1
|
Hieromnimon M, Regan DP, Lokken RP, Schook LB, Gaba RC, Schachtschneider KM. Single and multi-omic characterization of a porcine model of ethanol-induced hepatic fibrosis. Epigenetics 2025; 20:2471127. [PMID: 40040391 PMCID: PMC11901410 DOI: 10.1080/15592294.2025.2471127] [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: 07/18/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
Cirrhosis is a form of end-stage liver disease characterized by extensive hepatic fibrosis and loss of liver parenchyma. It is most commonly the result of long-term alcohol abuse in the United States. Large animal models of cirrhosis, as well as of one of its common long-term sequelae, HCC, are needed to study novel and emerging therapeutic interventions. In the present study, liver fibrosis was induced in the Oncopig cancer model, a large animal HCC model, via intrahepatic, intra-arterial ethanol infusion. Liver sections from five fibrosis induced and five age-matched controls were harvested for RNA-seq (mRNA and lncRNA), small RNA-seq (miRNA), and reduced representation bisulfite sequencing (RRBS; DNA methylation). Single- and multi-omic analysis was performed to investigate the transcriptomic and epigenomic mechanisms associated with fibrosis deposition in this model. A total of 3,439 genes, 70 miRNAs, 452 lncRNAs, and 7,715 methylation regions were found to be differentially regulated through individual single-omic analysis. Pathway analysis indicated differentially expressed genes were associated with collagen synthesis and turnover, hepatic metabolic functions such as ethanol and lipid metabolism, and proliferative and anti-proliferative pathways including PI3K and BAX/BCL signaling pathways. Multi-omic latent variable analysis demonstrated significant concordance with the single-omic analysis. lncRNA's associated with UHRF1BP1L and S1PR1 genes were found to reliably discriminate the two arms of the study. These genes were previously implicated in human cancer development and vasculogenesis, respectively. These findings support the validity and translatability of this model as a useful preclinical tool in the study of alcoholic liver disease and its treatment.
Collapse
Affiliation(s)
- Mark Hieromnimon
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel P. Regan
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, USA
| | - R. Peter Lokken
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Sus Clinicals Inc, Chicago, IL, USA
| | - Ron C. Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M. Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Sus Clinicals Inc, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Jiang C, Zeng X, Wang J, Wu X, Song L, Yang L, Li Z, Xie N, Yuan X, Wei Z, Guan Y. Andrographolide sulfonate alleviates rheumatoid arthritis by inhibiting glycolysis-mediated activation of PI3K/AKT to restrain Th17 cell differentiation. Chin J Nat Med 2025; 23:480-491. [PMID: 40274350 DOI: 10.1016/s1875-5364(25)60855-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 04/26/2025]
Abstract
Andrographolide sulfonate (AS) is a sulfonated derivative of andrographolide extracted from Andrographis paniculata (Burm.f.) Nees, and has been approved for several decades in China. The present study aimed to investigate the novel therapeutic application and possible mechanisms of AS in the treatment of rheumatoid arthritis. Results indicated that administration of AS by injection or gavage significantly reduced the paw swelling, improved body weights, and attenuated pathological changes in joints of rats with adjuvant-induced arthritis. Additionally, the levels of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and IL-1β in the serum and ankle joints were reduced. Bioinformatics analysis, along with the spleen index and measurements of IL-17 and IL-10 levels, suggested a potential relationship between AS and Th17 cells under arthritic conditions. In vitro, AS was shown to block Th17 cell differentiation, as evidenced by the reduced percentages of CD4+ IL-17A+ T cells and decreased expression levels of RORγt, IL-17A, IL-17F, IL-21, and IL-22, without affecting the cell viability and apoptosis. This effect was attributed to the limited glycolysis, as indicated by metabolomics analysis, reduced glucose uptake, and pH measurements. Further investigation revealed that AS might bind to hexokinase2 (HK2) to down-regulate the protein levels of HK2 but not glyceraldehyde-3-phosphate dehydrogenase (GAPDH) or pyruvate kinase M2 (PKM2), and overexpression of HK2 reversed the inhibition of AS on Th17 cell differentiation. Furthermore, AS impaired the activation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signals in vivo and in vitro, which was abolished by the addition of lactate. In conclusion, AS significantly improved adjuvant-induced arthritis (AIA) in rats by inhibiting glycolysis-mediated activation of PI3K/AKT to restrain Th17 cell differentiation.
Collapse
Affiliation(s)
- Chunhong Jiang
- Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology (Shantou University and The University of Hong Kong), Shantou University Medical College, Shantou 515041, China
| | - Xi Zeng
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
| | - Jia Wang
- Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology (Shantou University and The University of Hong Kong), Shantou University Medical College, Shantou 515041, China
| | - Xiaoqian Wu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
| | - Lijuan Song
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Yang
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
| | - Ze Li
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
| | - Ning Xie
- State Key Laboratory of Innovative Natural Medicine and TCM Injection, Ganzhou 341000, China
| | - Xiaomei Yuan
- Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology (Shantou University and The University of Hong Kong), Shantou University Medical College, Shantou 515041, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China.
| | - Yi Guan
- Guangdong-Hong Kong Joint Laboratory for Emerging Infectious Diseases, Joint Institute of Virology (Shantou University and The University of Hong Kong), Shantou University Medical College, Shantou 515041, China; State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3
|
Yan L, Niu X, Liang K, Guan F, Yu X, Ye Z, Huang M, Liang H, Zhong X, Zeng J. Assessing the Role of Polyamine Metabolites in Blood and the DNA Methylation of Mycobacterium Tuberculosis in Patients with Multidrug-Resistant Tuberculosis. Int J Med Sci 2025; 22:1762-1772. [PMID: 40225858 PMCID: PMC11983307 DOI: 10.7150/ijms.102568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/23/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Tuberculosis (TB) is the second largest infectious disease killer in China, and the increasing prevalence of drug-resistant TB patients complicates treatment efforts and raises associated costs. Research on the mechanisms and characteristics of drug-resistant TB contributes to the discovery of new drug targets and the development of new anti-tuberculosis drugs. Methods: In this study, high-performance liquid chromatography (HPLC) was used to detect the content of polyamine metabolites, while western blotting, qPCR and ELISA were used to detect the expression of polyamine metabolism-related enzymes. The Oxford Nanopore Technologies (ONT) sequencing was applied to profile DNA methylation in multidrug-resistant Mycobacterium tuberculosis (Mtb). Gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed on the screened differentially hypermethylated genes. Furthermore, STRING and Cytoscape software were used to construct a protein-protein interaction (PPI) network to identify the key genes. Results: The findings indicated the spermidine (SPD) and polyamine metabolism-related enzymes were elevated in the peripheral blood of TB patients. In addition, the production of polyamines and polyamine metabolism-related enzymes was increased in the peripheral blood of multidrug-resistant tuberculosis (MDR-TB) patients. GO and KEGG analyses showed that the differentially hypermethylated genes were mainly enriched in arginine metabolism. The PPI network analysis identified the top five key genes with the highest degrees: moaX, vapC49, vapB49, highA3 and nuoC. Conclusions: Polyamine metabolites were increased in the peripheral blood of MDR-TB patients. The differentially hypermethylated genes in multidrug-resistant Mtb are involved in the arginine biosynthetic process, the differentially methylated genes may play an important biological role in the multidrug resistance of Mtb.
