1
|
Sokei J, Kanefsky J, Sykes SM. Reprogramming of Fatty Acid Metabolism in Acute Leukemia. J Cell Physiol 2025; 240:e70000. [PMID: 39835485 DOI: 10.1002/jcp.70000] [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: 05/08/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Fatty acids are essential biomolecules that support several cellular processes, such as membrane structures, energy storage and production, as well as signal transduction. Accordingly, changes in fatty acid metabolism can have a significant impact on cell behavior, such as growth, survival, proliferation, differentiation, and motility. Therefore, it is not surprising that many aspects of fatty acid metabolism are frequently dysregulated in human cancer, including in highly aggressive blood cancers such as acute leukemia. The aims of this review are to summarize the aspects of fatty acid metabolism that are specifically coopted in acute leukemia as well as current preclinical strategies for targeting fatty acid metabolism in these cancers.
Collapse
Affiliation(s)
- Judith Sokei
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University in Saint Louis, St. Louis, Missouri, USA
| | - Joice Kanefsky
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University Health System, Philadelphia, Pennsylvania, USA
| | - Stephen M Sykes
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University in Saint Louis, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Nisticò C, Chiarella E. An Overview on Lipid Droplets Accumulation as Novel Target for Acute Myeloid Leukemia Therapy. Biomedicines 2023; 11:3186. [PMID: 38137407 PMCID: PMC10741140 DOI: 10.3390/biomedicines11123186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic reprogramming is a key alteration in tumorigenesis. In cancer cells, changes in metabolic fluxes are required to cope with large demands on ATP, NADPH, and NADH, as well as carbon skeletons. In particular, dysregulation in lipid metabolism ensures a great energy source for the cells and sustains cell membrane biogenesis and signaling molecules, which are necessary for tumor progression. Increased lipid uptake and synthesis results in intracellular lipid accumulation as lipid droplets (LDs), which in recent years have been considered hallmarks of malignancies. Here, we review current evidence implicating the biogenesis, composition, and functions of lipid droplets in acute myeloid leukemia (AML). This is an aggressive hematological neoplasm originating from the abnormal expansion of myeloid progenitor cells in bone marrow and blood and can be fatal within a few months without treatment. LD accumulation positively correlates with a poor prognosis in AML since it involves the activation of oncogenic signaling pathways and cross-talk between the tumor microenvironment and leukemic cells. Targeting altered LD production could represent a potential therapeutic strategy in AML. From this perspective, we discuss the main inhibitors tested in in vitro AML cell models to block LD formation, which is often associated with leukemia aggressiveness and which may find clinical application in the future.
Collapse
Affiliation(s)
- Clelia Nisticò
- Candiolo Cancer Institute, FPO-IRCCS, Department of Oncology, University of Torino, 10124 Candiolo, Italy
| | - Emanuela Chiarella
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy
| |
Collapse
|
3
|
Ramos Z, Garrick DJ, Blair HT, Vera B, Ciappesoni G, Kenyon PR. Genomic Regions Associated with Wool, Growth and Reproduction Traits in Uruguayan Merino Sheep. Genes (Basel) 2023; 14:167. [PMID: 36672908 PMCID: PMC9858812 DOI: 10.3390/genes14010167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The aim of this study was to identify genomic regions and genes associated with the fiber diameter (FD), clean fleece weight (CFW), live weight (LW), body condition score (BCS), pregnancy rate (PR) and lambing potential (LP) of Uruguayan Merino sheep. Phenotypic records of approximately 2000 mixed-age ewes were obtained from a Merino nucleus flock. Genome-wide association studies were performed utilizing single-step Bayesian analysis. For wool traits, a total of 35 genomic windows surpassed the significance threshold (PVE ≥ 0.25%). The proportion of the total additive genetic variance explained by those windows was 4.85 and 9.06% for FD and CFW, respectively. There were 42 windows significantly associated with LWM, which collectively explained 43.2% of the additive genetic variance. For BCS, 22 relevant windows accounted for more than 40% of the additive genetic variance, whereas for the reproduction traits, 53 genomic windows (24 and 29 for PR and LP, respectively) reached the suggestive threshold of 0.25% of the PVE. Within the top 10 windows for each trait, we identified several genes showing potential associations with the wool (e.g., IGF-1, TGFB2R, PRKCA), live weight (e.g., CAST, LAP3, MED28, HERC6), body condition score (e.g., CDH10, TMC2, SIRPA, CPXM1) or reproduction traits (e.g., ADCY1, LEPR, GHR, LPAR2) of the mixed-age ewes.
