1
|
Chen WR, Liao FY, Lu CY, Weng JR, Feng CH. Ice-bath-effervescence-assisted salting-out extraction combined with microwave-assisted derivatization for the detection of spermidine and spermine in whole blood. J Chromatogr A 2025; 1747:465807. [PMID: 40020286 DOI: 10.1016/j.chroma.2025.465807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Spermidine (Spd) and spermine (Spm) are biomarkers for various cancers. In the present study, we used 4-dimethylamino-1-naphthyl isothiocyanate as a derivatizing reagent to achieve microwave-assisted nucleophilic addition to Spd and Spm. We designed a strategy comprising an ice bath combined with salting out to increase the efficiency of Spd and Spm derivative extractions. Ice-bath-effervescence-assisted salting-out extraction (IEA-SOE) was conducted using a proton donor (citric acid) and an effervescent salt (KHCO3), which simultaneously provided salting-out and effervescence effects. The ice bath stopped the derivatization and improved the extraction efficiency of the IEA-SOE. Moreover, under optimized derivatization and extraction conditions, the total time required was only 10 min. Finally, the extraction layer was collected and analyzed using a narrow-bore liquid chromatograph equipped with a UV detector. The quantitative linear range was 1-200 μM, and the detection limit was 0.1 μM. The intra- and inter-batch relative standard deviations were in the range of 3.96-7.74 %, with relative errors in the range of -10.55-4.70 %. The proposed analytical method was successfully applied to determine Spd and Spm concentrations in whole-blood samples. Our method will serve as a useful tool for the accurate assessment of essential biomarkers in blood and subsequently, disease diagnosis.
Collapse
Affiliation(s)
- Wen-Rong Chen
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Fang-Yi Liao
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chi-Yu Lu
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jing-Ru Weng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Chia-Hsien Feng
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| |
Collapse
|
2
|
Zhang Y, Chen Q, Wang L, Geng H, Zhu Z, Lv C, Zhao Y, Wang X, Sun C, Chen P, Zhang C. Spatially-resolved characterization of the metabolic and N-glycan alterations in colorectal cancer using matrix-assisted laser desorption/ionization mass spectrometry imaging. RSC Adv 2025; 15:1838-1845. [PMID: 39839237 PMCID: PMC11747861 DOI: 10.1039/d4ra08100e] [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: 11/15/2024] [Accepted: 01/10/2025] [Indexed: 01/23/2025] Open
Abstract
Colorectal cancer is the second leading cause of cancer-related deaths worldwide, and its development typically involves complex metabolic reprogramming. By mapping the spatial distributions of metabolites and N-glycans in heterogeneous colorectal cancer tissues, we can elucidate cancer-associated metabolic and N-glycan changes. Herein, we combine mass spectrometry imaging-based metabolomics and N-glycomics to characterize the spatially resolved reprogramming of metabolites and N-glycans in colorectal cancer tissues. The metabolic characteristics of different regions of colorectal cancer were evaluated through the utilization of orthogonal partial least squares discriminant analysis. In combination with metabolic pathway enrichment analysis, significant alterations were identified in the fatty acid metabolism, arginine and proline metabolism of colorectal cancer. Cancer cell regions exhibited a marked upregulation of saturated fatty acids, monounsaturated fatty acids, polyamines, and histidine. Additionally, we discovered that the high-mannose N-glycans were predominantly distributed in tumor tissue regions, whereas complex N-glycans were more commonly found in the normal tissue regions adjacent to the tumor. Such findings provide new insights into the spatial signatures of metabolites and N-glycans in colorectal cancer, thereby offering a crucial basis for the diagnosis of colorectal cancer and potential vulnerabilities that might be targeted for cancer therapy.
Collapse
Affiliation(s)
- Yaqi Zhang
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Qiangjun Chen
- Department of Breast and Thyroid Surgery, Yi Du Central Hospital of Weifang, Shangdong Province No. 5168 Jiangjunshan Road Weifang 262500 China
| | - Lei Wang
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Haoyuan Geng
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Zihan Zhu
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Cancan Lv
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Yisheng Zhao
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Xiao Wang
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Chenglong Sun
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Panpan Chen
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250014 China
| | - Chao Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University Jinan 250012 China
| |
Collapse
|
3
|
Wu H, Fu M, Wu M, Cao Z, Zhang Q, Liu Z. Emerging mechanisms and promising approaches in pancreatic cancer metabolism. Cell Death Dis 2024; 15:553. [PMID: 39090116 PMCID: PMC11294586 DOI: 10.1038/s41419-024-06930-0] [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: 04/18/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Pancreatic cancer is an aggressive cancer with a poor prognosis. Metabolic abnormalities are one of the hallmarks of pancreatic cancer, and pancreatic cancer cells can adapt to biosynthesis, energy intake, and redox needs through metabolic reprogramming to tolerate nutrient deficiency and hypoxic microenvironments. Pancreatic cancer cells can use glucose, amino acids, and lipids as energy to maintain malignant growth. Moreover, they also metabolically interact with cells in the tumour microenvironment to change cell fate, promote tumour progression, and even affect immune responses. Importantly, metabolic changes at the body level deserve more attention. Basic research and clinical trials based on targeted metabolic therapy or in combination with other treatments are in full swing. A more comprehensive and in-depth understanding of the metabolic regulation of pancreatic cancer cells will not only enrich the understanding of the mechanisms of disease progression but also provide inspiration for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hao Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengdi Fu
- Department of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhen Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Qiyao Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
4
|
Xuan M, Gu X, Li J, Huang D, Xue C, He Y. Polyamines: their significance for maintaining health and contributing to diseases. Cell Commun Signal 2023; 21:348. [PMID: 38049863 PMCID: PMC10694995 DOI: 10.1186/s12964-023-01373-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/29/2023] [Indexed: 12/06/2023] Open
Abstract
Polyamines are essential for the growth and proliferation of mammalian cells and are intimately involved in biological mechanisms such as DNA replication, RNA transcription, protein synthesis, and post-translational modification. These mechanisms regulate cellular proliferation, differentiation, programmed cell death, and the formation of tumors. Several studies have confirmed the positive effect of polyamines on the maintenance of health, while others have demonstrated that their activity may promote the occurrence and progression of diseases. This review examines a variety of topics, such as polyamine source and metabolism, including metabolism, transport, and the potential impact of polyamines on health and disease. In addition, a brief summary of the effects of oncogenes and signaling pathways on tumor polyamine metabolism is provided. Video Abstract.