Collapse
Affiliation(s)
- Li Yan
- Dongguan Key Laboratory of Tuberculosis Prevention and Control, Dongguan Sixth People's Hospital, Dongguan 523008, Guangdong, China
| | - Xinxin Niu
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, Guangdong, China
| | - Kuidi Liang
- Dongguan Key Laboratory of Tuberculosis Prevention and Control, Dongguan Sixth People's Hospital, Dongguan 523008, Guangdong, China
| | - Feifeng Guan
- Dongguan Key Laboratory of Tuberculosis Prevention and Control, Dongguan Sixth People's Hospital, Dongguan 523008, Guangdong, China
| | - Xiaolin Yu
- Dongguan Key Laboratory of Tuberculosis Prevention and Control, Dongguan Sixth People's Hospital, Dongguan 523008, Guangdong, China
| | - Ziyu Ye
- Xinghai Institute of Cell, Guangdong Xianhua Institute for Medical Research, Dongguan 523808, Guangdong, China
| | - Mingyuan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, Guangdong, China
| | - Hancheng Liang
- Dongguan Key Laboratory of Tuberculosis Prevention and Control, Dongguan Sixth People's Hospital, Dongguan 523008, Guangdong, China
| | - Xinguang Zhong
- Dongguan Key Laboratory of Tuberculosis Prevention and Control, Dongguan Sixth People's Hospital, Dongguan 523008, Guangdong, China
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, Guangdong, China
- Xinghai Institute of Cell, Guangdong Xianhua Institute for Medical Research, Dongguan 523808, Guangdong, China
| |
Collapse
|
4
|
Cheng CW, Pedicini L, Alcala CM, Deligianni F, Smith J, Murray RD, Todd HJ, Forde N, McKeown L. RNA-seq analysis reveals transcriptome changes in livers from Efcab4b knockout mice. Biochem Biophys Rep 2025; 41:101944. [PMID: 40034259 PMCID: PMC11872658 DOI: 10.1016/j.bbrep.2025.101944] [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: 07/22/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
EFCAB4B is an evolutionarily conserved protein that encodes for the Rab GTPase Rab46, and the CRAC channel modulator, CRACR2A. Previous genome wide association studies have demonstrated the association of EFCAB4B variants in the progression of non-alcoholic fatty liver disease (NAFLD). In this study we show that mice with global depletion of Efcab4b -/- have significantly larger livers than their wild-type (WT) counterparts. We performed RNA-sequencing (RNA-seq) analysis of liver tissues to investigate differential global gene expression among Efcab4b -/- and WT mice. Of the 69 differentially expressed genes (DEGs), analyses of biological processes found significant enrichment in liver and bile development, with 6 genes (Pck1, Aacs, Onecut1, E2f8, Xbp1, and Hes1) involved in both processes. Specific consideration of possible roles of DEGs or their products in NAFLD progression to (NASH) and hepatocarcinoma (HCC), demonstrated DEGs in the livers of Efcab4b -/- mice had roles in molecular pathways including lipid metabolism, inflammation, ER stress and fibrosis. The results in this study provide additional insights into molecular mechanisms responsible for increasing susceptibility of liver injuries associated with EFCAB4B.
Collapse
Affiliation(s)
- Chew W. Cheng
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Lucia Pedicini
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Cintli Morales Alcala
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Fenia Deligianni
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Jessica Smith
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Ryan D. Murray
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Harriet J. Todd
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Niamh Forde
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| | - Lynn McKeown
- University of Leeds, Faculty of Medicine and Health, Leeds Institute of Cardiovascular and Metabolic Medicine, Leeds, LS2 9JT, UK
| |
Collapse
|
5
|
Jin R, Li H, Nan S, Wang H. FOXA1 co-activates circODC1 and ODC1 in HPV-positive cervical cancer cell growth. Syst Biol Reprod Med 2024; 70:113-123. [PMID: 38743820 DOI: 10.1080/19396368.2024.2311639] [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: 09/07/2022] [Accepted: 11/22/2023] [Indexed: 05/16/2024]
Abstract
As demonstrated in previous research, hsa_circ_0052602 (circODC1) is dynamically expressed in HPV-positive cervical cancer (CC). CircODC1 expression was quantified using qRT-PCR, and its role in CC cell growth was assessed via loss-of-function assays. Interactions between miR-607 and circODC1 or ODC1 were confirmed using bioinformatics and mechanistic assays. The association of FOXA1 with the circODC1 promoter was validated through ChIP and luciferase reporter assays. CircODC1 was highly expressed in HPV-positive CC cell lines, and its depletion significantly impeded malignant processes such as proliferation, migration, and invasion. We found that ODC1 also played an oncogenic role in HPV-positive CC cells. CircODC1 was shown to positively regulate ODC1 as a ceRNA, competitively binding to miR-607 to counteract its suppression of ODC1. HPV-associated FOXA1 was identified as a potential transcription factor of circODC1. Restoration experiments showed that overexpression of circODC1 could counterbalance the inhibitory effect of FOXA1 knockdown. These findings offer new insights into therapeutic strategies for HPV-positive CC patients.
Collapse
Affiliation(s)
- Rong Jin
- Department of Gynaecology and Obstetrics, the Fifth Center Hospital of Tianjin, Tianjin, China
| | - Hongfang Li
- Department of Gynaecology and Obstetrics, the First People's Hospital of Lanzhou, Lanzhou City, Gansu, China
| | - Shoushan Nan
- Department of Gastroenterology, the Fifth Center Hospital of Tianjin, Tianjin, China
| | - Huiju Wang
- Department of Gynaecology and Obstetrics, the Fifth Center Hospital of Tianjin, Tianjin, China
| |
Collapse
|
6
|
Sabir DK, Bin Jumah JA, Ancy I. Unveiling key drivers of hepatocellular carcinoma: a synergistic approach with network pharmacology, machine learning-driven ligand discovery and dynamic simulations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:1045-1070. [PMID: 39750056 DOI: 10.1080/1062936x.2024.2434577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) ranks fourth in cancer-related mortality worldwide. This study aims to uncover the genes and pathways involved in HCC through network pharmacology (NP) and to discover potential drugs via machine learning (ML)-based ligand screening. Additionally, toxicity prediction, molecular docking, and molecular dynamics (MD) simulations were conducted. NP study identified key genes related to HCC, particularly the enzymes AKT1 and GSK3β. Pathway analysis revealed that crucial pathways like PI3K-AKT and WNT signalling play pivotal roles in HCC progression. Using ML, potential inhibitors for AKT1 and GSK3β were identified, including CHEMBL2177361 and CHEMBL403354 for AKT1, and CHEMBL3652546 and CHEMBL4641631 for GSK3β. post-MD analyses, including RMSD, 2D-RMSD, RMSD cluster, RMSF, PCA, DCCM, residence time analysis, diffusion coefficient, autoencoder-based dimensionality reduction, FEL and MM/GBSA were performed to understand the protein-ligand interactions. The present study reveals the stable interactions of the inhibitors with AKT1 and GSK3β. The binding free energies of all the four complexes were -39.9, -46.8, -41.6, and -45.9 kcal/mol, respectively. This research provides novel insights into the genes and pathways involved in the progression and pathogenesis of HCC using bioinformatics tools. Furthermore, ML-based virtual screening identified potent inhibitors against the target proteins of HCC, such as AKT1 and GSK3β.