Collapse
Affiliation(s)
- Zully Ramos
- School of Agriculture and Environment, Massey University, Palmerston North 4410, New Zealand
| | - Dorian J. Garrick
- School of Agriculture and Environment, Massey University, Palmerston North 4410, New Zealand
| | - Hugh T. Blair
- School of Agriculture and Environment, Massey University, Palmerston North 4410, New Zealand
| | - Brenda Vera
- National Research Program on Meat and Wool Production, Instituto Nacional de Investigación Agropecuaria, INIA Las Brujas, Ruta 48 Km 10, Canelones 90100, Uruguay
| | - Gabriel Ciappesoni
- National Research Program on Meat and Wool Production, Instituto Nacional de Investigación Agropecuaria, INIA Las Brujas, Ruta 48 Km 10, Canelones 90100, Uruguay
| | - Paul R. Kenyon
- School of Agriculture and Environment, Massey University, Palmerston North 4410, New Zealand
| |
Collapse
|
4
|
High Expression of ACOT2 Predicts Worse Overall Survival and Abnormal Lipid Metabolism: A Potential Target for Acute Myeloid Leukemia. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2669114. [PMID: 36193167 PMCID: PMC9525752 DOI: 10.1155/2022/2669114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022]
Abstract
Acyl-CoA thioesterase (ACOT) plays a considerable role in lipid metabolism, which is closely related to the occurrence and development of cancer, nevertheless, its role has not been fully elucidated in acute myeloid leukemia (AML). To explore the role of ACOT2 in AML and to provide a potential therapeutic target for AML, the expression pattern of ACOT was investigated based on the TNMplot, Gene Expression Profiling Interactive Analysis (GEPIA), and Cancer Cell Line Encyclopedia (CCLE) database, and diagnostic value, prognostic value, and clinical phenotype of ACOT were explored based on data from The Cancer Genome Atlas (TCGA). Functional annotation and enrichment analysis of the common targets between ACOT2 coexpressed and AML-related genes were further performed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) analyses. The protein-protein interaction (PPI) network of ACOT2 coexpressed genes and functional ACOT2-related metabolites association network were constructed based on GeneMANIA and Human Metabolome Database. Among ACOTs, ACOT2 was highly expressed in AML compared to normal control subjects according to TNMplot, GEPIA, and CCLE database, which was significantly associated with poor overall survival (OS) in AML (
). Moreover, ACOT2 exhibited excellent diagnostic efficiency for AML (AUC: 1.000) and related to French-American-British (FAB) classification and cytogenetics. GO, KEGG, and GSEA analyses of 71 common targets between ACOT2 coexpressed and AML-related genes revealed that ACOT2 is closely related to ACOT1, ACOT4, enoyl-acyl carrier protein reductase, mitochondrial (MECR), puromycin-sensitive aminopeptidase (NPEPPS), SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1), and long-chain fatty acid-CoA ligase 1 (ACSL1) in PPI network, and plays a significant role in lipid metabolism, that is, involved in fatty acid elongation and biosynthesis of unsaturated fatty acids. Collectively, the increase of ACOT2 may be an important characteristic of worse OS and abnormal lipid metabolism, suggesting that ACOT2 may become a potential therapeutic target for AML.
Collapse
|
5
|
Li D, Liang J, Yang W, Guo W, Song W, Zhang W, Wu X, He B. A distinct lipid metabolism signature of acute myeloid leukemia with prognostic value. Front Oncol 2022; 12:876981. [PMID: 35957912 PMCID: PMC9359125 DOI: 10.3389/fonc.2022.876981] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy characterized by extensive genetic abnormalities that might affect the prognosis and provide potential drug targets for treatment. Reprogramming of lipid metabolism plays important roles in tumorigenesis and progression and has been newly recognized a new hallmark of malignancy, and some related molecules in the signal pathways could be prognostic biomarkers and potential therapeutic targets for cancer treatment. However, the clinical value of lipid metabolism reprogramming in AML has not been systematically explored. In this study, we aim to explore the clinical value of lipid metabolism reprogramming and develop a prognostic risk signature for AML. Methods We implemented univariate Cox regression analysis to identify the prognosis-related lipid metabolism genes, and then performed LASSO analysis to develop the risk signature with six lipid metabolism-related genes (LDLRAP1, PNPLA6, DGKA, PLA2G4A, CBR1, and EBP). The risk scores of samples were calculated and divided into low- and high-risk groups by the median risk score. Results Survival analysis showed the high-risk group hold the significantly poorer outcomes than the low-risk group. The signature was validated in the GEO datasets and displayed a robust prognostic value in the stratification analysis. Multivariate analysis revealed the signature was an independent prognostic factor for AML patients and could serve as a potential prognostic biomarker in clinical evaluation. Furthermore, the risk signature was also found to be closely related to immune landscape and immunotherapy response in AML. Conclusions Overall, we conducted a comprehensive analysis of lipid metabolism in AML and constructed a risk signature with six genes related to lipid metabolism for the malignancy, prognosis, and immune landscape of AML, and our study might contribute to better understanding in the use of metabolites and metabolic pathways as the potential prognostic biomarkers and therapeutic targets for AML.