Collapse
Affiliation(s)
- Mengjuan Xuan
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, College of Clinical Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Juan Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chen Xue
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Yuting He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
5
|
Bronson AS, Lilley CM, Zleczewski M, Mirza KM. Polyamine catabolism is concentrated in tumor-associated histiocytes in diffuse large B-cell lymphoma and classic Hodgkin lymphoma. Pathol Res Pract 2023; 248:154627. [PMID: 37343378 DOI: 10.1016/j.prp.2023.154627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023]
Abstract
Polyamines are cationic molecules necessary for cell survival, growth, and replication [1-5]. Polyamines come in a variety of structural forms and are principally regulated by two enzymes, spermine/spermidine acetyltransferase-1 (SAT1) and ornithine decarboxylase-1 (ODC1). SAT1 targets the polyamines spermidine and spermine for degradation via acetylation, while ODC1 is involved in converting the polyamine precursor molecule to more complex polyamines [6-8]. Polyamines and their regulatory enzymes have been implicated in tumor metastasis [9,10] and in crosstalk between oncogenes [11-13] in numerous types of cancer, but their role has never been evaluated in B-cell malignancies. In this study, we examine the expression of SAT1 in diffuse large B-cell lymphoma (DLBCL) and classic Hodgkin lymphoma (HL). We found that SAT1 is expressed in all examined cases of DLBCL (n = 15) and HL (n = 5), though the levels of expression across cases vary. We also note that SAT1 expression appears to be concentrated in tumor-associated histiocytes, rather than tumor cells in both DLBCL and HL. We propose that these findings indicate that the polyamine catabolic enzyme, SAT1, plays an unappreciated role in the pathogenesis of B-cell neoplasms.
Collapse
Affiliation(s)
- Adam S Bronson
- Loyola University Chicago Stritch School of Medicine, 2160 S 1st Ave, Maywood, IL 60153, USA
| | - Cullen M Lilley
- Loyola University Chicago Stritch School of Medicine, 2160 S 1st Ave, Maywood, IL 60153, USA
| | - Mathew Zleczewski
- Loyola University Medical Center Department of Pathology and Laboratory Medicine, 2160 S 1st Ave, Maywood, IL 60153, USA
| | - Kamran M Mirza
- Loyola University Chicago Stritch School of Medicine, 2160 S 1st Ave, Maywood, IL 60153, USA; Loyola University Medical Center Department of Pathology and Laboratory Medicine, 2160 S 1st Ave, Maywood, IL 60153, USA.
| |
Collapse
|
6
|
Perazzoli G, García-Valdeavero OM, Peña M, Prados J, Melguizo C, Jiménez-Luna C. Evaluating Metabolite-Based Biomarkers for Early Diagnosis of Pancreatic Cancer: A Systematic Review. Metabolites 2023; 13:872. [PMID: 37512579 PMCID: PMC10384620 DOI: 10.3390/metabo13070872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, with five-year survival rates around 10%. The only curative option remains complete surgical resection, but due to the delay in diagnosis, less than 20% of patients are eligible for surgery. Therefore, discovering diagnostic biomarkers for early detection is crucial for improving clinical outcomes. Metabolomics has become a powerful technology for biomarker discovery, and several metabolomic-based panels have been proposed for PDAC diagnosis, but these advances have not yet been translated into the clinic. Therefore, this review focused on summarizing metabolites identified for the early diagnosis of PDAC in the last five years. Bibliographic searches were performed in the PubMed, Scopus and WOS databases, using the terms "Biomarkers, Tumor", "Pancreatic Neoplasms", "Early Diagnosis", "Metabolomics" and "Lipidome" (January 2018-March 2023), and resulted in the selection of fourteen original studies that compared PDAC patients with subjects with other pancreatic diseases. These investigations showed amino acid and lipid metabolic pathways as the most commonly altered, reflecting their potential for biomarker research. Furthermore, other relevant metabolites such as glucose and lactate were detected in the pancreas tissue and body fluids from PDAC patients. Our results suggest that the use of metabolomics remains a robust approach to improve the early diagnosis of PDAC. However, these studies showed heterogeneity with respect to the metabolomics techniques used and further studies will be needed to validate the clinical utility of these biomarkers.
Collapse
Affiliation(s)
- Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Olga M García-Valdeavero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Mercedes Peña
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Cristina Jiménez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| |
Collapse
|
7
|
Zimmermann A, Hofer SJ, Madeo F. Molecular targets of spermidine: implications for cancer suppression. Cell Stress 2023; 7:50-58. [PMID: 37431488 PMCID: PMC10320397 DOI: 10.15698/cst2023.07.281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/12/2023] Open
Abstract
Spermidine is a ubiquitous, natural polyamine with geroprotective features. Supplementation of spermidine extends the lifespan of yeast, worms, flies, and mice, and dietary spermidine intake correlates with reduced human mortality. However, the crucial role of polyamines in cell proliferation has also implicated polyamine metabolism in neoplastic diseases, such as cancer. While depleting intracellular polyamine biosynthesis halts tumor growth in mouse models, lifelong external spermidine administration in mice does not increase cancer incidence. In contrast, a series of recent findings points to anti-neoplastic properties of spermidine administration in the context of immunotherapy. Various molecular mechanisms for the anti-aging and anti-cancer properties have been proposed, including the promotion of autophagy, enhanced translational control, and augmented mitochondrial function. For instance, spermidine allosterically activates mitochondrial trifunctional protein (MTP), a bipartite protein complex that mediates three of the four steps of mitochondrial fatty acid (β-oxidation. Through this action, spermidine supplementation is able to restore MTP-mediated mitochondrial respiratory capacity in naïve CD8+ T cells to juvenile levels and thereby improves T cell activation in aged mice. Here, we put this finding into the context of the previously described molecular target space of spermidine.