Collapse
Affiliation(s)
- D K Sabir
- Department of Medical Surgical Nursing, College of Nursing, Princess Nourahbint Abdulrahman University, Riyadh, Saudi Arabia
| | - J A Bin Jumah
- Nursing Administration & Education Department, College of Nursing, King Saud University, Riyadh, Saudi Arabia
| | - I Ancy
- Research and Development Center, Bioinnov Solutions LLP, Salem, India
| |
Collapse
|
7
|
Li YP, Huang ZJ, He QK, Li YX, Zhao XP, Ma ZQ, Qin MJ, Chen AW, Wei Q, Wang Y, Lu CH. Pirin Promotes the Progression of Non-Small-Cell Lung Cancer by Increasing ODC1 to Suppress Autophagy. J Proteome Res 2024; 23:1713-1724. [PMID: 38648079 DOI: 10.1021/acs.jproteome.3c00871] [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] [Indexed: 04/25/2024]
Abstract
Non-small-cell lung cancer (NSCLC), a common malignant tumor, requires deeper pathogenesis investigation. Autophagy is an evolutionarily conserved lysosomal degradation process that is frequently blocked during cancer progression. It is an urgent need to determine the novel autophagy-associated regulators in NSCLC. Here, we found that pirin was upregulated in NSCLC, and its expression was positively correlated with poor prognosis. Overexpression of pirin inhibited autophagy and promoted NSCLC proliferation. We then performed data-independent acquisition-based quantitative proteomics to identify the differentially expressed proteins (DEPs) in pirin-overexpression (OE) or pirin-knockdown (KD) cells. Among the pirin-regulated DEPs, ornithine decarboxylase 1 (ODC1) was downregulated in pirin-KD cells while upregulated along with pirin overexpression. ODC1 depletion reversed the pirin-induced autophagy inhibition and pro-proliferation effect in A549 and H460 cells. Immunohistochemistry showed that ODC1 was highly expressed in NSCLC cancer tissues and positively related with pirin. Notably, NSCLC patients with pirinhigh/ODC1high had a higher risk in terms of overall survival. In summary, we identified pirin and ODC1 as a novel cluster of prognostic biomarkers for NSCLC and highlighted the potential oncogenic role of the pirin/ODC1/autophagy axis in this cancer type. Targeting this pathway represents a possible therapeutic approach to treat NSCLC.
Collapse
Affiliation(s)
- Yan-Ping Li
- Research Laboratory of Zhuang & Yao Medicine, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, China
| | - Zi-Jia Huang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Quan-Kuo He
- Medical College of Guangxi University, Nanning, 530004 China
| | - Yi-Xiang Li
- Medical College of Guangxi University, Nanning, 530004 China
| | - Xiang-Pei Zhao
- Research Laboratory of Zhuang & Yao Medicine, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, China
| | - Zhong-Qi Ma
- Medical College of Guangxi University, Nanning, 530004 China
| | - Mei-Jing Qin
- Medical Experimental Center, The First People's Hospital of Nanning, Nanning Institute of Respiratory Diseases, Nanning, 530022 China
| | - Ai-Wen Chen
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiu Wei
- Medical Experimental Center, The First People's Hospital of Nanning, Nanning Institute of Respiratory Diseases, Nanning, 530022 China
| | - Yang Wang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Chun-Hua Lu
- Medical Experimental Center, The First People's Hospital of Nanning, Nanning Institute of Respiratory Diseases, Nanning, 530022 China
| |
Collapse
|
8
|
Su J, Xie Q, Xie L. Identification and validation of a metabolism-related gene signature for predicting the prognosis of paediatric medulloblastoma. Sci Rep 2024; 14:7540. [PMID: 38553479 PMCID: PMC10980764 DOI: 10.1038/s41598-024-57549-2] [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: 11/07/2023] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
Medulloblastoma (MB) is a malignant brain tumour that is highly common in children and has a tendency to spread to the brain and spinal cord. MB is thought to be a metabolically driven brain tumour. Understanding tumour cell metabolic patterns and characteristics can provide a promising foundation for understanding MB pathogenesis and developing treatments. Here, by analysing RNA-seq data of MB samples from the Gene Expression Omnibus (GEO) database, 12 differentially expressed metabolic-related genes (DE-MRGs) were chosen for the construction of a predictive risk score model for MB. This model demonstrated outstanding accuracy in predicting the outcomes of MB patients and served as a standalone predictor. An evaluation of functional enrichment revealed that the risk score showed enrichment in pathways related to cancer promotion and the immune response. In addition, a high risk score was an independent poor prognostic factor for MB in patients with different ages, sexes, metastasis stages and subgroups (SHH and Group 4). Consistently, the metabolic enzyme ornithine decarboxylase (ODC1) was upregulated in MB patients with poor survival time. Inhibition of ODC1 in primary and metastatic MB cell lines decreased cell proliferation, migration and invasion but increased immune infiltration. This study could aid in identifying metabolic targets for MB as well as optimizing risk stratification systems and individual treatment plans for MB patients via the use of a metabolism-related gene prognostic risk score signature.
Collapse
Affiliation(s)
- Jun Su
- Department of Neurosurgery, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan children's hospital), No. 86 Ziyuan Road, Changsha, 410007, Hunan, China
| | - Qin Xie
- Department of Neurosurgery, Xiangya Hospital, Central South University, No. 86 Xiangya Road, Changsha, 410008, Hunan, China
| | - Longlong Xie
- Pediatrics Research Institute of Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital Of Xiangya School of Medicine, Central South University (Hunan children's hospital), No. 86 Ziyuan Road, Changsha, 410007, Hunan, China.