Collapse
Affiliation(s)
- Ding Li
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiaming Liang
- Department of Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Yang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Wenbin Guo
- Department of Pathology, Pingtan Comprehensive Experimental Area Hospital, Fuzhou, China
| | - Wenping Song
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xuan Wu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People’s Hospital, Zhengzhou, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- *Correspondence: Baoxia He, ; Xuan Wu,
| | - Baoxia He
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Baoxia He, ; Xuan Wu,
| |
Collapse
|
6
|
Delaidelli A, Dunham C, Santi M, Negri GL, Triscott J, Zheludkova O, Golanov A, Ryzhova M, Okonechnikov K, Schrimpf D, Stichel D, Ellison DW, von Deimling A, Kool M, Pfister SM, Ramaswamy V, Korshunov A, Taylor MD, Sorensen PH. Clinically Tractable Outcome Prediction of non-WNT/non-SHH Medulloblastoma Based on TPD52 Immunohistochemistry in a Multicohort Study. Clin Cancer Res 2021; 28:116-128. [PMID: 34702771 DOI: 10.1158/1078-0432.ccr-21-2057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/12/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE International consensus and the 2021 WHO classification recognize eight molecular subgroups among non-WNT/non-SHH (Group 3/4) medulloblastoma, representing ~60% of tumors. However, very few clinical centers worldwide possess the technical capabilities to determine DNA-methylation profiles or other molecular parameters of high-risk for Group 3/4 tumors. As a result, biomarker-driven risk stratification and therapy assignment constitutes a major challenge in medulloblastoma research. Here, we identify an immunohistochemistry (IHC) marker as a clinically tractable method for improved medulloblastoma risk stratification. EXPERIMENTAL DESIGN We bioinformatically analyzed published medulloblastoma transcriptomes and proteomes identifying as a potential biomarker TPD52, whose IHC prognostic value was validated across three Group 3/4 medulloblastoma clinical cohorts (n = 387) treated with conventional therapies. RESULTS TPD52 IHC positivity represented a significant independent predictor of early relapse and death for Group 3/4 medulloblastoma (HRs between 3.67-26.7 [95% CIs between 1.00-706.23], p = 0.05, 0.017 and 0.0058). Cross-validated survival models incorporating TPD52 IHC with clinical features outperformed existing state-of-the-art risk stratification schemes, and reclassified ~50% of patients into more appropriate risk categories. Finally, TPD52 immunopositivity was a predictive indicator of poor response to chemotherapy (HR 12.66 [95% CI 3.53-45.40], p < 0.0001), suggesting important implication for therapeutic choices. CONCLUSIONS The current study redefines the approach to risk stratification in Group 3/4 medulloblastoma in global practice. Since integration of TPD52 IHC in classification algorithms significantly improved outcome prediction, this test could be rapidly adopted for risk stratification on a global scale, independently of advanced but technically challenging molecular profiling techniques.
Collapse
Affiliation(s)
- Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher Dunham
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, Children's and Women's Health Centre of British Columbia, and University of British Columbia, Vancouver, British Columbia, Canada
| | - Mariarita Santi
- Department of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gian Luca Negri
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
| | - Joanna Triscott
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Olga Zheludkova
- St Luka's Clinical Research Center for Children, Moscow, Russian Federation
| | - Andrey Golanov
- Neurosurgical NN Burdenko Institute, Moscow, Russian Federation
| | - Marina Ryzhova
- Neurosurgical NN Burdenko Institute, Moscow, Russian Federation
| | - Konstantin Okonechnikov
- Hopp Children's Cancer Center Heidelberg (KiTZ), Division of Pediatric Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)
| | - Daniel Schrimpf
- Department of Neuropathology of Heidelberg University and CCU Neuropathology, German Cancer Research Center, Heidelberg, Germany
| | - Damian Stichel
- Department of Neuropathology of Heidelberg University and CCU Neuropathology, German Cancer Research Center, Heidelberg, Germany
| | - David W Ellison
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Andreas von Deimling
- Department of Neuropathology of Heidelberg University and CCU Neuropathology, German Cancer Research Center, Heidelberg, Germany
| | - Marcel Kool
- Hopp Children's Cancer Center Heidelberg (KiTZ), Division of Pediatric Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Division of Pediatric Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)
- Department of Pediatric Hematology and Oncology, University Hospital, Heidelberg, Germany
| | - Vijay Ramaswamy
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrey Korshunov
- Department of Neuropathology of Heidelberg University and CCU Neuropathology, German Cancer Research Center, Heidelberg, Germany
| | - Michael D Taylor
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Loss-of-function mutations in the histone methyltransferase EZH2 promote chemotherapy resistance in AML. Sci Rep 2021; 11:5838. [PMID: 33712646 PMCID: PMC7955088 DOI: 10.1038/s41598-021-84708-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Chemotherapy resistance is the main impediment in the treatment of acute myeloid leukaemia (AML). Despite rapid advances, the various mechanisms inducing resistance development remain to be defined in detail. Here we report that loss-of-function mutations (LOF) in the histone methyltransferase EZH2 have the potential to confer resistance against the chemotherapeutic agent cytarabine. We identify seven distinct EZH2 mutations leading to loss of H3K27 trimethylation via multiple mechanisms. Analysis of matched diagnosis and relapse samples reveal a heterogenous regulation of EZH2 and a loss of EZH2 in 50% of patients. We confirm that loss of EZH2 induces resistance against cytarabine in the cell lines HEK293T and K562 as well as in a patient-derived xenograft model. Proteomics and transcriptomics analysis reveal that resistance is conferred by upregulation of multiple direct and indirect EZH2 target genes that are involved in apoptosis evasion, augmentation of proliferation and alteration of transmembrane transporter function. Our data indicate that loss of EZH2 results in upregulation of its target genes, providing the cell with a selective growth advantage, which mediates chemotherapy resistance.