Collapse
Affiliation(s)
- Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Sebastian J. Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| |
Collapse
|
8
|
Metabolic landscape dysregulation in bronchoalveolar lavage fluid of checkpoint inhibitor pneumonitis. Clin Immunol 2023; 247:109230. [PMID: 36646189 DOI: 10.1016/j.clim.2023.109230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
BACKGROUND Checkpoint inhibitor pneumonitis (CIP) is a potentially fatal adverse event resulting from immunotherapy in patients with malignant tumors. However, the pathogenesis of CIP remains poorly understood. METHODS We collected bronchoalveolar lavage fluid (BALF) from cohorts of patients with CIP, new-onset lung cancer (LC), and idiopathic pulmonary fibrosis (IPF). Non-targeted metabolomics analysis was conducted to analyze metabolic signatures. Flow cytometry was used to evaluate immune cell subsets. RESULTS Lymphocytes were predominant in the BALF of patients with CIP. A total of 903 metabolites were identified, among which lipid compounds were the most abundant. In a comparison between patients with CIP and LC, enrichment analysis of the altered metabolites showed suppressed amino sugar metabolism, and spermidine and spermine biosynthesis in the CIP group. Metabolism of alpha linolenic acid, linoleic acid, and their fatty acid derivatives was enriched in the CIP group relative to the IPF group. The twelve metabolites found to be enriched in the CIP group were positively correlated with the proportion of CD8+ T cells. One cluster of BALF metabolites, 57.14% of which were lipid molecules, was inversely correlated with the proportion of natural killer cells. CONCLUSIONS In this study, the metabolomic landscape of BALF in patients with CIP was determined. We elucidated suppressed tumor metabolic signatures, enhanced pulmonary inflammatory signaling, and the characteristics of responsible immune cells, which helps to understand the pathogenesis of CIP.
Collapse
|
9
|
Islam A, Shaukat Z, Hussain R, Gregory SL. One-Carbon and Polyamine Metabolism as Cancer Therapy Targets. Biomolecules 2022; 12:biom12121902. [PMID: 36551330 PMCID: PMC9775183 DOI: 10.3390/biom12121902] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer metabolic reprogramming is essential for maintaining cancer cell survival and rapid replication. A common target of this metabolic reprogramming is one-carbon metabolism which is notable for its function in DNA synthesis, protein and DNA methylation, and antioxidant production. Polyamines are a key output of one-carbon metabolism with widespread effects on gene expression and signaling. As a result of these functions, one-carbon and polyamine metabolism have recently drawn a lot of interest for their part in cancer malignancy. Therapeutic inhibitors that target one-carbon and polyamine metabolism have thus been trialed as anticancer medications. The significance and future possibilities of one-carbon and polyamine metabolism as a target in cancer therapy are discussed in this review.
Collapse
Affiliation(s)
- Anowarul Islam
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Zeeshan Shaukat
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Rashid Hussain
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Stephen L. Gregory
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Correspondence: ; Tel.: +61-0466987583
| |
Collapse
|
10
|
Dobrovolskaite A, Moots H, Tantak MP, Shah K, Thomas J, Dinara S, Massaro C, Hershberger PM, Maloney PR, Peddibhotla S, Sugarman E, Litherland S, Arnoletti JP, Jha RK, Levens D, Phanstiel O. Discovery of Anthranilic Acid Derivatives as Difluoromethylornithine Adjunct Agents That Inhibit Far Upstream Element Binding Protein 1 (FUBP1) Function. J Med Chem 2022; 65:15391-15415. [PMID: 36382923 PMCID: PMC10512781 DOI: 10.1021/acs.jmedchem.2c01350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Polyamine biosynthesis is regulated by ornithine decarboxylase (ODC), which is transcriptionally activated by c-Myc. A large library was screened to find molecules that potentiate the ODC inhibitor, difluoromethylornithine (DFMO). Anthranilic acid derivatives were identified as DFMO adjunct agents. Further studies identified the far upstream binding protein 1 (FUBP1) as the target of lead compound 9. FUBP1 is a single-stranded DNA/RNA binding protein and a master controller of specific genes including c-Myc and p21. We showed that 9 does not inhibit 3H-spermidine uptake yet works synergistically with DFMO to limit cell growth in the presence of exogenous spermidine. Compound 9 was also shown to inhibit the KH4 FUBP1-FUSE interaction in a gel shift assay, bind to FUBP1 in a ChIP assay, reduce both c-Myc mRNA and protein expression, increase p21 mRNA and protein expression, and deplete intracellular polyamines. This promising hit opens the door to new FUBP1 inhibitors with increased potency.
Collapse
Affiliation(s)
- Aiste Dobrovolskaite
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Holly Moots
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Mukund P Tantak
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Kunal Shah
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Jenna Thomas
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Sharifa Dinara
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Chelsea Massaro
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Paul M Hershberger
- Sanford Burnham Medical Research Institute, 6400 Sanger Road, Orlando, Florida 32827, United States
| | - Patrick R Maloney
- Sanford Burnham Medical Research Institute, 6400 Sanger Road, Orlando, Florida 32827, United States
| | | | - Eliot Sugarman
- Sanford Burnham Medical Research Institute, 6400 Sanger Road, Orlando, Florida 32827, United States
| | - Sally Litherland
- Advent Health Cancer Institute, 2520 North Orange Ave, Suite 104, Orlando, Florida 32804, United States
| | - Juan Pablo Arnoletti
- Advent Health Cancer Institute, 2520 North Orange Ave, Suite 104, Orlando, Florida 32804, United States
| | - Rajiv Kumar Jha
- Laboratory of Pathology, Center for Cancer Research, 10 Center Drive, Building 10, Room 2N106, Bethesda, Maryland 20892-1500, United States
| | - David Levens
- Laboratory of Pathology, Center for Cancer Research, 10 Center Drive, Building 10, Room 2N106, Bethesda, Maryland 20892-1500, United States
| | - Otto Phanstiel
- University of Central Florida, Biomolecular Research Annex, 12722 Research Parkway, Orlando, Florida 32826, United States
| |
Collapse
|
11
|
Teränen V, Nissinen S, Roine A, Antila A, Siiki A, Vaalavuo Y, Kumpulainen P, Oksala N, Laukkarinen J. Bile-volatile organic compounds in the diagnostics of pancreatic cancer and biliary obstruction: A prospective proof-of-concept study. Front Oncol 2022; 12:918539. [PMID: 36479080 PMCID: PMC9720309 DOI: 10.3389/fonc.2022.918539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/31/2022] [Indexed: 08/30/2023] Open
Abstract
OBJECTIVES Detection of volatile organic compounds (VOCs) from bodily fluids with field asymmetric waveform ion mobility spectrometry (FAIMS) and related methods has been studied in various settings. Preliminary results suggest that it is possible to detect prostate, colorectal, ovarian and pancreatic cancer from urine samples. In this study, our primary aim was to differentiate pancreatic cancer from pancreatitis and benign tumours of the pancreas by using bile samples obtained during endoscopic retrograde cholangiopancreatography (ERCP). Secondarily, we aimed to differentiate all pancreatic region malignancies from all other kinds of benign causes of biliary obstruction. METHODS A bile sample was successfully aspirated from 94 patients during ERCP in Tampere University Hospital. Hospital and patient records were prospectively followed up for at least two years after ERCP. Bile samples were analysed using a Lonestar chemical analyser (Owlstone, UK) using an ATLAS sampling system and a split-flow box. Diagnoses and corresponding data from the analyses were matched and divided into two subcategories for comparison. Statistical analysis was performed using linear discriminant analysis, support vector machines, and 5-fold cross-validation. RESULTS Pancreatic cancers (n=8) were differentiated from benign pancreatic lesions (n=9) with a sensitivity of 100%, specificity of 77.8%, and correct rate of 88%. All pancreatic region cancers (n=19) were differentiated from all other kinds of benign causes of biliary obstruction (n=75) with corresponding values of 21.1%, 94.7%, and 80.7%. The sample size was too small to try to differentiate pancreatic cancers from adjacent cancers. CONCLUSION Analysing bile VOCs using FAIMS shows promising capability in detecting pancreatic cancer and other cancers in the pancreatic area.