| |
Collapse
|
9
|
Zhang H, Li X, Liu Z, Lin Z, Huang K, Wang Y, Chen Y, Liao L, Wu L, Xie Z, Hou J, Zhang X, Liu H. Elevated expression of HIGD1A drives hepatocellular carcinoma progression by regulating polyamine metabolism through c-Myc-ODC1 nexus. Cancer Metab 2024; 12:7. [PMID: 38395945 PMCID: PMC10893642 DOI: 10.1186/s40170-024-00334-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Hypoxia contributes to cancer progression through various molecular mechanisms and hepatocellular carcinoma (HCC) is one of the most hypoxic malignancies. Hypoxia-inducible gene domain protein-1a (HIGD1A) is typically induced via epigenetic regulation and promotes tumor cell survival during hypoxia. However, the role of HIGD1A in HCC remains unknown. METHODS HIGD1A expression was determined in 24 pairs of human HCC samples and para-tumorous tissues. Loss-of-function experiments were conducted both in vivo and in vitro to explore the role of HIGD1A in HCC proliferation and metastasis. RESULTS Increased HIGD1A expression was found in HCC tissues and cell lines, which was induced by hypoxia or low-glucose condition. Moreover, HIGD1A knockdown in HCC cells arrested the cell cycle at the G2/M phase and promoted hypoxia-induced cell apoptosis, resulting in great inhibition of cell proliferation, migration, and invasion, as well as tumor xenograft formation. Interestingly, these anti-tumor effects were not observed in normal hepatocyte cell line L02. Further, HIGD1A knockdown suppressed the expression of ornithine decarboxylase 1 (ODC1), a rate-limiting enzyme of polyamine metabolism under c-Myc regulation. HIGD1A was found to bind with the c-Myc promoter region, and its knockdown decreased the levels of polyamine metabolites. Consistently, the inhibitory effect on HCC phenotype by HIGD1A silencing could be reversed by overexpression of c-Myc or supplementation of polyamines. CONCLUSIONS Our results demonstrated that HIGD1A activated c-Myc-ODC1 nexus to regulate polyamine synthesis and to promote HCC survival and malignant phenotype, implying that HIGD1A might represent a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Haixing Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoran Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziying Liu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zimo Lin
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kuiyuan Huang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiran Wang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Leyi Liao
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Leyuan Wu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhanglian Xie
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Hongyan Liu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Zhang X, Wu L, Jia L, Hu X, Yao Y, Liu H, Ma J, Wang W, Li L, Chen K, Liu B. The implication of integrative multiple RNA modification-based subtypes in gastric cancer immunotherapy and prognosis. iScience 2024; 27:108897. [PMID: 38318382 PMCID: PMC10839690 DOI: 10.1016/j.isci.2024.108897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/28/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Previous studies have focused on the impact of individual RNA modifications on tumor development. This study comprehensively investigated the effects of multiple RNA modifications, including m6A, alternative polyadenylation, pseudouridine, adenosine-to-inosine editing, and uridylation, on gastric cancer (GC). By analyzing 1,946 GC samples from eleven independent cohorts, we identified distinct clusters of RNA modification genes with varying survival rates and immunological characteristics. We assessed the chromatin activity of these RNA modification clusters through regulon enrichment analysis. A prognostic model was developed using Stepwise Regression and Random Survival Forest algorithms and validated in ten independent datasets. Notably, the low-risk group showed a more favorable prognosis and positive response to immune checkpoint blockade therapy. Single-cell RNA sequencing confirmed the abundant expression of signature genes in B cells and plasma cells. Overall, our findings shed light on the potential significance of multiple RNA modifications in GC prognosis, stemness development, and chemotherapy resistance.
Collapse
Affiliation(s)
- Xiangnan Zhang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Liuxing Wu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Liqing Jia
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Xin Hu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Yanxin Yao
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Huahuan Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Junfu Ma
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Wei Wang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Lian Li
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| | - Ben Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Key Laboratory of Prevention and Control of Human Major Diseases, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin 300060, China
| |
Collapse
|
11
|
Petersen J, Englmaier L, Artemov AV, Poverennaya I, Mahmoud R, Bouderlique T, Tesarova M, Deviatiiarov R, Szilvásy-Szabó A, Akkuratov EE, Pajuelo Reguera D, Zeberg H, Kaucka M, Kastriti ME, Krivanek J, Radaszkiewicz T, Gömöryová K, Knauth S, Potesil D, Zdrahal Z, Ganji RS, Grabowski A, Buhl ME, Zikmund T, Kavkova M, Axelson H, Lindgren D, Kramann R, Kuppe C, Erdélyi F, Máté Z, Szabó G, Koehne T, Harkany T, Fried K, Kaiser J, Boor P, Fekete C, Rozman J, Kasparek P, Prochazka J, Sedlacek R, Bryja V, Gusev O, Adameyko I. A previously uncharacterized Factor Associated with Metabolism and Energy (FAME/C14orf105/CCDC198/1700011H14Rik) is related to evolutionary adaptation, energy balance, and kidney physiology. Nat Commun 2023; 14:3092. [PMID: 37248239 PMCID: PMC10226981 DOI: 10.1038/s41467-023-38663-7] [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: 09/24/2021] [Accepted: 05/11/2023] [Indexed: 05/31/2023] Open
Abstract
In this study we use comparative genomics to uncover a gene with uncharacterized function (1700011H14Rik/C14orf105/CCDC198), which we hereby name FAME (Factor Associated with Metabolism and Energy). We observe that FAME shows an unusually high evolutionary divergence in birds and mammals. Through the comparison of single nucleotide polymorphisms, we identify gene flow of FAME from Neandertals into modern humans. We conduct knockout experiments on animals and observe altered body weight and decreased energy expenditure in Fame knockout animals, corresponding to genome-wide association studies linking FAME with higher body mass index in humans. Gene expression and subcellular localization analyses reveal that FAME is a membrane-bound protein enriched in the kidneys. Although the gene knockout results in structurally normal kidneys, we detect higher albumin in urine and lowered ferritin in the blood. Through experimental validation, we confirm interactions between FAME and ferritin and show co-localization in vesicular and plasma membranes.
Collapse
Affiliation(s)
- Julian Petersen
- Department of Orthodontics, University Leipzig Medical Center, Leipzig, Germany.
| | - Lukas Englmaier
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 1090, Vienna, Austria
| | - Artem V Artemov
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Irina Poverennaya
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Ruba Mahmoud
- Department of Orthodontics, University Leipzig Medical Center, Leipzig, Germany
| | - Thibault Bouderlique
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Ruslan Deviatiiarov
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Endocrinology Research Center, Moscow, Russia
| | - Anett Szilvásy-Szabó
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, 1083, Budapest, Hungary
| | - Evgeny E Akkuratov
- Department of Applied Physics, Royal Institute of Technology, Science for Life Laboratory, 171 65, Stockholm, Sweden
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, OX3 9DS, UK
| | - David Pajuelo Reguera
- Institute of Molecular Genetics of the Czech Academy of Science, Czech Centre for Phenogenomics, Vestec, Czech Republic
| | - Hugo Zeberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marketa Kaucka
- Max Planck Institute for Evolutionary Biology, Plön, 24306, Germany
| | - Maria Eleni Kastriti
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomasz Radaszkiewicz
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Kristína Gömöryová
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Sarah Knauth
- Department of Orthodontics, University Leipzig Medical Center, Leipzig, Germany
| | - David Potesil
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zbynek Zdrahal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Ranjani Sri Ganji
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Anna Grabowski
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Miriam E Buhl
- Institute of Pathology & Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Michaela Kavkova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Håkan Axelson
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Scheelevägen 2, Lund, Sweden
| | - David Lindgren
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Scheelevägen 2, Lund, Sweden
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Ferenc Erdélyi
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest, Hungary
| | - Zoltán Máté
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest, Hungary
| | - Gábor Szabó
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest, Hungary
| | - Till Koehne
- Department of Orthodontics, University Leipzig Medical Center, Leipzig, Germany
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Kaj Fried
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Peter Boor
- Institute of Pathology & Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, 1083, Budapest, Hungary
| | - Jan Rozman
- Institute of Molecular Genetics of the Czech Academy of Science, Czech Centre for Phenogenomics, Vestec, Czech Republic
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, avenue du Swing, 4367, Belvaux, Luxembourg
| | - Petr Kasparek
- Institute of Molecular Genetics of the Czech Academy of Science, Czech Centre for Phenogenomics, Vestec, Czech Republic
| | - Jan Prochazka
- Institute of Molecular Genetics of the Czech Academy of Science, Czech Centre for Phenogenomics, Vestec, Czech Republic
| | - Radislav Sedlacek
- Institute of Molecular Genetics of the Czech Academy of Science, Czech Centre for Phenogenomics, Vestec, Czech Republic
| | - Vitezslav Bryja
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Oleg Gusev
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria.