Collapse
|
8
|
Lee S, Suh HB, Choi SJ, Kang J, Kang JW, Kwon EJ, Kim HJ, Kim YH, Shin K. Identification of prognostic mRNAs in metastatic cutaneous melanoma. Melanoma Res 2020; 30:543-547. [PMID: 33003118 DOI: 10.1097/cmr.0000000000000697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cutaneous melanoma is the most common cause of skin cancer-related deaths worldwide. There is an urgent need to identify prognostic biomarkers to facilitate decision-making for treatment of metastatic cutaneous melanoma. Gene expression microarrays and RNA-seq technology have recently improved or changed current prognostic and therapeutic strategies for several cancers. However, according to the current melanoma staging system, prognosis is almost entirely dependent on clinicopathological features. To identify novel prognostic biomarkers, we investigated gene expression and clinical data for patients with cutaneous melanoma from three cohorts of The Cancer Genome Atlas and Gene Expression Omnibus. Kaplan-Meier survival analysis using median values of each gene as cutoff value revealed that nine genes (ABCC3, CAPS2, CCR6, CDCA8, CLU, DPF1, PTK2B, SATB1, and SYNE1) were statistically significant prognostic biomarkers of metastatic cutaneous melanoma in all three independent cohorts. Low expression of two genes (CDCA8 and DPF1) and high expression of seven genes (ABCC3, CAPS2, CCR6, CLU, PTK2B, SATB1, and SYNE) were significantly associated with positive metastatic cutaneous melanoma prognoses. In conclusion, we suggest nine novel prognostic biomarkers for cutaneous metastatic melanoma.
Collapse
Affiliation(s)
| | - Hie Bum Suh
- Department of Radiology, School of Medicine, Pusan National University
- Biomedical Research Institute, Pusan National University Hospital
| | | | | | | | | | | | - Yun Hak Kim
- Department of Biomedical Informatics
- Department of Anatomy, School of Medicine, Pusan National University
| | - Kihyuk Shin
- Biomedical Research Institute, Pusan National University Hospital
- Department of Dermatology
- Department of Dermatology, Pusan National University Yangsan Hospital
- Research Institute for Convergence of Biomedical Science and Technology, Yangsan, Korea
| |
Collapse
|
9
|
Kang JW, Kim Y, Lee Y, Myung K, Kim YH, Oh CK. AML poor prognosis factor, TPD52, is associated with the maintenance of haematopoietic stem cells through regulation of cell proliferation. J Cell Biochem 2020; 122:403-412. [PMID: 33166425 DOI: 10.1002/jcb.29869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/12/2020] [Accepted: 10/23/2020] [Indexed: 11/05/2022]
Abstract
Acute myeloid leukaemia (AML) is a blood cancer where undifferentiated myeloid cells are increased in the bone marrow and peripheral blood. As AML is dangerous and shows poor prognosis, many researchers categorised the relevant cytogenetic factors according to risk and prognosis. However, the specific reasons for poor cytogenetic factors remain unknown. We analysed a large data set from AML patients and found that TPD52 expression is elevated in patient groups with poor cytogenetic factors. As the amino acid sequence of TPD52 is evolutionally conserved in vertebrates, zebrafish embryos were used to investigate the function of TPD52. Since myeloid-biased haematopoietic stem cells (HSCs) are relevant to AML, the function of TPD52 in the development of HSCs was investigated. We determined that the zebrafish paralog, tpd52, is important for the maintenance of HSCs through regulation of cell proliferation. As tpd52 is linked to cell proliferation in zebrafish embryos, the proliferation-related gene, CD59, was correlated to TPD52 in every AML cohort with a high correlation coefficient. We suggest that TPD52 can be a novel therapeutic target for AML patients with poor cytogenetic factors. Additionally, more studies between TPD52 and CD59 will further increase the value of TPD52 as a novel target.
Collapse
Affiliation(s)
- Ji Wan Kang
- Interdisplinary Program of Genomic Science, Pusan National University, Yangsan, Republic of Korea
| | - Youngjoo Kim
- Interdisplinary Program of Genomic Science, Pusan National University, Yangsan, Republic of Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea.,Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Chang-Kyu Oh
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| |
Collapse
|
10
|
Endogenous retroviruses are a source of enhancers with oncogenic potential in acute myeloid leukaemia. Nat Commun 2020; 11:3506. [PMID: 32665538 PMCID: PMC7360734 DOI: 10.1038/s41467-020-17206-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterised by a series of genetic and epigenetic alterations that result in deregulation of transcriptional networks. One understudied source of transcriptional regulators are transposable elements (TEs), whose aberrant usage could contribute to oncogenic transcriptional circuits. However, the regulatory influence of TEs and their links to AML pathogenesis remain unexplored. Here we identify six endogenous retrovirus (ERV) families with AML-associated enhancer chromatin signatures that are enriched in binding of key regulators of hematopoiesis and AML pathogenesis. Using both locus-specific genetic editing and simultaneous epigenetic silencing of multiple ERVs, we demonstrate that ERV deregulation directly alters the expression of adjacent genes in AML. Strikingly, deletion or epigenetic silencing of an ERV-derived enhancer suppresses cell growth by inducing apoptosis in leukemia cell lines. This work reveals that ERVs are a previously unappreciated source of AML enhancers that may be exploited by cancer cells to help drive tumour heterogeneity and evolution. Transposable elements are a potential source of transcriptional regulators, but how these sequences contribute to oncogenesis remains poorly understood. Here, the authors identify endogenous retroviruses (ERVs) with acute myeloid leukemia (AML)-associated enhancer chromatin signatures, and provide evidence that ERV activation provides an additional layer of gene regulation in AML.