Collapse
Affiliation(s)
- Ville Teränen
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Samuli Nissinen
- Department of Internal Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Antti Roine
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anne Antila
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Antti Siiki
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Yrjö Vaalavuo
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Pekka Kumpulainen
- Department of Internal Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Niku Oksala
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Johanna Laukkarinen
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
12
|
Guo Z, Zhang X, Lin C, Huang Y, Zhong Y, Guo H, Zheng Z, Weng S. METTL3-IGF2BP3-axis mediates the proliferation and migration of pancreatic cancer by regulating spermine synthase m6A modification. Front Oncol 2022; 12:962204. [PMID: 36276112 PMCID: PMC9582246 DOI: 10.3389/fonc.2022.962204] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Spermine synthase (SMS) is an enzyme participating in polyamine synthesis; however, its function and role in pancreatic cancer remains elusive. Here we report that SMS is upregulated in pancreatic cancer and predicts a worse overall survival and significantly promotes the proliferation and migration of pancreatic cancer cells. Excessive SMS reduces the accumulation of spermidine by converting spermidine into spermine, which activates the phosphorylation of serine/threonine kinase (AKT) and epithelial-mesenchymal transition (EMT) signaling pathway, thereby inhibiting pancreatic cancer cell proliferation and invasion. Moreover, SMS was identified as the direct target of both methyltransferase like 3 (METTL3) and insulin like growth factor 2 mRNA binding protein 3 (IGF2BP3), which directly bind to the m6A modification sites of SMS and inhibit mRNA degradation. Knockdown of METTL3 or IGF2BP3 significantly reduced the SMS protein expression and inhibited the migration of pancreatic cancer. We propose a novel regulatory mechanism in which the METTL3-IGF2BP3 axis mediates the mRNA degradation of SMS in an m6A-dependent manner to regulate spermine/spermidine conversion, which regulates AKT phosphorylation and EMT activation, thereby inducing tumor progression and migration in pancreatic cancer.
Collapse
Affiliation(s)
- Zhenyun Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiang Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chengjie Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yue Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yun Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hailing Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhou Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shangeng Weng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
- *Correspondence: Shangeng Weng,
| |
Collapse
|
13
|
ATP13A3 facilitates polyamine transport in human pancreatic cancer cells. Sci Rep 2022; 12:4045. [PMID: 35260637 PMCID: PMC8904813 DOI: 10.1038/s41598-022-07712-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/18/2022] [Indexed: 01/01/2023] Open
Abstract
The purpose of this study is to provide an increased understanding of the molecular mechanisms responsible for mammalian polyamine transport, a process that has been a long-standing 'black box' for the polyamine field. Here, we describe how ATP13A3, a P-type ATPase, functions as a polyamine transporter in response to different polyamine stimuli and polyamine-targeted therapies in highly proliferating pancreatic cancer cells. We assessed the expression, cellular localization and the response of the human ATP13A3 protein to polyamine treatments in different pancreatic cancer cell lines using Western blot and immunofluorescence microscopy. Using CRISPR mutagenesis and radiolabeled polyamine uptake assays, we investigated the role of ATP13A3 protein in polyamine transport. Highly metastatic cancer cells with high polyamine import express higher levels of the full-length ATP13A3 compared to cells with slow proliferation and low import activity. Highlighting its role in polyamine trafficking, the localization of ATP13A3 is altered in the presence of polyamine stimuli and polyamine-targeted therapies in these cells. Using CRISPR mutagenesis, we demonstrate that the first membrane-associated domain of this protein is critical and indispensable for its function as a spermidine and spermine transporter in cells. Further analysis of existing databases revealed that pancreatic cancer patients with high expression of ATP13A3 have decreased overall survival consistent with the role of intracellular polyamines in supporting tumor growth. Our studies shed light on the mysterious polyamine transport process in human cells and clearly establishes ATP13A3 as an intrinsic component of the spermidine and spermine transport system in humans.
Collapse
|
14
|
Potential Metabolite Markers for Pancreatic Cancer Identified by Metabolomic Analysis of Induced Cancer-Associated Fibroblasts. Cancers (Basel) 2022; 14:cancers14061375. [PMID: 35326527 PMCID: PMC8945883 DOI: 10.3390/cancers14061375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Fibroblasts in normal tissues conduct energy metabolism via oxidative phosphorylation (OXPHOS). However, cancer-associated fibroblasts (CAFs) produce energy (i.e., ATP) via glycolysis. Nonetheless, whether intracellular metabolism transitions from OXPHOS to glycolysis when normal tissue fibroblasts differentiate into CAFs remains to be determined. Here, we established an experimental system and induced the in vitro differentiation of mesenchymal stem cells to CAFs and performed detailed metabolomic and RNA sequencing analyses. We found that the intracellular metabolic pathway was reprogrammed to the glycolytic pathway when mesenchymal stem cells were co-cultured with pancreatic cancer cells. Furthermore, we identified CAF-specific metabolites that were expressed post reprogramming. These metabolites have also been observed in pancreatic cancer mouse models, suggesting their potential as cancer biomarkers. Abstract Cancer-associated fibroblasts (CAFs) in the tumor microenvironment perform glycolysis to produce energy, i.e., ATP. Since the origin of CAFs is unidentified, it is not determined whether the intracellular metabolism transitions from oxidative phosphorylation (OXPHOS) to glycolysis when normal tissue fibroblasts differentiate into CAFs. In this study, we established an experimental system and induced the in vitro differentiation of mesenchymal stem cells (MSCs) to CAFs. Additionally, we performed metabolomic and RNA-sequencing analyses before and after differentiation to investigate changes in the intracellular metabolism. Consequently, we discovered that OXPHOS, which was the primary intracellular metabolism in MSCs, was reprogrammed to glycolysis. Furthermore, we analyzed the metabolites in pancreatic tumor tissues in a mice model. The metabolites extracted as candidates in the in vitro experiments were also detected in the in vivo experiments. Thus, we conclude that normal tissue fibroblasts that differentiate into CAFs undergo a metabolic reprogramming from OXPHOS to glycolysis. Moreover, we identified the CAF-specific metabolites expressed during metabolic reprogramming as potential future biomarkers for pancreatic cancer.