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
12
|
Zhang T, Sun Q, Shen C, Qian Y. Circular RNA circ_0003028 regulates cell development through modulating miR-498/ornithine decarboxylase 1 axis in hepatocellular carcinoma. Anticancer Drugs 2023; 34:507-518. [PMID: 36730287 DOI: 10.1097/cad.0000000000001457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Circular RNA has been revealed to participate in multiple biological functions and contribute to various diseases' progression. This study aims to clarify the role of circ_0003028 and its potential molecular mechanism in hepatocellular carcinoma (HCC). The levels of circ_0003028, miR-498, and ornithine decarboxylase 1 (ODC1) mRNA were examined by quantitative real-time PCR. The cell proliferation ability was detected via 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, colony formation, and 5-ethynyl-2'-deoxyuridine assays. The apoptotic rate was evaluated through flow cytometry. The migration and invasion capacity was tested by using wound healing assay and transwell assay. The protein levels of E-cadherin, N-cadherin, and vimentin were measured by western blot assay. The ceRNA regulatory mechanism of circ_0003028 was observed via dual-luciferase reporter and RNA pull-down assays. The mice xenograft models were constructed to confirm the oncogenicity of circ_0003028 in HCC in vivo . Circ_0003028 and ODC1 were upregulated, whereas miR-498 was downregulated in HCC tissues and cells. Circ_0003028 knockdown inhibited cell proliferation and metastasis, and promoted apoptosis. MiR-498 was a direct target of circ_0003028, and inhibition of miR-498 reversed the inhibitory effect of circ_0003028 silencing on HCC progression. Moreover, ODC1 was a direct target of miR-498 and ODC1 overexpression abated the anticancer roles of miR-498 in HCC. Additionally, circ_0003028 regulated ODC1 expression by sponging miR-498. Finally, we found that circ_0003028 could induce epithelial-mesenchymal transition of HCC cells by exosome pathway. In brief, the results demonstrated that circ_0003028 exerted tumourigenicity roles via miR-498/ODC1 signaling axis, providing a promising biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Tao Zhang
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei
| | - Qikai Sun
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei
| | - Chao Shen
- CT Room, Hunan Zhongya Imaging Clinic, Changsha, China
| | - Yeben Qian
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei
| |
Collapse
|
13
|
Imran M, Sachdeva G, Menon S, Das D, Davuluri S, Acharya K, Chaudhari U. Therapeutic metformin concentrations positively regulate proliferation in endometrial epithelial cells via mTOR activation and augmented mitochondrial strength. Can J Physiol Pharmacol 2023; 101:52-64. [PMID: 36322951 DOI: 10.1139/cjpp-2022-0307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metformin, an antidiabetic drug, has recently been repositioned in the treatment of several nondiabetic disorders, including reproductive disorders such as polycystic ovarian syndrome, where it improves endometrial functions. In vitro studies employing supratherapeutic concentrations (5-20 mmol/L) of metformin have reported antiproliferative effects on endometrial epithelial and stromal cells. However, animal and human studies have revealed that therapeutic serum concentrations of metformin range between 20 and 70 µmol/L. In the present study, the effect of therapeutic concentrations of metformin was studied on endometrial epithelial cells (EECs). Therapeutic concentrations of metformin induced proliferation in Ishikawa and HEC-1A cells. The proliferation of EECs was found to be mammalian target of rapamycin (mTOR) dependent. Interestingly, therapeutic metformin concentrations were not able to activate the classical AMP-activated protein kinase (AMPK) signaling. On the contrary, supratherapeutic metformin concentration (10 mmol/L) inhibited mTOR and activated AMPK signaling. Microarray analysis of metformin-treated HEC-1A cells revealed dose-dependent differential effects on biological pathways associated with translation, ribosomal RNA processing, mitochondrial translation, and cell proliferation. Therapeutic concentrations of metformin upregulated mitochondrial number as demonstrated by increased MitoTracker™ Red staining and enhanced succinate dehydrogenase expression; however, higher concentration (10 mmol/L) abrogated the same. Our results suggest that therapeutic concentrations of metformin augment mitochondrial strength and induce mTOR-dependent endometrial cell proliferation.
Collapse
Affiliation(s)
- M Imran
- Cell Physiology and Pathology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Geetanjali Sachdeva
- Cell Physiology and Pathology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Shyla Menon
- Stem Cell Biology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Dhanjit Das
- Stem Cell Biology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | | | - Kshitish Acharya
- Shodhaka Life Sciences Pvt. Ltd., Bengaluru, Karnataka, India.,Institute of Bioinformatics and Applied Biotechnology (IBAB), Bengaluru, Karnataka, India
| | - Uddhav Chaudhari
- Cell Physiology and Pathology Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| |
Collapse
|
14
|
Investigation of the Therapeutic Effect of Total Alkaloids of Corydalis saxicola Bunting on CCl 4-Induced Liver Fibrosis in Rats by LC/MS-Based Metabolomics Analysis and Network Pharmacology. Metabolites 2022; 13:metabo13010009. [PMID: 36676934 PMCID: PMC9866371 DOI: 10.3390/metabo13010009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Liver fibrosis is a pathological result of liver injury that usually leads to a pathophysiological wound healing response. The total alkaloids of Corydalis saxicola Bunting (TACS) have been used for hepatoprotective effects on the liver. However, its exact therapeutic mechanisms of liver fibrosis are not yet well understood. To explore the potential anti-fibrosis mechanism of TACS, metabolomics coupled with network pharmacology were applied to reveal the underlying mechanisms. Ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS) combined with multivariate statistical analyses were performed to estimate changes in metabolic profiles. As a result, a total of 23 metabolites in rats with liver fibrosis were altered; of these, 11 had been downregulated and 12 had been upregulated compared with the control group. After TACS treatment, the levels of 13 metabolites were significantly restored compared with the CCl4-treated group, of which 4 metabolites were up-regulated and 9 metabolites were down-regulated. Many of these metabolites are involved in the bile acid metabolism, glutathione metabolism, tryptophan metabolism and purine metabolism. Then, three key targets, including cytochrome P450 family1 subfamily A member 1 (CYP1A1), ornithine decarboxylase 1 (OCD1) and monoamine oxidase Type B (MAOB) were predicted as potential therapeutic targets of TACS against liver fibrosis through network pharmacology analysis. Finally, palmatine, tetrahydropalmatine and dehydrocavidine were screened as potential active compounds responsible for the anti-fibrosis effect of TACS by molecular docking analysis. This study reveals that TACS exerted anti-fibrosis effects by regulating the liver metabolic pathway with multiple components and multiple targets, which is helpful to further clarify the hepatoprotective mechanisms of natural plant extracts.
