metabolomics. Cell Titer Glo was used to determine cell sensitivity to 2-deoxyglucose in media containing either fructose or glucose. RESULTS We found that neither the tissue of origin nor expression level of any single gene related to fructose catabolism determine the fructolytic ability. However, cells cultured chronically in fructose can develop fructolytic ability. SLC2A5, encoding the fructose transporter, GLUT5, was specifically upregulated in these cells. Overexpression of GLUT5 in non-fructolytic cells enabled growth in fructose-containing media across cells of different origins. GLUT5 permitted fructose to flux through glycolysis using hexokinase (HK) and not ketohexokinase (KHK). CONCLUSIONS We show that GLUT5 is a robust and generalizable driver of fructose-dependent cell proliferation. This indicates that fructose uptake is the limiting factor for fructose-mediated cell proliferation. We further demonstrate that cellular proliferation with fructose is independent of KHK.
Collapse
Affiliation(s)
- Roger J Liang
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Samuel Taylor
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-I MD-PhD program, New York, NY, 10065, USA
| | - Navid Nahiyaan
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Junho Song
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Charles J Murphy
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shuyuan Cheng
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ting-Wei Hsu
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shakti Ramsamooj
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Rahul Grover
- Weill Cornell Medical College, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Seo-Kyoung Hwang
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Bryan Ngo
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kyu Y Rhee
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
10
|
Chen C, Hao X, Lai X, Liu L, Zhu J, Shao H, Huang D, Gu H, Zhang T, Yu Z, Xie L, Zhang X, Yang Y, Xu J, Zhao Y, Lu Z, Zheng J. Oxidative phosphorylation enhances the leukemogenic capacity and resistance to chemotherapy of B cell acute lymphoblastic leukemia. SCIENCE ADVANCES 2021; 7:eabd6280. [PMID: 33692103 PMCID: PMC7946372 DOI: 10.1126/sciadv.abd6280] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 01/22/2021] [Indexed: 05/11/2023]
Abstract
How metabolic status controls the fates of different types of leukemia cells remains elusive. Using a SoNar-transgenic mouse line, we demonstrated that B cell acute lymphoblastic leukemia (B-ALL) cells had a preference in using oxidative phosphorylation. B-ALL cells with a low SoNar ratio (SoNar-low) had enhanced mitochondrial respiration capacity, mainly resided in the vascular niche, and were enriched with more functional leukemia-initiating cells than that of SoNar-high cells in a murine B-ALL model. The SoNar-low cells were more resistant to cytosine arabinoside (Ara-C) treatment. cyclic adenosine 3',5'-monophosphate response element-binding protein transactivated pyruvate dehydrogenase complex component X and cytidine deaminase to maintain the oxidative phosphorylation level and Ara-C-induced resistance. SoNar-low human primary B-ALL cells also had a preference for oxidative phosphorylation. Suppressing oxidative phosphorylation with several drugs sufficiently attenuated Ara-C-induced resistance. Our study provides a unique angle for understanding the potential connections between metabolism and B-ALL cell fates.