Collapse
|
11
|
Wang Y, Hu F, Li JY, Nie RC, Chen SL, Cai YY, Shu LL, Deng DJ, Xu JB, Liang Y. Systematic Construction and Validation of a Metabolic Risk Model for Prognostic Prediction in Acute Myelogenous Leukemia. Front Oncol 2020; 10:540. [PMID: 32373530 PMCID: PMC7186449 DOI: 10.3389/fonc.2020.00540] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/25/2020] [Indexed: 01/11/2023] Open
Abstract
Background: Acute myelogenous leukemia (AML) is a heterogeneous disease with recurrent gene mutations and variations in disease-associated gene expression, which may be useful for prognostic prediction. Methods: RNA matrix and clinical data of AML were downloaded from GEO, TCGA, and TARGET databases. Prognostic metabolic genes were identified by LASSO analysis to establish a metabolic model. Prognostic accuracy of the model was quantified by time-dependent receiver operating characteristic curves and the area under the curve (AUC). Survival analysis was performed by log-rank tests. Enriched pathways in different metabolic risk statuses were evaluated by gene set enrichment analyses (GSEA). Results: We identified nine genes to construct a prognostic model of shorter survival in the high-risk vs. low-risk group. The prognostic model showed good predictive efficacy, with AUCs for 5-year overall survival of 0.78 (0.73-0.83), 0.76 (0.62-0.89), and 0.66 (0.57-0.75) in the training, adult external, and pediatric external cohorts, respectively. Multivariable analysis demonstrated that the metabolic signature had independent prognostic value with hazard ratios of 2.75 (2.06-3.66), 1.89 (1.09-3.29), and 1.96 (1.00-3.84) in the training, adult external, and pediatric external cohorts, respectively. Combining metabolic signatures and classic prognostic factors improved 5-year overall survival prediction compared to the prediction by classic prognostic factors (p < 0.05). GSEA revealed that most pathways were metabolism-related, indicating potential mechanisms. Conclusion: We identified dysregulated metabolic features in AML and constructed a prognostic model to predict the survival of patients with AML.
Collapse
Affiliation(s)
- Yun Wang
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fang Hu
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jin-Yuan Li
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Run-Cong Nie
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Si-Liang Chen
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan-Yu Cai
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ling-Ling Shu
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - De-Jun Deng
- Department of Oncology and Hematology, Shenzhen Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Jing-Bo Xu
- Department of Hematology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yang Liang
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
12
|
Lee D, Ha M, Hong CM, Kim J, Park SM, Park D, Sohn DH, Shin HJ, Yu HS, Kim CD, Kang CD, Han ME, Oh SO, Kim YH. GABRQ expression is a potential prognostic marker for patients with clear cell renal cell carcinoma. Oncol Lett 2019; 18:5731-5738. [PMID: 31788046 PMCID: PMC6865077 DOI: 10.3892/ol.2019.10960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/27/2019] [Indexed: 01/08/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer. Novel biomarkers of ccRCC may provide crucial information on tumor features and prognosis. The present study aimed to determine whether the expression of γ-aminobutyric acid (GABA) A receptor subunit θ (GABRQ) could serve as a novel prognostic marker of ccRCC. GABA is the main inhibitory neurotransmitter in the brain that activates the receptor GABAA, which is comprised of three subunit isoforms: GABRA3, GABRB3 and GABRQ. A recent study reported that GABRQ is involved in the initiation and progression of hepatocellular carcinoma; however, the role of GABRQ in ccRCC remains unknown. In the present study, clinical and transcriptomic data were obtained from cohorts of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). Differential GABRQ expression levels among early (TI and II), late (TIII and IV), nonmetastatic (M0) and metastatic (M1, primary tumor) stages of ccRCC samples were then identified. Furthermore, the use of GABRQ as a prognostic gene was analyzed using Uno's C-index based on the time-dependent area under the curve (AUC), the AUC of the receiver operating characteristic curve at 5 years, the Kaplan-Meier survival curve and multivariate analysis. The survival curve analysis revealed that low GABRQ mRNA expression was significantly associated with a poor prognosis of ccRCC (P<0.001 and P=0.0012 for TCGA and ICGC data, respectively). In addition, analyses of the C-index and AUC values further supported this discriminatory power. Furthermore, the prognostic value of GABRQ mRNA expression was confirmed by multivariate Cox regression analysis. Taken together, these results suggested that GABRQ mRNA expression may be considered as a novel prognostic biomarker of ccRCC.