Collapse
|
15
|
Chen M, Nie Z, Huang D, Gao Y, Cao H, Zheng L, Zhang S. Development of a polyamine gene expression score for predicting prognosis and treatment response in clear cell renal cell carcinoma. Front Immunol 2022; 13:1048204. [PMID: 36505496 PMCID: PMC9732944 DOI: 10.3389/fimmu.2022.1048204] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Backgrounds Polyamine metabolism (PM) is closely related to the tumor microenvironment (TME) and is involved in antitumor immunity. Clear cell renal cell carcinoma (ccRCC) not only has high immunogenicity but also has significant metabolic changes. However, the role of PM in the immune microenvironment of ccRCC remains unclear. This study aimed to reveal the prognostic value of PM-related genes (PMRGs) expression in ccRCC and their correlation with the TME. Methods The expression levels PMRGs in different cells were characterized with single-cell sequencing analysis. The PMRG expression pattern of 777 ccRCC patients was evaluated based on PMRGs. Unsupervised clustering analysis was used in identifying PMRG expression subtypes, and Lasso regression analysis was used in developing polyamine gene expression score (PGES), which was validated in external and internal data sets. The predictive value of PGES for immunotherapy was validated in the IMvigor210 cohort. Multiple algorithms were used in analyzing the correlation between PGES and immune cells. The sensitivity of PGES to chemotherapeutic drugs was analyzed with the "pRRophetic" package. We validated the genes that develop PGES in tissue samples. Finally, weighted gene co-expression network analysis was used in identifying the key PMRGs closely related to ccRCC, and cell function experiments were carried out. Results PMRGs were abundantly expressed on tumor cells, and PMRG expression was active in CD8+ T cells and fibroblasts. We identified three PMRG expression subtypes. Cancer and immune related pathways were active in PMRG expression cluster A, which had better prognosis. PGES exhibited excellent predictive value. The high-PGES group was characterized by high immune cell infiltration, high expression of T cell depletion markers, high tumor mutation burden and tumor immune dysfunction and exclusion, was insensitive to immunotherapy but sensitive to sunitinib, temsirolimus, and rapamycin, and had poor prognosis. Spermidine synthetase (SRM) has been identified as a key gene and is highly expressed in ccRCC at RNA and protein levels. SRM knockdown can inhibit ccRCC cell proliferation, migration, and invasion. Conclusions We revealed the biological characteristics of PMRG expression subtypes and developed PGES to accurately predict the prognosis of patients and response to immunotherapy.
Collapse
|
16
|
Transcriptome Analysis Reveals Critical Factors For Survival After Adenovirus Serotype 4 Infection. Poult Sci 2022; 102:102150. [PMID: 36989855 PMCID: PMC10070941 DOI: 10.1016/j.psj.2022.102150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/05/2022] [Accepted: 08/16/2022] [Indexed: 11/24/2022] Open
Abstract
Fowl adenovirus serotype-4 (FAdV-4) is highly lethal to poultry, making it one of the leading causes of economic losses in the poultry industry. However, a small proportion of poultry can survive after FAdV-4 infection. It is unclear whether there are genetic factors that protect chickens from FAdV-4 infection. Therefore, the livers from chickens uninfected with FAdV-4 (Normal), dead after FAdV-4 infection (Dead) or surviving after FAdV-4 infection (Survivor) were collected for RNA-seq, and 2,649 differentially expressed genes (DEGs) were identified. Among these, many immune-related cytokines and chemokines were significantly upregulated in the Dead group compared with the Survivor group, which might indicate that death is related to an excessive inflammatory immune response (cytokine storm). Subsequently, the KEGG results for DEGs specifically expressed in each comparison group indicated that cell cycle and apoptosis-related DEGs were upregulated and metabolism-related DEGs were downregulated in the Dead group, which also validated the reliability of the samples. Furthermore, GO and KEGG results showed DEGs expressed in all three groups were mainly associated with cell cycle. Among them, BRCA1, CDK1, ODC1, and MCM3 were screened as factors that might influence FAdV-4 infection. The qPCR results demonstrated that these 4 factors were not only upregulated in the Dead group but also significantly upregulated in the LMH cells after 24 h infection by FAdV-4. Moreover, interfering with BRCA1, CDK1, ODC1, and MCM3 significantly attenuated viral replication of FAdV-4. And interfering of BRCA1, CDK1, and MCM3 had more substantial hindering effects. These results provided novel insights into the molecular changes following FAdV-4 infection but also shed light on potential factors driving the survival of FAdV-4 infection in chickens.
Collapse
|
17
|
Tham SM, Rahmat JN, Chiong E, Wu Q, Esuvaranathan K, Mahendran R. Intravesical High Dose BCG Tokyo and Low Dose BCG Tokyo with GMCSF+IFN α Induce Systemic Immunity in a Murine Orthotopic Bladder Cancer Model. Biomedicines 2021; 9:biomedicines9121766. [PMID: 34944584 PMCID: PMC8698822 DOI: 10.3390/biomedicines9121766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 11/21/2022] Open
Abstract
This study evaluates a short therapy schedule for bladder cancer using BCG Tokyo. BCG Tokyo was evaluated in vitro using bone marrow derived dendritic cells, neutrophils, RAW macrophages and the murine bladder cancer cell line, MB49PSA, and compared to other BCG strains. BCG Tokyo > BCG TICE at inducing cytokine production. In vivo, high dose (1 × 107 colony forming units (cfu)) and low dose (1 × 106 cfu) BCG Tokyo with and without cytokine genes (GMCSF + IFNα) were evaluated in C57BL/6J mice (n = 12–16 per group) with orthotopically implanted MB49PSA cells. Mice were treated with four instillations of cytokine gene therapy and BCG therapy. Both high dose BCG alone and low dose BCG combined with cytokine gene therapy were similarly effective. In the second part the responsive groups, mice (n = 27) were monitored by urinary PSA analysis for a further 7 weeks after therapy cessation. More mice were cured at day 84 than at day 42 confirming activation of the immune system. Cured mice resisted the re-challenge with subcutaneous tumors unlike naïve, age matched mice. Antigen specific T cells recognizing BCG, HY and PSA were identified. Thus, fewer intravesical instillations, with high dose BCG Tokyo or low dose BCG Tokyo with GMCSF + IFNα gene therapy, can induce effective systemic immunity.
Collapse
Affiliation(s)
- Sin Mun Tham
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, 1E Kent Ridge Road, Singapore 119228, Singapore; (S.M.T.); (J.N.R.); (E.C.); (K.E.)
| | - Juwita N. Rahmat
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, 1E Kent Ridge Road, Singapore 119228, Singapore; (S.M.T.); (J.N.R.); (E.C.); (K.E.)
- Department of Bioengineering, National University of Singapore, Singapore 119077, Singapore
| | - Edmund Chiong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, 1E Kent Ridge Road, Singapore 119228, Singapore; (S.M.T.); (J.N.R.); (E.C.); (K.E.)
- Department of Urology, National University Hospital, National University Health System, Singapore 119228, Singapore;
| | - Qinghui Wu
- Department of Urology, National University Hospital, National University Health System, Singapore 119228, Singapore;
| | - Kesavan Esuvaranathan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, 1E Kent Ridge Road, Singapore 119228, Singapore; (S.M.T.); (J.N.R.); (E.C.); (K.E.)
- Department of Urology, National University Hospital, National University Health System, Singapore 119228, Singapore;
| | - Ratha Mahendran
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 8, 1E Kent Ridge Road, Singapore 119228, Singapore; (S.M.T.); (J.N.R.); (E.C.); (K.E.)
