1
|
Lin F, Li J, Zhou L, Yi R, Chen Y, He S. Targeting vulnerability in tumor therapy: Dihydroorotate dehydrogenase. Life Sci 2025; 371:123612. [PMID: 40187643 DOI: 10.1016/j.lfs.2025.123612] [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: 01/16/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Dihydroorotate dehydrogenase (DHODH) is a key enzyme in the de novo pyrimidine biosynthetic pathway and a recognized therapeutic target in various diseases. In oncology research, DHODH has gained increasing importance and become a hot target for various tumor therapy studies. This review highlights three key points: (1) DHODH enables its diverse biological functions through its unique structural features and dominates the regulation of tumor metabolism and cell fate; (2) DHODH activates oncogenic signals, drives metastatic adaptation, and remodels drug resistance networks in tumors, making it a metabolic-signaling dual hub; and (3) DHODH inhibitors have shown significant efficacy in preclinical models of various tumors but face multiple challenges in clinical trials, including drug-related limitations and external constraints. Given these challenges, future research should explore DHODH inhibitors as a foundation for overcoming technological and translational barriers while establishing a systematic framework for the clinical application of DHODH-targeted tumor therapies.
Collapse
Affiliation(s)
- Fu Lin
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Jiaxin Li
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Lei Zhou
- Laboratory of Pathogen Biology and Immunology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Rigui Yi
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Yingge Chen
- School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Shuai He
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China.
| |
Collapse
|
2
|
Liu Z, Li Z, Guo Y, Li Y, Xuan H. The protective effects of propolis against lipopolysaccharide-induced acute liver injury by modulating serum metabolites and gut flora. Sci Rep 2025; 15:16959. [PMID: 40374745 PMCID: PMC12081765 DOI: 10.1038/s41598-025-01343-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Propolis has significant hepatoprotective effects, but the active components, targets, and mechanisms have not been fully elucidated. Here, we integrated network pharmacology, serum metabolomics, and 16 S rRNA sequencing to disclose the hepatoprotective effects of Chinese propolis (CP) by lipopolysaccharide (LPS)-induced acute liver injury (ALI) in mice. The core active ingredients of CP against ALI, including quercetin, luteolin, and kaempferol, can bind stably to pro-inflammatory factors such as TNF-α, IL-6, IL-1β, and IFN-γ. CP and its active ingredient quercetin obviously alleviated LPS-induced ALI in mice and downregulated the levels of pro-inflammatory genes (Tnf-α, Il-1β, Il-6, Mcp-1, Ifn-γ, and Cox-2) while increasing the protein expression levels of the antioxidant factors Nrf2 and HO-1. Untargeted serum metabolomics analysis indicated that CP and quercetin ameliorated LPS-induced metabolic disorders mainly by modulating the ascorbate and aldarate metabolisms. 16 S rRNA sequencing demonstrated that CP and quercetin modulated the gut microbiota, augmenting the relative abundance of anti-inflammatory bacteria like Lactobacillus and Dubosiella and diminishing the pro-inflammatory bacteria like Alistipes. Spearman correlation analysis revealed that there existed significant correlations among inflammatory factors, gut microbiota, and differential metabolites of serum after propolis pretreatment. Our research indicated that propolis effectively alleviated pathological damage in LPS-induced ALI mice mainly through partially restoring the ecology of gut flora and metabolic disorders to reduce inflammation.
Collapse
Affiliation(s)
- Zhengxin Liu
- School of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Zongze Li
- School of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Yuyang Guo
- School of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Yajing Li
- The Department of Biopharmaceutical Technology, Zhejiang Institute of Economics and Trade, Hangzhou, 310018, China.
| | - Hongzhuan Xuan
- School of Life Sciences, Liaocheng University, Liaocheng, 252059, China.
| |
Collapse
|
3
|
Song C, Li Q, Zhang J, Hu W. Uridine Phosphorylase 1 as a Biomarker Associated with Glycolysis in Acute Lung Injury. Inflammation 2025:10.1007/s10753-025-02270-z. [PMID: 39969741 DOI: 10.1007/s10753-025-02270-z] [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: 11/06/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
The specific pathogenesis of acute lung injury (ALI) is complex and not yet clear, and the clinical treatment methods are relatively limited. It is of great clinical significance to explore its pathogenesis and effective molecular targets. Here, we identified an ALI biomarker (UPP1) associated with uridine metabolism by a systematic bioinformatics approach. It was also confirmed to be associated with the glycolytic pathway in the mouse ALI model. In addition, drug sensitivity analysis based on the CMAP database identified three UPP1-associated drugs (CAY10585, XL147 and IOX2) that may be useful in the treatment of ALI. Molecular docking and molecular dynamics simulations further confirmed the stability of the binding between UPP1 and the three drugs. In conclusion, this study confirms that the uridine metabolism gene UPP1 associated with glycolysis is a key biomarker of ALI and provides valuable insights into the potential application of CAY10585, XL147 and IOX2 in ALI.