Collapse
|
15
|
Nissinen SI, Venäläinen M, Kumpulainen P, Roine A, Häkkinen MR, Vepsäläinen J, Oksala N, Rantanen T. Discrimination between Pancreatic Cancer, Pancreatitis and Healthy Controls Using Urinary Polyamine Panel. Cancer Control 2022; 28:10732748211039762. [PMID: 35135363 PMCID: PMC8832577 DOI: 10.1177/10732748211039762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKROUND Polyamines play an important role in cellular proliferation, and the change in polyamine metabolism is reported in various cancers. We searched for urinary polyamine signature for distinguishing between pancreatic cancer, premalignant lesions of the pancreas (PLP), acute and chronic pancreatitis, and controls. METHODS Patients and controls were prospectively recruited in three Finnish hospitals between October 2013 and June 2016. The patients provided a urine sample at the time of the diagnosis. The panel of 14 polyamines was obtained in a single run with mass spectrometry. The polyamine concentrations were analysed with quadratic discriminant analysis and cross-validated with leave-one-out cross-validation. RESULTS Sixty-eight patients with pancreatic cancer, 36 with acute pancreatitis, 18 with chronic pancreatitis and 7 with PLP were recruited, as were 53 controls. The combination of 4 polyamines - acetylputrescine, diacetylspermidine, N8-acetylspermidine and diacetylputrescine - distinguished pancreatic cancer and PLP from controls (sensitivity = 94%, specificity = 68% and AUC = 0.88). The combination of diacetylspermidine, N8-acetylspermidine and diacetylspermine distinguished acute pancreatitis from controls (sensitivity = 94%, specificity = 92%, AUC = 0.98). The combination of acetylputrescine, diacetylspermidine and diacetylputrescine distinguished chronic pancreatitis from controls (sensitivity = 98%, specificity = 71%, AUC = 0.93). CONCLUSIONS Optimally selected urinary polyamine panels discriminate between pancreatic cancer and controls, as well as between acute and chronic pancreatitis and controls.
Collapse
Affiliation(s)
- Samuli I Nissinen
- Department of Internal Medicine, School of Medicine, 205537University of Eastern Finland, Kuopio, Finland.,Department of Internal Medicine, 3701Kanta-Häme Central Hospital, Hämeenlinna, Finland
| | - Markus Venäläinen
- Department of Internal Medicine, School of Medicine, 205537University of Eastern Finland, Kuopio, Finland
| | | | - Antti Roine
- Faculty of Medicine and Health Technology, 7840Tampere University, Tampere, Finland
| | - Merja R Häkkinen
- School of Pharmacy, Biocenter Kuopio, 205537University of Eastern Finland, Kuopio, Finland
| | - Jouko Vepsäläinen
- School of Pharmacy, Biocenter Kuopio, 205537University of Eastern Finland, Kuopio, Finland
| | - Niku Oksala
- Faculty of Medicine and Health Technology, 7840Tampere University, Tampere, Finland.,Centre for Vascular Surgery and Interventional Radiology, 60670Tampere University Hospital, Tampere, Finland
| | - Tuomo Rantanen
- Department of Surgery, School of Medicine, 205537University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
16
|
Nakkina SP, Gitto SB, Pandey V, Parikh JG, Geerts D, Maurer HC, Olive KP, Phanstiel O, Altomare DA. Differential Expression of Polyamine Pathways in Human Pancreatic Tumor Progression and Effects of Polyamine Blockade on Tumor Microenvironment. Cancers (Basel) 2021; 13:6391. [PMID: 34945011 PMCID: PMC8699198 DOI: 10.3390/cancers13246391] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death. Existing therapies only moderately improve pancreatic ductal adenocarcinoma (PDAC) patient prognosis. The present study investigates the importance of the polyamine metabolism in the pancreatic tumor microenvironment. Relative mRNA expression analysis identified differential expression of polyamine biosynthesis, homeostasis, and transport mediators in both pancreatic epithelial and stromal cells from low-grade pancreatic intraepithelial neoplasia (PanIN-1) or primary PDAC patient samples. We found dysregulated mRNA levels that encode for proteins associated with the polyamine pathway of PDAC tumors compared to early lesions. Next, bioinformatic databases were used to assess expression of select genes involved in polyamine metabolism and their impact on patient survival. Higher expression of pro-polyamine genes was associated with poor patient prognosis, supporting the use of a polyamine blockade therapy (PBT) strategy for inhibiting pancreatic tumor progression. Moreover, PBT treatment of syngeneic mice injected intra-pancreatic with PAN 02 tumor cells resulted in increased survival and decreased tumor weights of PDAC-bearing mice. Histological assessment of PBT-treated tumors revealed macrophage presence and significantly increased expression of CD86, a T cell co-stimulatory marker. Collectively, therapies which target polyamine metabolism can be used to disrupt tumor progression, modulate tumor microenvironment, and extend overall survival.
Collapse
Affiliation(s)
- Sai Preethi Nakkina
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| | - Sarah B. Gitto
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.G.); (V.P.)
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Veethika Pandey
- Ovarian Cancer Research Center, Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.B.G.); (V.P.)