- Correspondence: ; Tel.: +65-6601-3982; Fax: +65-6777-8427
| |
Collapse
|
18
|
Ke M, Qian J, Hao F, Li X, Wu H, Luo X, Xu B, Gu C, Yang Y. Acupuncture Synergized With Bortezomib Improves Survival of Multiple Myeloma Mice via Decreasing Metabolic Ornithine. Front Oncol 2021; 11:779562. [PMID: 34804983 PMCID: PMC8596548 DOI: 10.3389/fonc.2021.779562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy worldwide in urgent need for novel therapeutic strategies. Since Velcade (bortezomib) was approved for the treatment of relapsed/refractory MM in 2003, we have seen considerable improvement in extending MM patient survival. However, most patients are fraught with high recurrence rate and incurability. Acupuncture is known for alleviating patient symptoms and improving the quality of life, but it is not well investigated in MM, especially in combination with bortezomib. In this study, we employed LC-MS and UHPLC-MS together with bioinformatics methods to test serum samples from 5TMM3VT MM murine model mice with four different treatments [control (C) group, bortezomib (V) treatment group, acupuncture (A) group, and combined (VA) group]. MM mice in group VA had longer survival time than mice in group A or group V. Joint pathway analysis indicated the underlying arginine and proline metabolism pathway among the 32 significantly decreased metabolites in group VA. CCK-8 assay and in vivo experiments validated that ornithine, the metabolite of arginine, promoted MM cell proliferation. In addition, gene expression omnibus (GEO) database analysis suggested that MM patients with higher ornithine decarboxylase 1 (ODC1) expression were evidently associated with poor overall survival. In summary, this study demonstrates the synergistic effects of acupuncture and bortezomib on extending the survival of MM model mice and provides potential therapeutic targets in the treatment of MM.
Collapse
Affiliation(s)
- Mengying Ke
- Large Data Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.,School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Qian
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Hao
- Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinying Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongjie Wu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xian Luo
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Xu
- Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- Large Data Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.,School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
19
|
Kozinova M, Joshi S, Ye S, Belinsky MG, Sharipova D, Farma JM, Reddy SS, Litwin S, Devarajan K, Campos AR, Yu Y, Schwartz B, von Mehren M, Rink L. Combined Inhibition of AKT and KIT Restores Expression of Programmed Cell Death 4 (PDCD4) in Gastrointestinal Stromal Tumor. Cancers (Basel) 2021; 13:cancers13153699. [PMID: 34359600 PMCID: PMC8345102 DOI: 10.3390/cancers13153699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
The majority of gastrointestinal stromal tumor (GIST) patients develop resistance to the first-line KIT inhibitor, imatinib mesylate (IM), through acquisition of secondary mutations in KIT or bypass signaling pathway activation. In addition to KIT, AKT is a relevant target for inhibition, since the PI3K/AKT pathway is crucial for IM-resistant GIST survival. We evaluated the activity of a novel pan-AKT inhibitor, MK-4440 (formerly ARQ 751), as monotherapy and in combination with IM in GIST cell lines and preclinical models with varying IM sensitivities. Dual inhibition of KIT and AKT demonstrated synergistic effects in IM-sensitive and -resistant GIST cell lines. Proteomic analyses revealed upregulation of the tumor suppressor, PDCD4, in combination treated cells. Enhanced PDCD4 expression correlated to increased cell death. In vivo studies revealed superior efficacy of MK-4440/IM combination in an IM-sensitive preclinical model of GIST compared with either single agent. The combination demonstrated limited efficacy in two IM-resistant models, including a GIST patient-derived xenograft model possessing an exon 9 KIT mutation. These studies provide strong rationale for further use of AKT inhibition in combination with IM in primary GIST; however, alternative agents will need to be tested in combination with AKT inhibition in the resistant setting.
Collapse
Affiliation(s)
- Marya Kozinova
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (M.K.); (S.J.); (S.Y.); (M.G.B.); (D.S.); (M.v.M.)
- Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Shalina Joshi
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (M.K.); (S.J.); (S.Y.); (M.G.B.); (D.S.); (M.v.M.)
| | - Shuai Ye
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (M.K.); (S.J.); (S.Y.); (M.G.B.); (D.S.); (M.v.M.)
| | - Martin G. Belinsky
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (M.K.); (S.J.); (S.Y.); (M.G.B.); (D.S.); (M.v.M.)
| | - Dinara Sharipova
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (M.K.); (S.J.); (S.Y.); (M.G.B.); (D.S.); (M.v.M.)
| | - Jeffrey M. Farma
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (J.M.F.); (S.S.R.)
| | - Sanjay S. Reddy
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (J.M.F.); (S.S.R.)
| | - Samuel Litwin
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (S.L.); (K.D.)
| | - Karthik Devarajan
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (S.L.); (K.D.)
| | - Alex Rosa Campos
- Proteomics Core Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
| | - Yi Yu
- ArQuIe Inc., A Wholly-Owned Subsidiary of Merck & Co., Inc. (Known as MSD Outside the United States and Canada), Kenilworth, NJ 07033, USA; (Y.Y.); (B.S.)
| | - Brian Schwartz
- ArQuIe Inc., A Wholly-Owned Subsidiary of Merck & Co., Inc. (Known as MSD Outside the United States and Canada), Kenilworth, NJ 07033, USA; (Y.Y.); (B.S.)
| | - Margaret von Mehren
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (M.K.); (S.J.); (S.Y.); (M.G.B.); (D.S.); (M.v.M.)
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Lori Rink
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (M.K.); (S.J.); (S.Y.); (M.G.B.); (D.S.); (M.v.M.)
- Correspondence: ; Tel.: +1-(215)-214-1608
| |
Collapse
|
20
|
Ajit K, Murphy BD, Banerjee A. Elucidating evolutionarily conserved mechanisms of diapause regulation using an in silico approach. FEBS Lett 2021; 595:1350-1374. [PMID: 33650678 DOI: 10.1002/1873-3468.14064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/02/2021] [Accepted: 02/19/2021] [Indexed: 11/11/2022]
Abstract
Embryonic diapause is an enigmatic phenomenon that appears in diverse species. Although regulatory mechanisms have been established, there is much to be discovered. Herein, we have made the first comprehensive attempt to elucidate diapause regulatory mechanisms using a computational approach. We found transcription factors unique to promoters of genes in diapause species. From pathway analysis and STRING PPI networks, the signaling pathways regulated by these unique transcription factors were identified. The pathways were then consolidated into a model to combine various known mechanisms of diapause regulation. This work also highlighted certain transcription factors that may act as 'master transcription factors' to regulate the phenomenon. Promoter analysis further suggested evidence for independent evolution for some of regulatory elements involved in diapause.