Collapse
Affiliation(s)
- Congkuan Song
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Provincial Clinical Research Center for Cancer, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Qingqing Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jinjin Zhang
- Department of Critical Care Medicine, Wuhan Fourth Hospital, Wusheng Road, Wuhan, China
| | - Weidong Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China.
- Hubei Provincial Clinical Research Center for Cancer, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China.
| |
Collapse
|
4
|
Xue Q, Zhou X, Wang Y, Liu Y, Li X, Xiong C, Liu X, Huang Y. Mass spectrometry imaging reveals spatial metabolic variation and the crucial role of uridine metabolism in liver injury caused by Schistosoma japonicum. PLoS Negl Trop Dis 2025; 19:e0012854. [PMID: 39933005 PMCID: PMC11813095 DOI: 10.1371/journal.pntd.0012854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/18/2025] [Indexed: 02/13/2025] Open
Abstract
Schistosomiasis is the second most important parasitic disease worldwide. Schistosomiasis japonica is a unique species endemic to southern China, and schistosomiasis is characterized by severe liver injury, inflammation, liver granuloma, and subsequent liver fibrosis. However, the pathological mechanism of this disease remains unclear. Mass spectrometry imaging (MSI) is a versatile technique that integrates the molecular specificity of mass spectrometry (MS) with spatial imaging information, which could provide an accurate method for observing disease progression. In this study, we used an air flow-assisted desorption electrospray ionization (AFADESI-MSI) platform to detect a wide range of metabolites and visualize their distribution in the liver tissue of mice infected with Schistosoma japonicum. In the negative ion mode analysis, 21 and 25 different metabolites were detected in the early and chronic stages of infection, respectively. Thirteen characteristic metabolites and 3 metabolic pathways related to disease development may be involved in the chronicity of schistosomiasis. There were more than 32 and 40 region-specific changes in the abundance of a wide range of metabolites (including carbohydrates, amino acids, nucleotides, and fatty acids) in the livers of mice at two different infection times, which also revealed the heterogeneous metabolic characteristics of the liver egg granulomas of S. japonicum. In a chronic infection model with S. japonicum, oral treatment with praziquantel significantly alleviated most metabolic disorders, including fatty acid and pyrimidine metabolism. Surprisingly, Upase1, a key enzyme in uridine metabolism, was significantly upregulated 6 weeks after infection, and liver uridine levels were negatively correlated with the abundance of multiple lipid-associated metabolites. Further studies revealed that in vitro uridine supplementation inhibited the activation of LX-2 cells, restored the homeostasis of fatty acid metabolism through the peroxisome proliferator-activated receptor γ (PPARγ) pathway, and played an antifibrotic role. Our findings provide new insights into the molecular mechanisms of S. japonicum-induced liver fibrosis and the potential of targeting uridine metabolism in disease therapy.
Collapse
Affiliation(s)
- Qingkai Xue
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xiangyu Zhou
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yuyan Wang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi, Jiangsu, China
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yiyun Liu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi, Jiangsu, China
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojing Li
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi, Jiangsu, China
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunrong Xiong
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xinjian Liu
- Department of Pathogen Biology, Key Laboratory of Antibody Techniques of National Health Commission, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuzheng Huang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
- Tropical Diseases Research Center, Nanjing Medical University, Wuxi, Jiangsu, China
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Semmler G, Petrenko O, Lozano JJ, Shalaby S, Sánchez-Avila JI, Marella N, Hannich T, Wöran K, Balcar L, Simbrunner B, Lampichler K, Mozayani B, Trauner M, Mandorfer M, Reiberger T, García-Pagán JC, Scheiner B. Metabolomic profiles differentiate between porto-sinusoidal vascular disorder, cirrhosis, and healthy individuals. JHEP Rep 2024; 6:101208. [PMID: 39624234 PMCID: PMC11609546 DOI: 10.1016/j.jhepr.2024.101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/30/2024] [Accepted: 08/27/2024] [Indexed: 04/03/2025] Open
Abstract