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jignesh G. Parikh
- Department of Pathology, Orlando VA Medical Center, 13800 Veterans Way, Orlando, FL 32827, USA;
| | - Dirk Geerts
- Department of Medical Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Hans Carlo Maurer
- Internal Medicine II, School of Medicine, Technische Universität München, 81675 Munich, Germany;
| | - Kenneth P. Olive
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA;
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Otto Phanstiel
- Department of Medical Education, College of Medicine, University of Central Florida, Orlando, FL 32826, USA
| | - Deborah A. Altomare
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
17
|
Wodtke R, Pietzsch J, Löser R. Solid-Phase Synthesis of Selectively Mono-Fluorobenz(o)ylated Polyamines as a Basis for the Development of 18F-Labeled Radiotracers. Molecules 2021; 26:molecules26227012. [PMID: 34834103 PMCID: PMC8625420 DOI: 10.3390/molecules26227012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Polyamines are highly attractive vectors for tumor targeting, particularly with regards to the development of radiolabeled probes for imaging by positron emission (PET) and single-photon emission computed tomography (SPECT). However, the synthesis of selectively functionalized derivatives remains challenging due to the presence of multiple amino groups of similar reactivity. In this work, we established a synthetic methodology for the selective mono-fluorobenz(o)ylation of various biogenic diamines and polyamines as lead compounds for the perspective development of substrate-based radiotracers for targeting polyamine-specific membrane transporters and enzymes such as transglutaminases. For this purpose, the polyamine scaffold was constructed by solid-phase synthesis of the corresponding oxopolyamines and subsequent reduction with BH3/THF. Primary and secondary amino groups were selectively protected using Dde and Boc as protecting groups, respectively, in orientation to previously reported procedures, which enabled the selective introduction of the reporter groups. For example, N1-FBz-spermidine, N4-FBz-spermidine, N8-FBz-spermidine, and N1-FBz-spermine and N4-FBz-spermine (FBz = 4-fluorobenzoyl) were obtained in good yields by this approach. The advantages and disadvantages of this synthetic approach are discussed in detail and its suitability for radiolabeling was demonstrated for the solid-phase synthesis of N1-[18F]FBz-cadaverine.
Collapse
Affiliation(s)
- Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany;
- Correspondence: (R.W.); (R.L.); Tel.: +49-351-260-3923 (R.W.); +49-351-260-3658 (R.L.)
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany;
- Faculty of Chemistry and Food Chemistry, School of Science, Technische University Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Reik Löser
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany;
- Faculty of Chemistry and Food Chemistry, School of Science, Technische University Dresden, Mommsenstraße 4, 01069 Dresden, Germany
- Correspondence: (R.W.); (R.L.); Tel.: +49-351-260-3923 (R.W.); +49-351-260-3658 (R.L.)
| |
Collapse
|
18
|
Novita Sari I, Setiawan T, Seock Kim K, Toni Wijaya Y, Won Cho K, Young Kwon H. Metabolism and function of polyamines in cancer progression. Cancer Lett 2021; 519:91-104. [PMID: 34186159 DOI: 10.1016/j.canlet.2021.06.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/11/2021] [Accepted: 06/22/2021] [Indexed: 01/18/2023]
Abstract
Polyamines are essential for the proliferation, differentiation, and development of eukaryotes. They include spermine, spermidine, and the diamine precursor putrescine, and are low-molecular-weight, organic polycations with more than two amino groups. Their intracellular concentrations are strictly maintained within a specific physiological range through several regulatory mechanisms in normal cells. In contrast, polyamine metabolism is dysregulated in many neoplastic states, including cancer. In various types of cancer, polyamine levels are elevated, and crosstalk occurs between polyamine metabolism and oncogenic pathways, such as mTOR and RAS pathways. Thus, polyamines might have potential as therapeutic targets in the prevention and treatment of cancer. The molecular mechanisms linking polyamine metabolism to carcinogenesis must be unraveled to develop novel inhibitors of polyamine metabolism. This overview describes the nature of polyamines, their association with carcinogenesis, the development of polyamine inhibitors and their potential, and the findings of clinical trials.
Collapse
Affiliation(s)
- Ita Novita Sari
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Tania Setiawan
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Kwang Seock Kim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Yoseph Toni Wijaya
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea; Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea.
| | - Hyog Young Kwon
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea; Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea.
| |
Collapse
|
19
|
A naphthalimide-polyamine conjugate preferentially accumulates in hepatic carcinoma metastases as a lysosome-targeted antimetastatic agent. Eur J Med Chem 2021; 221:113469. [PMID: 33965862 DOI: 10.1016/j.ejmech.2021.113469] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/31/2022]
Abstract
Disseminated tumors lead to approximately 90% of cancer-associated deaths especially for hepatocellular carcinoma (HCC), indicating the imperative need of antimetastatic drugs and the ineffectiveness of current therapies. Recently polyamine derivatives have been identified as a promising prospect in dealing with metastatic tumors. Herein, a novel class of naphthalimide-polyamine conjugates 8a-8d, 13a-13c, 17 and 21 were synthesized and the mechanism was further determined. The polyamine conjugate 13b displayed remarkably elevated anti-tumor and anti-metastatic effects (76.01% and 75.02%) than the positive control amonafide (46.91% and 55.77%) at 5 mg/kg in vivo. The underlying molecular mechanism indicated that in addition to induce DNA damage by up-regulating p53 and γH2AX, 13b also targeted lysosome to modulate polyamine metabolism and function in a totally different way from that of amonafide. Furthermore, the HMGB1/p62/LC3II/LC3I and p53/SSAT/β-catenin pathways were mainly involved in the inhibition of 13b-induced HCC metastasis by targeting polyamine transporters (PTs) overexpressed in HCC. At last, 13b down-regulated the concentrations of Put, Spd and Spm by modulating polyamine metabolism key enzymes SSAT and PAO, which favored the suppression of fast growing tumor cells. Taken together, our study implies a promising strategy for naphthalimide conjugates to treat terminal cancer of HCC by targeting autophagy and tumor microenvironment with reduced toxicities and notable activities.