Collapse
Affiliation(s)
- Chiqi Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoxin Hao
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoyun Lai
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ligen Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun Zhu
- Department of Hematology, Shanghai Zhaxin Hospital, Shanghai 200434, China
| | - Hongfang Shao
- Center of Reproductive Medicine, Shanghai Sixth People's Hospital, 600 Yishan Road Shanghai 200233, China
| | - Dan Huang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hao Gu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tinghua Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhuo Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Xie
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaocui Zhang
- Department of Hematology, Shanghai Zhaxin Hospital, Shanghai 200434, China
| | - Yi Yang
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Yuzheng Zhao
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhigang Lu
- The Fifth People's Hospital of Shanghai, Fudan University, and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
11
|
Zeng T, Fedeli MA, Tanda F, Wang Y, Yang D, Xue B, Jia L, Palmieri G, Sechi LA, Kelvin DJ. Whole-exome Sequencing of Prostate Cancer in Sardinian Identify Recurrent UDP-glucuronosyltransferase Amplifications. J Cancer 2021; 12:438-450. [PMID: 33391440 PMCID: PMC7738997 DOI: 10.7150/jca.48433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/25/2020] [Indexed: 02/05/2023] Open
Abstract
Globally, prostate cancer is the third most common cancer in the world, and the second most common cancer in men. However, rates for incidence and mortality vary considerably with race, ethnicity, and geography. Over 97 significantly mutated genes that have been identified in prostate cancer; however, a lack of genomic prostate cancer studies focusing on different racial and ethnic groups and racial mixing pose a serious challenge to universalize these findings. The Sardinian population is an isolated Mediterranean population that has a high frequency of centenarians and a much lower incidence of prostate cancer than found in males in mainland Europe. Here, we conducted a genomic prostate cancer study on a Sardinian cohort diagnosed with local prostate cancer. Our data reveals a low rate of ERG fusion in Sardinian prostate cancer. Interestingly, we identified a novel BTBD7-SLC2A5 fusion that occurred in 13% of the patients. We also found that the UGT2B4 on 4q13.2 was amplified in 20% of the Sardinian patients but rarely amplified in patients of other population. These observations underscore the importance of the inter-population molecular heterogeneity of prostate cancer. In addition, we examined the expression of UGT2B4 in 497 prostate cancer patients derived from The Cancer Genome Atlas database. We found that high expression of UGT2B4 was associated with low-grade prostate cancer and upregulation of UGT2B4 in tumors was associated with upregulation of metabolism pathways such as 'de novo' IMP biosynthetic process, glutamine and monocarboxylic acid metabolism. These data provide insight into clinical relevance and functional mechanism of UGT2B4. Further understanding functional mechanism of UGT2B4 amplification and BTBD7-SLC2A5 fusion will aid in developing drugs to benefit the prostate cancer patients.
Collapse
Affiliation(s)
- Tiansheng Zeng
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou Guangdong, China
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Maria Antonietta Fedeli
- Department of Scienze Mediche Chirurgiche e Sperimentali, first affiliated Hospital of 33445Sassari University
| | - Francesco Tanda
- Department of Scienze Mediche Chirurgiche e Sperimentali, first affiliated Hospital of 33445Sassari University
| | - Yuyong Wang
- Department of Urology, affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, China
| | - Dongsheng Yang
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou Guangdong, China
| | - Bei Xue
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou Guangdong, China
| | - Lisha Jia
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou Guangdong, China
| | - Giuseppe Palmieri
- Institute of Genetic and Biomedical Research (IRGB), Head, National Research Council (CNR), 07100 Sassari, Italy
| | - Leonardo A Sechi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- ✉ Corresponding authors: J. Kelvin, E-mail: ; and Leonardo A. Sechi, E-mail: . Co-corresponding authors equally contributed to this work
| | - David J. Kelvin
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou Guangdong, China
- Department of Scienze Mediche Chirurgiche e Sperimentali, first affiliated Hospital of 33445Sassari University
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
- Canadian Center for Vaccinology, IWK, Halifax, Nova Scotia, Canada