Background & Aims Porto-sinusoidal vascular disorder (PSVD) is a rare and diagnostically challenging vascular liver disease. This study aimed to identify distinct metabolomic signatures in patients with PSVD or cirrhosis to facilitate non-invasive diagnosis and elucidate perturbed metabolic pathways. Methods Serum samples from 20 healthy volunteers (HVs), 20 patients with histologically confirmed PSVD or 20 patients with cirrhosis were analyzed. Metabolites were measured using liquid chromatography-mass spectrometry. Differential abundance was evaluated with Limma's moderated t-statistics. Artificial neural network and support vector machine models were developed to classify PSVD against cirrhosis or HV metabolomic profiles. An independent cohort was used for validation. Results A total of 283 metabolites were included for downstream analysis. Clustering effectively separated PSVD from HV metabolomes, although a subset of patients with PSVD (n = 5, 25%) overlapped with those with cirrhosis. Differential testing revealed significant PSVD-linked metabolic perturbations, including pertubations in taurocholic and adipic acids, distinguishing patients with PSVD from both HVs and those with cirrhosis. Alterations in pyrimidine, glycine, serine, and threonine pathways were exclusively associated with PSVD. Machine learning models utilizing selected metabolic signatures reliably differentiated the PSVD group from HVs or patients with cirrhosis using only 4 to 6 metabolites. Validation in an independent cohort demonstrated the high discriminative ability of taurocholic acid (AUROC 0.899) for patients with PSVD vs. HVs and the taurocholic acid/aspartic acid ratio (AUROC 0.720) for PSVD vs. cirrhosis. Conclusions High-throughput metabolomics enabled the identification of distinct metabolic profiles that differentiate between PSVD, cirrhosis, and healthy individuals. Unique alterations in the glycine, serine, and threonine pathways suggest their potential involvement in PSVD pathogenesis. Impact and implications Porto-sinusoidal vascular disorder (PSVD) is a vascular liver disease that can lead to pre-sinusoidal portal hypertension in the absence of cirrhosis, with poorly understood pathophysiology and no established treatment. Our study demonstrates that analyzing the serum metabolome could reveal distinct metabolic signatures in patients with PSVD, including alterations in the pyrimidine, glycine, serine and threonine pathways, potentially shedding light on the disease's underlying pathways. These findings could enable earlier and non-invasive diagnosis of PSVD, potentially reducing reliance on invasive procedures like liver biopsy and guiding diagnostic pathways.
Collapse
Affiliation(s)
- Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Oleksandr Petrenko
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | | | - Sarah Shalaby
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). CIBEREHD (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas). Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE-Liver). Departament de Medicina i Ciències de la Salut. Universitat de Barcelona, Spain
| | - Juan I. Sánchez-Avila
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Nara Marella
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Hannich
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Katharina Wöran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Lorenz Balcar
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Katharina Lampichler
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Behrang Mozayani
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Juan-Carlos García-Pagán
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). CIBEREHD (Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas). Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE-Liver). Departament de Medicina i Ciències de la Salut. Universitat de Barcelona, Spain
| | - Bernhard Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Ortega-Vallbona R, Palomino-Schätzlein M, Tolosa L, Benfenati E, Ecker GF, Gozalbes R, Serrano-Candelas E. Computational Strategies for Assessing Adverse Outcome Pathways: Hepatic Steatosis as a Case Study. Int J Mol Sci 2024; 25:11154. [PMID: 39456937 PMCID: PMC11508863 DOI: 10.3390/ijms252011154] [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: 09/20/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The evolving landscape of chemical risk assessment is increasingly focused on developing tiered, mechanistically driven approaches that avoid the use of animal experiments. In this context, adverse outcome pathways have gained importance for evaluating various types of chemical-induced toxicity. Using hepatic steatosis as a case study, this review explores the use of diverse computational techniques, such as structure-activity relationship models, quantitative structure-activity relationship models, read-across methods, omics data analysis, and structure-based approaches to fill data gaps within adverse outcome pathway networks. Emphasizing the regulatory acceptance of each technique, we examine how these methodologies can be integrated to provide a comprehensive understanding of chemical toxicity. This review highlights the transformative impact of in silico techniques in toxicology, proposing guidelines for their application in evidence gathering for developing and filling data gaps in adverse outcome pathway networks. These guidelines can be applied to other cases, advancing the field of toxicological risk assessment.