Collapse
Affiliation(s)
- Kamal Ajit
- Department of Biological Sciences, BITS Pilani KK Birla Goa Campus, Goa, India
| | - Bruce D Murphy
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médicine Vétérinaire, Université Montréal, St-Hyacinthe, QC, Canada
| | - Arnab Banerjee
- Department of Biological Sciences, BITS Pilani KK Birla Goa Campus, Goa, India
| |
Collapse
|
21
|
Wang H, Zhao Q, Dong W, Yang L, Lu K, Guo X, Liu H, Wei H, Cheng Y, Wu Z, Li S. Radiosynthesis and evaluation of N 5-(2- 18F-fluoropropanyl) ornithine as a potential agent for tumor PET imaging. Nucl Med Biol 2021; 94-95:98-105. [PMID: 33621898 DOI: 10.1016/j.nucmedbio.2021.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/30/2021] [Accepted: 02/04/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Studies have confirmed that tumorigenesis is related to an imbalance of polyamine metabolism and over-expression of oncogenes resulting in the up-regulation of ornithine decarboxylase (ODC, the first rate-limiting enzyme for regulating intracellular polyamines biosynthesis), which has become a target for anti-tumor therapy. In this study, an ornithine derivative, N5-(2-[18F]fluoropropionyl) ornithine (N5-[18F]FPO), has been prepared and its potential utility for tumor PET imaging evaluated. METHODS N5-[18F]FPO was successfully prepared via a nucleophilic fluorination reaction and a subsequent efficient deprotection step. The in vitro and in vivo stability were determined by HPLC conducted in fetal bovine serum, saline and rat urine. Cellular uptake studies were conducted in HepG2 cells and the biodistribution and micro-PET/CT imaging performed in normal ICR mice and three tumor-bearing mice models, respectively. RESULTS Total synthesis time of N5-[18F]FPO was about 80 min with a radiochemical yield of 15% ± 6% (uncorrected, based on 18F-, n = 6) and a high radiochemical stability can be seen in vitro and vivo. The N5-[18F]FPO exhibited fast uptake in HepG2 cells and the cellular uptake ability of N5-[18F]FPO can be inhibited by L-ornithine and DFMO, which indicated that the transport pathway of N5-[18F]FPO is similar to that of L-ornithine, interacting with ODC after being transported into the cell. The biodistribution and micro-PET/CT images demonstrate that N5-[18F]FPO was excreted by the urinary system, and excellent tumor visualization with high tumor-to-background ratios can be observed in the three tumor-bearing mice models studied. CONCLUSION All the above results suggest that N5-[18F]FPO has the potential to be a novel radiotracer for imaging ODC expression in solid tumors.
Collapse
Affiliation(s)
- Hongliang Wang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China.
| | - Qinan Zhao
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Weixuan Dong
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Liu Yang
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Keyi Lu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Xiaoshan Guo
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Hua Wei
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Yan Cheng
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China
| | - Zhifang Wu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China.
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Shanxi Key Laboratory of Molecular Imaging, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China; Molecular Imaging Precision Medicine Collaborative Innovation Center of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, People's Republic of China.
| |
Collapse
|
22
|
Zeng Z, Lan J, Lei S, Yang Y, He Z, Xue Y, Chen T. Simultaneous Inhibition of Ornithine Decarboxylase 1 and Pyruvate Kinase M2 Exerts Synergistic Effects Against Hepatocellular Carcinoma Cells. Onco Targets Ther 2020; 13:11697-11709. [PMID: 33244237 PMCID: PMC7683510 DOI: 10.2147/ott.s240535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose Previously, we showed that lactate promoted the proliferation and mobility of hepatocellular carcinoma (HCC) cells by increasing the expression of ornithine decarboxylase 1 (ODC1). In this study, we determined the relationship between ODC1 and pyruvate kinase M2 (PKM2, a key lactate metabolism enzyme), and determined the combined effects of difluoromethylornithine (DFMO; an ODC1 inhibitor) and compound 3k (a PKM2 inhibitor) on HCC cells. Methods First, the relationship between PKM2 and ODC1 was analyzed using Western blotting, Cell Counting Kit (CCK)-8 assays, transwell assays, bioinformatics, quantitative real-time fluorescent PCR (qRT-PCR), and immunohistochemical staining. Thereafter, the ODC1 inhibitor DFMO and the PKM2 inhibitor compound 3k were employed. Their combined effects on HCC cell proliferation and mobility were evaluated via CCK-8 assay, flow cytometry, a subcutaneous xenograft tumor model in mice, wound healing assays, and transwell assays. Additionally, the effects of DFMO and compound 3k on the epithelial–mesenchymal transition phenotype and the AKT/GSK-3β/β-catenin pathway were explored using Western blotting and immunofluorescence. Results PKM2 knockdown significantly decreased the ODC1 expression, and the proliferation and invasion of HCC cells, while ODC1 overexpression reversed the inhibitory effects of PKM2 knockdown. Similarly, inhibition of ODC1 also decreased the expression of PKM2 via reducing the c-myc-induced transcription. PKM2 was co-expressed with ODC1 in HCC samples, while simultaneously upregulated PKM2 and ODC1 led to the poorest survival outcome. DFMO and compound 3k synergistically inhibited HCC cell proliferation, induced apoptosis, and suppressed cell mobility, as well as the EMT phenotype and the AKT/GSK-3β/β-catenin pathway. The AKT activator SC79 reversed the inhibitory effects. Conclusion PKM2/ODC1 are involved in a positive feedback loop. The simultaneous inhibition of ODC1 and PKM2 using DFMO and compound 3k exerts synergistic effects against HCC cells via the AKT/GSK-3β/β-catenin pathway. Thus, DFMO combined with compound 3k may be a novel effective strategy for treating HCC.
Collapse
Affiliation(s)
- Zhirui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| | - Jinzhi Lan
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| | - Shan Lei
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| | - Yushi Yang
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| | - Yan Xue
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| | - Tengxiang Chen
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550009, People's Republic of China
| |
Collapse
|
23
|
Li J, Meng Y, Wu X, Sun Y. Polyamines and related signaling pathways in cancer. Cancer Cell Int 2020; 20:539. [PMID: 33292222 PMCID: PMC7643453 DOI: 10.1186/s12935-020-01545-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Polyamines are aliphatic compounds with more than two amino groups that play various important roles in human cells. In cancer, polyamine metabolism dysfunction often occurs, and regulatory mechanisms of polyamine. This review summarizes the existing research on the metabolism and transport of polyamines to study the association of oncogenes and related signaling pathways with polyamines in tumor cells. Drugs that regulate enzymes have been developed for cancer treatment, and in the future, more attention should be paid to treatment strategies that simultaneously modulate polyamine metabolism and carcinogenic signaling pathways. In addition, the polyamine pathway is a potential target for cancer chemoprevention. As an irreversible suicide inhibitor of the ornithine decarboxylase (a vital enzyme of polyamine synthesis), Difluoro-methylornithine had been shown to have the chemoprevention effect on cancer. Therefore, we summarized and analyzed the chemoprophylaxis effect of the difluoromethylornithine in this systematic review.
Collapse
Affiliation(s)
- Jiajing Li
- Department of Otorhinolaryngology-Head and Neck Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China.,Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yan Meng
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, China
| | - Xiaolin Wu
- Department of Pathophysiology, Prostate Diseases Prevention and Treatment Research Center, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yuxin Sun
- Department of Otorhinolaryngology-Head and Neck Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
24
|
Guerra MF, Lacoste MG, Anzulovich AC, Makinistian L. Magnetic fields, cancer and circadian rhythms: hypotheses on the relevance of intermittence and cycling. Proc Biol Sci 2019; 286:20192337. [PMID: 31795871 DOI: 10.1098/rspb.2019.2337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- María Florencia Guerra
- Departamento de Física, Instituto de Física Aplicada (INFAP), Universidad Nacional de San Luis y Consejo Nacional de Investigaciones Científicas y Técnicas, Ejército de los Andes 950, 5700 San Luis, Argentina.,Laboratorio de Cronobiología, IMIBIO-SL (CONICET-UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - María Gabriela Lacoste
- Laboratorio de Cronobiología, IMIBIO-SL (CONICET-UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina.,Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Ana Cecilia Anzulovich
- Laboratorio de Cronobiología, IMIBIO-SL (CONICET-UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina.,Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Leonardo Makinistian
- Departamento de Física, Instituto de Física Aplicada (INFAP), Universidad Nacional de San Luis y Consejo Nacional de Investigaciones Científicas y Técnicas, Ejército de los Andes 950, 5700 San Luis, Argentina
| |
Collapse
|