Collapse
|
20
|
Xu J, Zhu S, Xu L, Liu X, Ding W, Wang Q, Chen Y, Deng H. CA9 Silencing Promotes Mitochondrial Biogenesis, Increases Putrescine Toxicity and Decreases Cell Motility to Suppress ccRCC Progression. Int J Mol Sci 2020; 21:E5939. [PMID: 32824856 PMCID: PMC7460829 DOI: 10.3390/ijms21165939] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 02/04/2023] Open
Abstract
Carbonic anhydrase IX (CA9), a pH-regulating transmembrane protein, is highly expressed in solid tumors, and particularly in clear cell renal cell carcinoma (ccRCC). The catalytic mechanisms of CA9 are well defined, but its roles in mediating cell migration/invasion and survival in ccRCC remain to be determined. Here, we confirmed that the mRNA expression of CA9 in ccRCC was significantly higher than that in para-carcinoma tissues from analysis of the datasets in The Cancer Genome Atlas. CA9 knockdown upregulated oxidative phosphorylation-associated proteins and increased mitochondrial biogenesis, resulting in the reversal of the Warburg phenotype and the inhibition of cell growth. Our study revealed that CA9 knockdown upregulated mitochondrial arginase 2 (ARG2), leading to the accumulation of putrescine, which suppressed ccRCC proliferation. Surfaceomics analysis revealed that CA9 knockdown downregulated proteins associated with extracellular matrix (ECM)-receptor interaction and cell adhesion, resulting in decreased cell migration. CA9 silencing also downregulated amino acid transporters, leading to reduced cellular amino acids. Collectively, our data show that CA9 knockdown suppresses proliferation via metabolic reprogramming and reduced cell migration, reaffirming that CA9 is a potential therapeutic target for ccRCC treatment.
Collapse
Affiliation(s)
- Jiatong Xu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Songbiao Zhu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Lina Xu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Xiaohui Liu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Wenxi Ding
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Qingtao Wang
- Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100043, China;
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (J.X.); (S.Z.); (L.X.); (X.L.); (W.D.)
| |
Collapse
|
21
|
Ma J, Li Y, Li L, Yue K, Liu H, Wang J, Xi Z, Shi M, Zhao S, Ma Q, Liu S, Guo S, Liu J, Hou L, Wang C, Wang PG, Tian Z, Xie S. A Polyamine-Based Dinitro-Naphthalimide Conjugate as Substrates for Polyamine Transporters Preferentially Accumulates in Cancer Cells and Minimizes Side Effects in vitro and in vivo. Front Chem 2020; 8:166. [PMID: 32328475 PMCID: PMC7160362 DOI: 10.3389/fchem.2020.00166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/25/2020] [Indexed: 01/10/2023] Open
Abstract
Naphthalimides, such as amonafide and mitonafide in clinical trials, have been developed as antitumor agents for orthotopic tumor. However, the serious side effects in cancer patients limit their applications. Herein, a new class of polyamine-based naphthalimide conjugates 5a-5c, 7a-7b, and 11a-11b with and without the alkylation of the distant nitrogen in the polyamine chain were synthesized and the mechanism was determined. Compared with amonafide, dinitro-naphthalimide conjugate 5c with a 4,3-cyclopropyl motif preferentially accumulates in cancer cells and minimizes side effects in vitro and in vivo. More importantly, 5c at the dosage of as low as 3 mg/kg (57.97%) displays better antitumor effects than the positive control amonafide (53.27%) at 5 mg/kg in vivo. And a remarkably elevated antitumor activity and a reduced toxicity are also observed for 5c at 5 mg/kg (65.90%). The upregulated p53 and the apoptotic cells (73.50%) indicate that the mechanism of 5c to induce apoptosis may result from its enhanced DNA damage. Further investigation indicates that in addition to target DNA, 5c can modulate the polyamine homeostasis by upregulating polyamine oxidase (PAO) in a different way from that of amonafide. And also by targeting PTs overexpressed in most of cancer cells, 5c downregulates the contents of Put, Spd, and Spm, which are in favor of suppressing fast-growing tumor cells. Our study implies a promising strategy for naphthalimide conjugates to treat hepatic carcinoma with notable activities and reduced toxicities at a low dosage.
Collapse
Affiliation(s)
- Jing Ma
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Yingguang Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Linrong Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Kexin Yue
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Hanfang Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Jiajia Wang
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medicine Science, Henan University, Kaifeng, China
| | - Zhuoqing Xi
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China.,Henan University of Science and Technology Second Affiliated Hospital, Luoyang, China
| | - Man Shi
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Sihan Zhao
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Qi Ma
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Sitong Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Shudi Guo
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Jianing Liu
- School of Medicine, Henan University Minsheng College, Kaifeng, China
| | - Lili Hou
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Chaojie Wang
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| | - Peng George Wang
- Southern University of Science and Technology, School of Medicine, Shenzhen, China
| | - Zhiyong Tian
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| | - Songqiang Xie
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, Kaifeng, China
| |
Collapse
|
22
|
Venkat S, Tisdale AA, Schwarz JR, Alahmari AA, Maurer HC, Olive KP, Eng KH, Feigin ME. Alternative polyadenylation drives oncogenic gene expression in pancreatic ductal adenocarcinoma. Genome Res 2020; 30:347-360. [PMID: 32029502 PMCID: PMC7111527 DOI: 10.1101/gr.257550.119] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/04/2020] [Indexed: 01/08/2023]
Abstract
Alternative polyadenylation (APA) is a gene regulatory process that dictates mRNA 3'-UTR length, resulting in changes in mRNA stability and localization. APA is frequently disrupted in cancer and promotes tumorigenesis through altered expression of oncogenes and tumor suppressors. Pan-cancer analyses have revealed common APA events across the tumor landscape; however, little is known about tumor type-specific alterations that may uncover novel events and vulnerabilities. Here, we integrate RNA-sequencing data from the Genotype-Tissue Expression (GTEx) project and The Cancer Genome Atlas (TCGA) to comprehensively analyze APA events in 148 pancreatic ductal adenocarcinomas (PDACs). We report widespread, recurrent, and functionally relevant 3'-UTR alterations associated with gene expression changes of known and newly identified PDAC growth-promoting genes and experimentally validate the effects of these APA events on protein expression. We find enrichment for APA events in genes associated with known PDAC pathways, loss of tumor-suppressive miRNA binding sites, and increased heterogeneity in 3'-UTR forms of metabolic genes. Survival analyses reveal a subset of 3'-UTR alterations that independently characterize a poor prognostic cohort among PDAC patients. Finally, we identify and validate the casein kinase CSNK1A1 (also known as CK1alpha or CK1a) as an APA-regulated therapeutic target in PDAC. Knockdown or pharmacological inhibition of CSNK1A1 attenuates PDAC cell proliferation and clonogenic growth. Our single-cancer analysis reveals APA as an underappreciated driver of protumorigenic gene expression in PDAC via the loss of miRNA regulation.