Collapse
Affiliation(s)
- Rita Ortega-Vallbona
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| | - Martina Palomino-Schätzlein
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, 46026 Valencia, Spain;
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos, 28029 Madrid, Spain
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Gerhard F. Ecker
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek Platz 2, 1090 Wien, Austria;
| | - Rafael Gozalbes
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
- MolDrug AI Systems S.L., Olimpia Arozena Torres 45, 46108 Valencia, Spain
| | - Eva Serrano-Candelas
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| |
Collapse
|
7
|
Chen ZY, Panga MJ, Zhang X, Qiao S, Chen S, Appiah C, Zhao Y. Estrogen alleviates liver fibrosis and restores metabolic homeostasis in ovariectomy-induced liver injury and carbon tetrachloride (CCl 4) exposure. Eur J Pharmacol 2024; 978:176774. [PMID: 38936452 DOI: 10.1016/j.ejphar.2024.176774] [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: 03/18/2024] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024]
Abstract
AIM Given estrogen's recognized regulatory influence on diverse metabolic and immune functions, this study sought to explore its potential impact on fibrosis and elucidate the underlying metabolic regulations. METHODS Female mice underwent ovary removal surgery, followed by carbon tetrachloride (CCl4) administration to induce liver injury. Biochemical index analysis and histopathological examination were then conducted. The expression levels of alpha-smooth muscle actin (α-SMA), transforming growth factor-β (TGF-β), and collagen type 1 alpha 1 chain (COL1A1) were assessed using western blotting to further elucidate the extent of liver injury. Finally, metabolite extraction and metabolomic analysis were performed to evaluate metabolic changes. RESULTS Ovary removal exacerbated CCl4-induced liver damage, while estrogen supplementation provided protection against hepatic changes resulting from OVX. Furthermore, estrogen mitigated liver injury induced by CCl4 treatment in vivo. Estrogen supplementation significantly restored liver damage induced by OVX and CCl4. Comparative analysis revealed significant alterations in pathways including aminoacyl-tRNA biosynthesis, glycine, serine, and threonine metabolism, lysine degradation, and taurine and hypotaurine metabolism in estrogen treatment. CONCLUSION Estrogen supplementation alleviates liver injury induced by OVX and CCl4, highlighting its protective effects against fibrosis and associated metabolic alterations.
Collapse
Affiliation(s)
- Zi Yi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Mogellah John Panga
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Xiangrui Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Shuai Qiao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Shitian Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Clara Appiah
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China.
| |
Collapse
|
8
|
Tan Z, Shen P, Wen Y, Sun HY, Liang HY, Qie HJ, Dai RW, Gao Y, Huang Z, Zhou W, Tang LJ. Assessment of metabolomic variations among individuals returning to plain areas after exposure to high altitudes: a metabolomic analysis of human plasma samples with high-altitude de-acclimatization syndrome. Front Mol Biosci 2024; 11:1375360. [PMID: 38962282 PMCID: PMC11220191 DOI: 10.3389/fmolb.2024.1375360] [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: 02/14/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND High altitude de-acclimatization (HADA) is gradually becoming a public health concern as millions of individuals of different occupations migrate to high-altitude areas for work due to economic growth in plateau areas. HADA affects people who return to lower elevations after exposure to high altitudes. It causes significant physiological and functional changes that can negatively impact health and even endanger life. However, uncertainties persist about the detailed mechanisms underlying HADA. METHODS We established a population cohort of individuals with HADA and assessed variations in metabolite composition. Plasm samples of four groups, including subjects staying at plain (P) and high altitude (H) as well as subjects suffering from HADA syndrome with almost no reaction (r3) and mild-to-moderate reaction (R3) after returning to plain from high altitude, were collected and analyzed by Liquid Chromatography-Mass Spectrometry metabolomic. Multivariate statistical analyses were used to explore significant differences and potential clinical prospect of metabolites. RESULT Although significantly different on current HADAS diagnostic symptom score, there were no differences in 17 usual clinical indices between r3 and R3. Further multivariate analyses showed isolated clustering distribution of the metabolites among the four groups, suggesting significant differences in their metabolic characteristics. Through K-means clustering analysis, we identified 235 metabolites that exhibited patterns of abundance change consistent with phenotype of HADA syndrome. Pathway enrichment analysis indicated a high influence of polyunsaturated fatty acids under high-altitude conditions. We compared the metabolites between R3 and r3 and found 107 metabolites with differential abundance involved in lipid metabolism and oxidation, suggesting their potential role in the regulation of oxidative stress homeostasis. Among them, four metabolites might play a key role in the occurrence of HADA, including 11-beta-hydroxyandrosterone-3-glucuronide, 5-methoxyindoleacetate, 9,10-epoxyoctadecenoic acid, and PysoPC (20:5). CONCLUSION We observed the dynamic variation in the metabolic process of HADA. Levels of four metabolites, which might be provoking HADA mediated through lipid metabolism and oxidation, were expected to be explore prospective indices for HADA. Additionally, metabolomics was more efficient in identifying environmental risk factors than clinical examination when dramatic metabolic disturbances underlying the difference in symptoms were detected, providing new insights into the molecular mechanisms of HADAS.