Collapse
Affiliation(s)
- Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Mihyang Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Chae Mi Hong
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Jayoung Kim
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Su Min Park
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Dongsu Park
- Department of Molecular Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Skeletal Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Ho Jin Shin
- Department of Hematology-Oncology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Hak-Sun Yu
- Department of Parasitology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Chi-Dug Kang
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea.,Department of Biochemistry, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea.,Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do 50612, Republic of Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| |
Collapse
|
13
|
Ha M, Kim DW, Kim J, Hong CM, Park SM, Woo IA, Kim MY, Koo H, Namkoong J, Kim J, Han ME, Song P, Hur J, Kang CD, Kim YH, Lee D, Oh SO. Prognostic role of the beta-2 adrenergic receptor in clear cell renal cell carcinoma. Anim Cells Syst (Seoul) 2019; 23:365-369. [PMID: 31700702 PMCID: PMC6830282 DOI: 10.1080/19768354.2019.1658638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/31/2019] [Accepted: 08/14/2019] [Indexed: 12/18/2022] Open
Abstract
The beta-2 adrenergic receptor (ADRB2) regulates the proliferation, apoptosis, angiogenesis, migration, and metastasis of cancer cells. However, its function in the progression of clear cell renal cell carcinoma (ccRCC) is unknown. Here, we report that ADRB2 can be a novel prognostic factor for patients with ccRCC. The differential expression of ADRB2 in low-stage (stages I and II), high-stage (stages III and IV), low-grade (grades I and II), and high-grade (grades III and IV) ccRCC was identified in cohorts of patients from The Cancer Genome Atlas and the International Cancer Genome Consortium. We evaluated ADRB2 expression as a prognostic factor using the Kaplan-Meier survival curve, multivariate analysis, time-dependent area under the curve (AUC) of Uno's C-index, and AUC of the receiver operating characteristics (ROC) at five years. Kaplan-Meier analysis revealed that reduced ADRB2 expression is associated with poor prognosis in ccRCC patients. Analysis of C-indices and AUC-ROC further confirmed this result. Moreover, multivariate analysis confirmed the prognostic significance of ADRB2 expression. Collectively, these findings suggest that ADRB2 is a potential prognostic factor for ccRCC.
Collapse
Affiliation(s)
- Mihyang Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dong Woo Kim
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jayoung Kim
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Chae Mi Hong
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Su Min Park
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - In Ae Woo
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Min Yong Kim
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hyunjun Koo
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jin Namkoong
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jaehyun Kim
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Parkyong Song
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jin Hur
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Chi-Dug Kang
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Department of Biomedical Informatics, and Biomedical Research Institute, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| |
Collapse
|
14
|
Aqrawi LA, Galtung HK, Guerreiro EM, Øvstebø R, Thiede B, Utheim TP, Chen X, Utheim ØA, Palm Ø, Skarstein K, Jensen JL. Proteomic and histopathological characterisation of sicca subjects and primary Sjögren's syndrome patients reveals promising tear, saliva and extracellular vesicle disease biomarkers. Arthritis Res Ther 2019; 21:181. [PMID: 31366407 PMCID: PMC6670195 DOI: 10.1186/s13075-019-1961-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022] Open
Abstract
Background Mononuclear cell infiltration of exocrine glands, production of Ro/SSA and La/SSB autoantibodies, along with oral and ocular dryness, are characteristic features of primary Sjögren’s syndrome (pSS). Non-SS sicca subjects, an underexplored group in relation to pSS, display similar sicca symptoms, with possible mild signs of inflammation in their salivary glands, yet with no serological detection of autoantibody production. In this study, we investigated inflammatory manifestations in the salivary gland tissue, tear fluid and saliva of non-SS subjects, as compared to pSS patients and healthy individuals. Methods Fifteen non-SS, 10 pSS and 10 healthy subjects were included in the analyses. Histological evaluation of salivary gland biopsies was performed. Liquid chromatography-mass spectrometry (LC-MS) was conducted on tear fluid and stimulated whole saliva, and proteomic biomarker profiles were generated. Extracellular vesicle (EVs) isolation and characterisation from both fluids were also combined with LC-MS. The LC-MS data were analysed for quantitative differences between patient and control groups using Scaffold. Database for Annotation, Visualization and Integrated Discovery (DAVID) and Functional Enrichment Analysis Tool (FunRich) were applied for functional analyses. Results Histopathological evaluation of salivary gland biopsies showed implications of milder inflammation in non-SS subjects through mononuclear cell infiltration, fibrosis and fatty replacement, as compared to pSS patients. Although unaffected in the non-SS group, upregulation of proinflammatory pathways and proteins involved in ubiquitination (LMO7 and HUWE1) and B cell differentiation (TPD52) were detected in tear fluid of pSS patients. Moreover, overexpression of proteins STOM, ANXA4 and ANXA1, regulating cellular innate and adaptive immunological pathways, were further identified in EVs from tear fluid of pSS patients. Finally, whole saliva and EVs isolated from whole saliva of pSS patients expressed proteins vital for innate MHC class I cellular regulation (NGAL) and T cell activation (CD44). Conclusions Non-SS sicca subjects may show implications of mild inflammation in their glandular tissue, while their protein profile was strikingly more similar to healthy controls than to pSS patients. Hence, the tear and salivary biomarkers identified could be implemented as potential non-invasive diagnostic tools that may aid in increasing diagnostic accuracy when evaluating non-SS subjects and pSS patients and monitoring disease progression. Electronic supplementary material The online version of this article (10.1186/s13075-019-1961-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lara A Aqrawi
- Department of Oral Surgery and Oral Medicine, University of Oslo, Oslo, Norway
| | - Hilde Kanli Galtung
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.