Collapse
Affiliation(s)
- Swati Venkat
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
| | - Arwen A Tisdale
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
| | - Johann R Schwarz
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
| | - Abdulrahman A Alahmari
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
| | - H Carlo Maurer
- Klinikum rechts der Isar, II. Medizinische Klinik, Technische Universität München, 81675 Munich, Germany
| | - Kenneth P Olive
- Herbert Irving Comprehensive Cancer Center, Department of Medicine, Division of Digestive and Liver Diseases, Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York 10032, USA
| | - Kevin H Eng
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
| | - Michael E Feigin
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, USA
| |
Collapse
|
23
|
Liu H, Ma J, Li Y, Yue K, Li L, Xi Z, Zhang X, Liu J, Feng K, Ma Q, Liu S, Guo S, Wang PG, Wang C, Xie S. Polyamine-Based Pt(IV) Prodrugs as Substrates for Polyamine Transporters Preferentially Accumulate in Cancer Metastases as DNA and Polyamine Metabolism Dual-Targeted Antimetastatic Agents. J Med Chem 2019; 62:11324-11334. [PMID: 31765154 DOI: 10.1021/acs.jmedchem.9b01641] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diverse platinum drug candidates have been designed to improve inhibitory potency and overcome resistance for orthotopic tumors. However, the antimetastatic properties have rarely been reported. We herein report that homospermidineplatin (4a), a polyamine-Pt(IV) prodrug, can potently inhibit tumor growth in situ and reverse cisplatin resistance as expected, and more importantly, 4a displays remarkably elevated antimetastatic activity in vivo (65.7%), compared to those of cisplatin (27.0%) and oxaliplatin (19.6%). The underlying molecular mechanism indicates that in addition to targeting nuclear DNA, 4a can modulate polyamine metabolism and function in a manner different from that of cisplatin. By upregulating SSAT and PAO, 4a downregulates the concentrations of Put, Spd, and Spm, which favors the suppression of fast-growing tumor cells. Moreover, the p53/SSAT/β-catenin and PAO/ROS/GSH/GSH-Px pathways are involved in the inhibition of 4a-induced tumor metastasis. Our study implies a promising strategy for the design of platinum drugs for the treatment of terminal cancer.
Collapse
Affiliation(s)
- Hanfang Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Jing Ma
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Yingguang Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Kexin Yue
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Linrong Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Zhuoqing Xi
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
- Henan University of Science and Technology Second Affiliated Hospital , Luoyang 471000 , China
| | - Xiao Zhang
- The Key Laboratory of Natural Medicine and Immuno-Engineering , Henan University , Kaifeng 475004 , China
| | - Jianing Liu
- School of Medicine , Henan University Minsheng College , Kaifeng 475004 , China
| | - Kai Feng
- School of Medicine , Henan University Minsheng College , Kaifeng 475004 , China
| | - Qi Ma
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Sitong Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Shudi Guo
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Peng George Wang
- The State Key Laboratory of Microbial Technology and National Glycoengineering Research Center , Shandong University , Qingdao 266237 , China
| | - Chaojie Wang
- The Key Laboratory of Natural Medicine and Immuno-Engineering , Henan University , Kaifeng 475004 , China
| | - Songqiang Xie
- School of Pharmacy, Institute of Chemical Biology , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| |
Collapse
|
24
|
Li D, Qian X, Xu P, Wang X, Li Z, Qian J, Yao J. Identification of lncRNAs and Their Functional Network Associated with Chemoresistance in SW1990/GZ Pancreatic Cancer Cells by RNA Sequencing. DNA Cell Biol 2018; 37:839-849. [PMID: 30113217 DOI: 10.1089/dna.2018.4312] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Duguang Li
- The Second Clinical College of Dalian Medical University, Dalian, China
| | - Xiaowei Qian
- Yangzhou University Medical College, Yangzhou, China
| | - Peng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Xiaodong Wang
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Zhennan Li
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Jianjun Qian
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| | - Jie Yao
- Department of Hepatobiliary and Pancreatic Surgery, Northern Jiangsu People's Hospital, Clinic Medical College of Yangzhou University, Yangzhou, China
| |
Collapse
|
25
|
Alpha-Difluoromethylornithine, an Irreversible Inhibitor of Polyamine Biosynthesis, as a Therapeutic Strategy against Hyperproliferative and Infectious Diseases. Med Sci (Basel) 2018; 6:medsci6010012. [PMID: 29419804 PMCID: PMC5872169 DOI: 10.3390/medsci6010012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
The fluorinated ornithine analog α-difluoromethylornithine (DFMO, eflornithine, ornidyl) is an irreversible suicide inhibitor of ornithine decarboxylase (ODC), the first and rate-limiting enzyme of polyamine biosynthesis. The ubiquitous and essential polyamines have many functions, but are primarily important for rapidly proliferating cells. Thus, ODC is potentially a drug target for any disease state where rapid growth is a key process leading to pathology. The compound was originally discovered as an anticancer drug, but its effectiveness was disappointing. However, DFMO was successfully developed to treat African sleeping sickness and is currently one of few clinically used drugs to combat this neglected tropical disease. The other Food and Drug Administration (FDA) approved application for DFMO is as an active ingredient in the hair removal cream Vaniqa. In recent years, renewed interest in DFMO for hyperproliferative diseases has led to increased research and promising preclinical and clinical trials. This review explores the use of DFMO for the treatment of African sleeping sickness and hirsutism, as well as its potential as a chemopreventive and chemotherapeutic agent against colorectal cancer and neuroblastoma.
Collapse
|
26
|
Elevated Polyamines in Saliva of Pancreatic Cancer. Cancers (Basel) 2018; 10:cancers10020043. [PMID: 29401744 PMCID: PMC5836075 DOI: 10.3390/cancers10020043] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 12/13/2022] Open
Abstract
Detection of pancreatic cancer (PC) at a resectable stage is still difficult because of the lack of accurate detection tests. The development of accurate biomarkers in low or non-invasive biofluids is essential to enable frequent tests, which would help increase the opportunity of PC detection in early stages. Polyamines have been reported as possible biomarkers in urine and saliva samples in various cancers. Here, we analyzed salivary metabolites, including polyamines, using capillary electrophoresis-mass spectrometry. Salivary samples were collected from patients with PC (n = 39), those with chronic pancreatitis (CP, n = 14), and controls (C, n = 26). Polyamines, such as spermine, N₁-acetylspermidine, and N₁-acetylspermine, showed a significant difference between patients with PC and those with C, and the combination of four metabolites including N₁-acetylspermidine showed high accuracy in discriminating PC from the other two groups. These data show the potential of saliva as a source for tests screening for PC.
Collapse
|