Collapse
Affiliation(s)
- Zhen Tan
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Pan Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yi Wen
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hong-yu Sun
- Department of Central Lab, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hong-yin Liang
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hua-ji Qie
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Rui-wu Dai
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhu Huang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Li-jun Tang
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Zhang W, Zhang L, Zhi L, Qi J, He J. A Mendelian randomization study of the entire phenome to explore the causal links between epilepsy. Brain Behav 2024; 14:e3602. [PMID: 38898641 PMCID: PMC11186849 DOI: 10.1002/brb3.3602] [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: 06/13/2023] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVE The causes and triggering factors of epilepsy are still unknown. The results of genome-wide association studies can be utilized for a phenome-wide association study using Mendelian randomization (MR) to identify potential risk factors for epilepsy. METHODS This study utilizes two-sample MR analysis to investigate whether 316 phenotypes, including lifestyle, environmental factors, blood biomarker, and more, are causally associated with the occurrence of epilepsy. The primary analysis employed the inverse variance weighted (IVW) model, while complementary MR analysis methods (MR Egger, Wald ratio) were also employed. Sensitivity analyses were also conducted to evaluate heterogeneity and pleiotropy. RESULTS There was no evidence of a statistically significant causal association between the examined phenotypes and epilepsy following Bonferroni correction (p < 1.58 × 10-4) or false discovery rate correction. The results of the MR analysis indicate that the frequency of tiredness or lethargy in the last 2 weeks (p = 0.042), blood uridine (p = 0.003), blood propionylcarnitine (p = 0.041), and free cholesterol (p = 0.044) are suggestive causal risks for epilepsy. Lifestyle choices, such as sleep duration and alcohol consumption, as well as biomarkers including steroid hormone levels, hippocampal volume, and amygdala volume were not identified as causal factors for developing epilepsy (p > 0.05). CONCLUSIONS Our study provides additional insights into the underlying causes of epilepsy, which will serve as evidence for the prevention and control of epilepsy. The associations observed in epidemiological studies may be partially attributed to shared biological factors or lifestyle confounders.
Collapse
Affiliation(s)
- Wei Zhang
- Department of NeurosurgeryBeijing Fengtai HospitalBeijingChina
| | - Li‐Ming Zhang
- Department of NeurosurgeryJinan University Affiliated 999 Brain HospitalGuangzhouChina
| | - Lin Zhi
- Department of NeurosurgeryBeijing Fengtai HospitalBeijingChina
| | - Ji Qi
- Department of NeurosurgeryBeijing Fengtai HospitalBeijingChina
| | - Jue He
- Department of NeurosurgeryBeijing Fengtai HospitalBeijingChina
| |
Collapse
|
10
|
Strefeler A, Blanco-Fernandez J, Jourdain AA. Nucleosides are overlooked fuels in central carbon metabolism. Trends Endocrinol Metab 2024; 35:290-299. [PMID: 38423899 DOI: 10.1016/j.tem.2024.01.013] [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: 11/02/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
From our daily nutrition and synthesis within cells, nucleosides enter the bloodstream and circulate throughout the body and tissues. Nucleosides and nucleotides are classically viewed as precursors of nucleic acids, but recently they have emerged as a novel energy source for central carbon metabolism. Through catabolism by nucleoside phosphorylases, the ribose sugar group is released and can provide substrates for lower steps in glycolysis. In environments with limited glucose, such as at sites of infection or in the tumor microenvironment (TME), cells can use, and may even require, this alternative energy source. Here, we discuss the implications of these new findings in health and disease and speculate on the potential new roles of nucleosides and nucleic acids in energy metabolism.
Collapse
Affiliation(s)
- Abigail Strefeler
- Department of Immunobiology, University of Lausanne, Ch. des Boveresses 155, CP51, 1066 Epalinges, Switzerland
| | - Joan Blanco-Fernandez
- Department of Immunobiology, University of Lausanne, Ch. des Boveresses 155, CP51, 1066 Epalinges, Switzerland
| | - Alexis A Jourdain
- Department of Immunobiology, University of Lausanne, Ch. des Boveresses 155, CP51, 1066 Epalinges, Switzerland.