| | - Eduarda M Guerreiro
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Reidun Øvstebø
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Tor Paaske Utheim
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.,Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
| | - Xiangjun Chen
- Department of Oral Surgery and Oral Medicine, University of Oslo, Oslo, Norway.,The Norwegian Dry Eye Clinic, Oslo, Norway
| | - Øygunn Aass Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,The Norwegian Dry Eye Clinic, Oslo, Norway
| | - Øyvind Palm
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | - Kathrine Skarstein
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
15
|
Ha M, Jeong H, Roh JS, Lee B, Han ME, Oh SO, Sohn DH, Kim YH. DYSF expression in clear cell renal cell carcinoma: A retrospective study of 2 independent cohorts. Urol Oncol 2019; 37:735-741. [PMID: 31377166 DOI: 10.1016/j.urolonc.2019.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/28/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Renal cell carcinoma (RCC) is the most typical type of kidney cancer in adults. Hypercalcemia is a well known paraneoplastic syndrome associated with RCC and recent studies have reported that hypercalcemia is closely related to the poor prognosis of RCC patients. Clear cell RCC (ccRCC) is the most common and aggressive subtype of RCC. Although the histological classification of RCC is important for determination of appropriate treatment strategies, effective biomarkers for predicting prognosis of ccRCC have not yet been identified. Since calcium levels affect the prognosis of RCC patients, we evaluated whether the calcium-sensing genes on the plasma membrane, including those encoding calcium channels, CaSR, GPRC6a, and DYSF, could be used as biomarkers to predict the prognosis of ccRCC patients. METHODS Information from 537 patients from The Cancer Genome Atlas (TCGA; n = 446) and International Cancer Genome Consortium (ICGC; n = 91) was used in this study. Among these genes, DYSF was the only gene whose expression correlated with overall survival of both TGCA and ICGC patients. RESULTS Although DYSF gene expression was higher in ccRCC tissue than in normal kidney tissue, Kaplan-Meier curves showed that the survival rate of ccRCC patients with high DYSF expression was significantly higher than that of patients with low DYSF expression (TCGA, P < 0.0001; ICGC, P = 0.0011). We also validated the potential of DYSF as a prognostic biomarker for ccRCC by conducting a time-dependent area under the curve (AUC) analysis and 5-years receiver operating characteristic curve analysis. Finally, multivariate regression analysis revealed that the expression of DYSF is independent of other prognostic parameters (TCGA, P = 0.017; ICGC, P = 0.006). CONCLUSIONS These results suggested that DYSF may play a suppressive role in the progression of ccRCC and could act as a promising prognostic biomarker for predicting the survival of ccRCC patients.
Collapse
Affiliation(s)
- Mihyang Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hoim Jeong
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jong Seong Roh
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea.
| | - Yun Hak Kim
- Department of Anatomy and Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
| |
Collapse
|
16
|
Goh TS, Ha M, Lee JS, Jeong DC, Jung ES, Han ME, Kim YH, Oh SO. Prognostic significance of EIF4G1 in patients with pancreatic ductal adenocarcinoma. Onco Targets Ther 2019; 12:2853-2859. [PMID: 31043796 PMCID: PMC6472433 DOI: 10.2147/ott.s202101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Advances in genomics have greatly improved the survival rate in cancer patients. However, due to genetic heterogeneity, pancreatic ductal adenocarcinoma (PDAC) is still difficult to diagnose early, and its survival rate is extremely low. Therefore, we identified biomarkers that predict the prognosis of PDAC patients using independent cohort data. Materials and methods To develop a novel prognostic biomarker, we used the gene expression and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Kaplan-Meier survival curve using median values of genes as cutoff showed that EIF4G1 was the only statistically significant gene in the 3 cohorts. We analyzed the prognostic significance of EIF4G1 using the time-dependent area under the curve (AUC) of Uno's C-index, the AUC value of the receiver operating characteristics (ROC) at 3 years, and multivariate Cox analysis. We also compared EIF4G1 levels between tumors and matched non-tumor tissues. Results EIF4G1 is the only prognostic gene in patients with PDAC, which was selected by Kaplan-Meier survival analysis. The survival curve showed that high expression of EIF4G1 was associated with poor prognosis of PDAC with a good discriminative ability in 3 independent cohorts. The risk stratifying ability of EIF4G1 was demonstrated by analyzing C-indices and AUC values. Multivariate Cox regression confirmed its prognostic significance. EIF4G1 expression was significantly higher in PDAC tissues than in the matched normal tissues. Conclusion EIF4G1 could be used as a novel prognostic marker for PDAC and to determine suitable treatment options.