- ✉ Corresponding authors: J. Kelvin, E-mail: ; and Leonardo A. Sechi, E-mail: . Co-corresponding authors equally contributed to this work
| |
Collapse
|
12
|
Lai B, Lai Y, Zhang Y, Zhou M, Sheng L, OuYang G. The Solute Carrier Family 2 Genes Are Potential Prognostic Biomarkers in Acute Myeloid Leukemia. Technol Cancer Res Treat 2020; 19:1533033819894308. [PMID: 31918632 PMCID: PMC7099669 DOI: 10.1177/1533033819894308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aims: The solute carrier family 2 (SLC2) genes are comprised of 14 members which are essential for the maintenance of glucose uptake and survival of tumour cells. This study was performed to investigate the associations of SLC2 family gene expression with mortality in acute myeloid leukemia (AML). Methods: Clinical features and SLC2 family gene expression data were obtained from The Cancer Genome Atlas and Gene Expression Omnibus database. The associations between SLC2 family gene expression and clinicopathologic features were analyzed using linear regression model. Kaplan-Meier survival, univariate, multivariate survival analyses and validation analysis were performed to analyze the associations between SLC2 family gene expression and patients’ overall survival. Results: Patient mortality was positively associated with age and cytogenetic risk in AML patients. Kaplan-Meier survival analysis suggested that patients with high SLC2A5 and SLC2A10 expression showed poorer survival than those with low SLC2A5 and SLC2A10 expression. In contrast, patients with high SLC2A13 expression exhibited better prognosis than those with low SLC2A13 expression (P < 0.05 for all cases, log rank test). Multivariate survival analysis and validation analysis confirmed that high expression of SLC2A5 and SLC2A10 and low expression of SLC2A13 were associated with increased mortality (P = 0.00, Odd ratio [OR]:4.05, 95% Confidence Interval [CI]: 1.73-10.22; P = 0.00, OR: 3.66, 95% CI: 1.54-9.25; and P = 0.01, OR: 0.26, 95% CI: 0.09-0.68, respectively). Conclusion: SLC family gene expression, such as SLC2A5, SLC2A10 and SLC2A13, was significantly associated with prognosis of AML patients, their expression levels might become useful prognostic biomarkers in AML.
Collapse
Affiliation(s)
- Binbin Lai
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Yanli Lai
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Yanli Zhang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Miao Zhou
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Lixia Sheng
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| | - Guifang OuYang
- Department of Hematology, Ningbo First Hospital, Ningbo, Zhejiang Province, China
| |
Collapse
|
13
|
Jin C, Gong X, Shang Y. GLUT5 increases fructose utilization in ovarian cancer. Onco Targets Ther 2019; 12:5425-5436. [PMID: 31371983 PMCID: PMC6635899 DOI: 10.2147/ott.s205522] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/20/2019] [Indexed: 01/01/2023] Open
Abstract
Background: Fructose is one of the most common dietary carbohydrates in the whole world, and recent studies have found that fructose consumption is closely related to the oncogenesis and development of tumors, however, very few studies have focused on the fructose in ovarian cancer. GLUT5 (Glucose transporter type 5), as a specific fructose transporter in mammalian cells, has also been found highly expressed in many cancers. Methods: In this study, we investigated the abilities of proliferation, colony formation, and migration of ovarian cancer cells in fructose medium, and then silenced GLUT5 in ovarian cancer cells to explore the role GLUT5 in fructose metabolism in ovarian cancer. Results: The results showed that the ovarian cancer cells had similar abilities of proliferation and migration in fructose medium and glucose medium, but silencing GLUT5 could significantly inhibit these abilities in fructose medium. Meanwhile, we found that GLUT5 was higher expressed in ovarian cancer tissues, and its expression correlated significantly with tumor malignancy and poor survival of ovarian cancer patients. Furthermore, the results of animal experiments also demonstrated that intake too much fructose could prominently increase tumor volume, and silencing GLUT5 could significantly inhibit tumor proliferation. Conclusion: In conclusion, we demonstrate that ovarian cancer cells could utilize fructose for their growth, and restricting the fructose intake or targeting GLUT5 may be efficacious strategies for ovarian cancer therapy.
Collapse
Affiliation(s)
- Cuiping Jin
- Department of Gynaecology and Obstetrics, Tianjin Hospital, Tianjin 300211, People's Republic of China
| | - Xiaojin Gong
- Department of Gynaecology and Obstetrics, Tianjin Hospital, Tianjin 300211, People's Republic of China
| | - Yumin Shang
- Department of Gynaecology and Obstetrics, Tianjin Hospital, Tianjin 300211, People's Republic of China
| |
Collapse
|