| |
Collapse
|
11
|
Yan X, Liu H, Huang M, Zhang Y, Zeng B. Integrative proteomics and metabolomics explore the effect and mechanism of Qiyin granules on improving nonalcoholic fatty liver disease. Heliyon 2024; 10:e27075. [PMID: 38444462 PMCID: PMC10912341 DOI: 10.1016/j.heliyon.2024.e27075] [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: 09/20/2023] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as a prominent global health concern, representing a substantial burden within the spectrum of chronic liver diseases. Despite its escalating prevalence, a definitive therapeutic strategy or efficacious pharmacological intervention for NAFLD has yet to receive official approval to date. While Fu Fang Qiyin granules have exhibited efficacy in addressing NAFLD, the intricacies of their underlying mechanism of action remain inadequately elucidated. In this study, we substantiated the ameliorative impact of Qiyin on highfat diet (HFD)induced NAFLD in rat models. The results of metabonomics showed that 108 potential biomarkers in serum and urine related to amino acid metabolism, energy metabolism, and pyrimidine metabolism, have returned to normal levels compared to the model group. Hepatic transcriptomics further indicated that Qiyin potentially confers protective effects against NAFLD by mediating liver inflammation and fibrosis through lumican (LUM) and decorin (DCN). In summation, our investigation provides compelling evidence affirming the therapeutic promise of Qiyin for NAFLD. It elucidates the underlying mechanistic pathways, furnishing a compelling rationale for its prospective clinical application.
Collapse
Affiliation(s)
- Xuehua Yan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, People's Republic of China
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, 830017, People's Republic of China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Urumqi, Xinjiang, 830017, People's Republic of China
| | - Hongbing Liu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, 830017, People's Republic of China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Urumqi, Xinjiang, 830017, People's Republic of China
| | - Meng Huang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, 830017, People's Republic of China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Urumqi, Xinjiang, 830017, People's Republic of China
| | - Yujie Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, People's Republic of China
| | - Binfang Zeng
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, 830017, People's Republic of China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, Urumqi, Xinjiang, 830017, People's Republic of China
| |
Collapse
|
12
|
Huneault HE, Gent AE, Cohen CC, He Z, Jarrell ZR, Kamaleswaran R, Vos MB. Validation of a screening panel for pediatric metabolic dysfunction-associated steatotic liver disease using metabolomics. Hepatol Commun 2024; 8:e0375. [PMID: 38407264 PMCID: PMC10898657 DOI: 10.1097/hc9.0000000000000375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/19/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as NAFLD, is the most common liver disease in children. Liver biopsy remains the gold standard for diagnosis, although more efficient screening methods are needed. We previously developed a novel NAFLD screening panel in youth using machine learning applied to high-resolution metabolomics and clinical phenotype data. Our objective was to validate this panel in a separate cohort, which consisted of a combined cross-sectional sample of 161 children with stored frozen samples (75% male, 12.8±2.6 years of age, body mass index 31.0±7.0 kg/m2, 81% with MASLD, 58% Hispanic race/ethnicity). METHODS Clinical data were collected from all children, and high-resolution metabolomics was performed using their fasting serum samples. MASLD was assessed by MRI-proton density fat fraction or liver biopsy and cardiometabolic factors. Our previously developed panel included waist circumference, triglycerides, whole-body insulin sensitivity index, 3 amino acids, 2 phospholipids, dihydrothymine, and 2 unknowns. To improve feasibility, a simplified version without the unknowns was utilized in the present study. Since the panel was modified, the data were split into training (67%) and test (33%) sets to assess the validity of the panel. RESULTS Our present highest-performing modified model, with 4 clinical variables and 8 metabolomics features, achieved an AUROC of 0.92, 95% sensitivity, and 80% specificity for detecting MASLD in the test set. CONCLUSIONS Therefore, this panel has promising potential for use as a screening tool for MASLD in youth.
Collapse
Affiliation(s)
- Helaina E. Huneault
- Nutrition & Health Sciences Program, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Alasdair E. Gent
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Catherine C. Cohen
- Section of Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Zhulin He
- Department of Pediatrics, Pediatric Biostatistics Core, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Zachery R. Jarrell
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, Georgia, USA
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Miriam B. Vos
- Nutrition & Health Sciences Program, Laney Graduate School, Emory University, Atlanta, Georgia, USA
- Department of Pediatrics, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Yang Y, Ye Y, Deng Y, Gao L. Uridine and its role in metabolic diseases, tumors, and neurodegenerative diseases. Front Physiol 2024; 15:1360891. [PMID: 38487261 PMCID: PMC10937367 DOI: 10.3389/fphys.2024.1360891] [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: 01/04/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
Uridine is a pyrimidine nucleoside found in plasma and cerebrospinal fluid with a concentration higher than the other nucleosides. As a simple metabolite, uridine plays a pivotal role in various biological processes. In addition to nucleic acid synthesis, uridine is critical to glycogen synthesis through the formation of uridine diphosphate glucose in which promotes the production of UDP-GlcNAc in the hexosamine biosynthetic pathway and supplies UDP-GlcNAc for O-GlcNAcylation. This process can regulate protein modification and affect its function. Moreover, Uridine has an effect on body temperature and circadian rhythms, which can regulate the metabolic rate and the expression of metabolic genes. Abnormal levels of blood uridine have been found in people with diabetes and obesity, suggesting a link of uridine dysregulation and metabolic disorders. At present, the role of uridine in glucose metabolism and lipid metabolism is controversial, and the mechanism is not clear, but it shows the trend of long-term damage and short-term benefit. Therefore, maintaining uridine homeostasis is essential for maintaining basic functions and normal metabolism. This article summarizes the latest findings about the metabolic effects of uridine and the potential of uridine metabolism as therapeutic target in treatment of metabolic disorders.