Collapse
Affiliation(s)
- Tae Sik Goh
- Department of Orthopaedic Surgery and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Mihyang Ha
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea,
| | - Jung Sub Lee
- Department of Orthopaedic Surgery and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Dae Cheon Jeong
- Deloitte Analytics Group, Deloitte Consulting LLC, Seoul, Republic of Korea
| | - Eun Sang Jung
- Department of Bioenvironmental Energy, College of Life & Resources Science, Pusan National University, Miryang, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea,
| | - Yun Hak Kim
- Department of Anatomy and Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Republic of Korea, .,Biomedical Research Institute, Pusan National University, Busan, Republic of Korea,
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Republic of Korea,
| |
Collapse
|
17
|
Jeon YH, Ha M, Kim SW, Kim MJ, Lee CS, Oh CK, Han ME, Oh SO, Kim YH. Evaluation of the prognostic significances of γ-secretase genes in pancreatic cancer. Oncol Lett 2019; 17:4614-4620. [PMID: 30944650 PMCID: PMC6444455 DOI: 10.3892/ol.2019.10113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/19/2019] [Indexed: 12/19/2022] Open
Abstract
With the growing requirement for novel prognostic biomarkers for pancreatic cancer, many studies have focused on clinical and/or genomic variables. Although many studies have been performed, carbohydrate antigen 19-9 is the only biomarker in clinical use. Therefore, the present study examined whether γ-secretase genes, including presenilin (PSEN), nicastrin (NCSTN), presenilin enhancer protein 2 (PSENEN), and anterior pharynx-defective 1 (APH1-), could serve as prognostic factors for pancreatic cancer. The cohorts selected included >100 pancreatic cancer patients. Patient data were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GSE21501). The prognostic roles of the γ-secretase genes were analyzed by several survival analysis methods. Among the γ-secretase genes, the prognosis tended to be worse in the 2 cohorts with increasing expression of PSEN1, APH1A, and PSENEN, while the remaining genes were the opposite in the 2 cohorts. Notably, although the patient characteristics were quite different, APH1A was statistically significantly associated with prognosis in the 2 cohorts. The hazard ratio of APH1A for overall survival was 1.598 (TCGA) and 2.724 (GSE21501). These results contribute to the study of γ-secretase in pancreatic cancer. We believe that γ-secretase, particularly APH1A, will be a new prognostic biomarker for pancreatic cancer.
Collapse
Affiliation(s)
- Yun Ho Jeon
- Department of Pediatrics, Busan St. Mary's Hospital, Busan 48575, Republic of Korea
| | - Mihyang Ha
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sung Won Kim
- Department of Pediatrics, Busan St. Mary's Hospital, Busan 48575, Republic of Korea
| | - Mun Ju Kim
- Department of Pediatrics, Busan St. Mary's Hospital, Busan 48575, Republic of Korea
| | - Chi-Seung Lee
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Chang-Kyu Oh
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea.,Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
18
|
Ha M, Jeong H, Roh JS, Lee B, Lee D, Han ME, Oh SO, Sohn DH, Kim YH. VNN3 is a potential novel biomarker for predicting prognosis in clear cell renal cell carcinoma. Anim Cells Syst (Seoul) 2019; 23:112-117. [PMID: 30949398 PMCID: PMC6440501 DOI: 10.1080/19768354.2019.1583126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/13/2019] [Accepted: 02/10/2019] [Indexed: 02/07/2023] Open
Abstract
Although pathological observations provide approximate prognoses, it is difficult to achieve prognosis in patients with existing prognostic factors. Therefore, it is very important to find appropriate biomarkers to achieve accurate cancer prognosis. Renal cell carcinoma (RCC) has several subtypes, the discrimination of which is crucial for proper treatment. Here, we present a novel biomarker, VNN3, which is used to prognose clear cell renal cell carcinoma (ccRCC), the most common and aggressive subtype of kidney cancer. Patient information analyzed in our study was extracted from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) cohorts. VNN3 expression was considerably higher in stages III and IV than in stages I and II. Moreover, Kaplan–Meier curves associated high VNN3 expression with poor prognoses (TCGA, p < .0001; ICGC, p = .00076), confirming that ccRCC prognosis can be predicted via VNN3 expression patterns. Consistent with all patient results, the prognosis of patients with higher VNN3 expression was worse in both low stage (I and II) and high stage (III and IV) (TCGA, p < 0.0001 in stage I and II; ICGC, p = 0.028 in stage I and II; TCGA, p = 0.005 in stage III and IV). Area under the curve and receiver operating characteristic curves supported our results that highlighted VNN3 expression as a suitable ccRCC biomarker. Multivariate analysis also verified the prognostic performance of VNN3 expression (TCGA, p < .001; ICGC, p = .017). Altogether, we suggest that VNN3 is applicable as a new biomarker to establish prognosis in patients with ccRCC.
Collapse
Affiliation(s)
- Mihyang Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hoim Jeong
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jong Seong Roh
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|