Collapse
Affiliation(s)
- Yueyuan Yang
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yahong Ye
- Department of Internal Medicine, QuanZhou Women’s and Children’s Hospital, QuanZhou, China
| | - Yingfeng Deng
- Department of Diabetes and Cancer Metabolism, City of Hope, Duarte, CA, United States
| | - Ling Gao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Zheng A, Wei C, Liu J, Bu N, Liu D. Deciphering the Mechanism by Which Carbon Dioxide Extends the Shelf Life of Raw Milk: A Microbiomics- and Metabolomics-Based Approach. Molecules 2024; 29:329. [PMID: 38257241 PMCID: PMC10819274 DOI: 10.3390/molecules29020329] [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: 12/01/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Microbial community succession in raw milk determines its quality and storage period. In this study, carbon dioxide (CO2) at 2000 ppm was used to treat raw milk to investigate the mechanism of extending the shelf life of raw milk by CO2 treatment from the viewpoint of microbial colonies and metabolites. The results showed that the shelf life of CO2-treated raw milk was extended to 16 days at 4 °C, while that of the control raw milk was only 6 days. Microbiomics analysis identified 221 amplicon sequence variants (ASVs) in raw milk, and the alpha diversity of microbial communities increased (p < 0.05) with the extension of storage time. Among them, Pseudomonas, Actinobacteria and Serratia were the major microbial genera responsible for the deterioration of raw milk, with a percentage of 85.7%. A combined metagenomics and metabolomics analysis revealed that microorganisms altered the levels of metabolites, such as pyruvic acid, glutamic acid, 5'-cmp, arginine, 2-propenoic acid and phenylalanine, in the raw milk through metabolic activities, such as ABC transporters, pyrimidine metabolism, arginine and proline metabolism and phenylalanine metabolism, and reduced the shelf life of raw milk. CO2 treatment prolonged the shelf life of raw milk by inhibiting the growth of Gram-negative aerobic bacteria, such as Acinetobacter guillouiae, Pseudomonas fluorescens, Serratia liquefaciens and Pseudomonas simiae.
Collapse
Affiliation(s)
- Anran Zheng
- School of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (A.Z.)
- School of Food Science and Engineering, Ningxia University, Yinchuan 750021, China
| | - Chaokun Wei
- School of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (A.Z.)
- School of Food Science and Engineering, Ningxia University, Yinchuan 750021, China
| | - Jun Liu
- School of Life Sciences, Hubei Normmal University, Huangshi 435002, China;
| | - Ningxia Bu
- School of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (A.Z.)
- School of Food Science and Engineering, Ningxia University, Yinchuan 750021, China
| | - Dunhua Liu
- School of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; (A.Z.)
- School of Food Science and Engineering, Ningxia University, Yinchuan 750021, China
| |
Collapse
|
15
|
Meng HH, Liu WY, Zhao WL, Zheng Q, Wang JS. Study on the acute toxicity of trichlorfon and its breakdown product dichlorvos to goldfish (Carassius auratus) based on 1H NMR metabonomics. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:125664-125676. [PMID: 38001290 DOI: 10.1007/s11356-023-31012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2023] [Indexed: 11/26/2023]
Abstract
Trichlorfon, one of the most widely used organophosphate insecticides, is commonly employed in aquaculture and agriculture to combat parasitic infestations. However, its inherent instability leads to rapid decomposition into dichlorvos (DDVP), increasing its toxicity by eightfold. Therefore, the environmental effects of trichlorfon in real-world scenarios involve the combined effects of trichlorfon and its degradation product, DDVP. In this study, we systematically investigated the degradation of trichlorfon in tap water over time using HPLC and LC-MS/MS analysis. Subsequently, an experiment was conducted to assess the acute toxicity of trichlorfon and DDVP on goldfish (Carassius auratus), employing a 1H NMR-based metabolic approach in conjunction with serum biochemistry, histopathological inspection, and correlation network analysis. Exposure to trichlorfon and its degradation product DDVP leads to increased lipid peroxidation, reduced antioxidant activity, and severe hepatotoxicity and nephrotoxicity in goldfish. Based on the observed pathological changes and metabolite alterations, short-term exposure to trichlorfon significantly affected the liver and kidney functions of goldfish, while exerting minimal influence on the brain, potentially due to the presence of the blood-brain barrier. The changes in the metabolic profile indicated that trichlorfon and DDVP influenced several pathways, including oxidative stress, protein synthesis, energy metabolism, and nucleic acid metabolism. This study demonstrated the applicability and potential of 1H NMR-based metabonomics in pesticide environmental risk assessment, providing a feasible method for the comprehensive study of pesticide toxicity in water environments.
Collapse
Affiliation(s)
- Hui-Hui Meng
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Wen-Ya Liu
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Wen-Long Zhao
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Qi Zheng
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Jun-Song Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China.
| |
Collapse
|
16
|
Pascut D, Giraudi PJ, Banfi C, Ghilardi S, Tiribelli C, Bondesan A, Caroli D, Minocci A, Grugni G, Sartorio A. Proteome profiling identifies circulating biomarkers associated with hepatic steatosis in subjects with Prader-Willi syndrome. Front Endocrinol (Lausanne) 2023; 14:1254778. [PMID: 38034016 PMCID: PMC10684934 DOI: 10.3389/fendo.2023.1254778] [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: 07/07/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Prader-Willi syndrome (PWS) is a rare genetic disorder characterized by loss of expression of paternal chromosome 15q11.2-q13 genes. Individuals with PWS exhibit unique physical, endocrine, and metabolic traits associated with severe obesity. Identifying liver steatosis in PWS is challenging, despite its lower prevalence compared to non-syndromic obesity. Reliable biomarkers are crucial for the early detection and management of this condition associated with the complex metabolic profile and cardiovascular risks in PWS. Methods Circulating proteome profiling was conducted in 29 individuals with PWS (15 with steatosis, 14 without) using the Olink Target 96 metabolism and cardiometabolic panels. Correlation analysis was performed to identify the association between protein biomarkes and clinical variables, while the gene enrichment analysis was conducted to identify pathways linked to deregulated proteins. Receiver operating characteristic (ROC) curves assessed the discriminatory power of circulating protein while a logistic regression model evaluated the potential of a combination of protein biomarkers. Results CDH2, CTSO, QDPR, CANT1, ALDH1A1, TYMP, ADGRE, KYAT1, MCFD, SEMA3F, THOP1, TXND5, SSC4D, FBP1, and CES1 exhibited a significant differential expression in liver steatosis, with a progressive increase from grade 1 to grade 3. FBP1, CES1, and QDPR showed predominant liver expression. The logistic regression model, -34.19 + 0.85 * QDPR*QDPR + 0.75 * CANT1*TYMP - 0.46 * THOP1*ALDH1A, achieved an AUC of 0.93 (95% CI: 0.63-0.99), with a sensitivity of 93% and specificity of 80% for detecting steatosis in individuals with PWS. These biomarkers showed strong correlations among themselves and were involved in an interconnected network of 62 nodes, related to seven metabolic pathways. They were also significantly associated with cholesterol, LDL, triglycerides, transaminases, HbA1c, FLI, APRI, and HOMA, and showed a negative correlation with HDL levels. Conclusion The biomarkers identified in this study offer the potential for improved patient stratification and personalized therapeutic protocols.
Collapse
Affiliation(s)
- Devis Pascut
- Liver Cancer Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
| | - Pablo J. Giraudi
- Metabolic Liver Disease Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics, and Network analysis, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Stefania Ghilardi
- Unit of Functional Proteomics, Metabolomics, and Network analysis, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Claudio Tiribelli
- Liver Cancer Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
- Metabolic Liver Disease Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
| | - Adele Bondesan
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
| | - Diana Caroli
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
| | - Alessandro Minocci
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Division of Metabolic Diseases, Piancavallo-Verbania, Italy
| | - Graziano Grugni
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Division of Auxology, Piancavallo-Verbania, Italy
| | - Alessandro Sartorio
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
| |
Collapse
|
17
|
Huang T, Wu Y, Huang L, Lin R, Li Z, Wang X, Wu P, Huang L. Mechanism of the Effect of Compound Anoectochilus roxburghii (Wall.) Lindl. Oral Liquid in Treating Alcoholic Rat Liver Injury by Metabolomics. Drug Des Devel Ther 2023; 17:3409-3428. [PMID: 38024538 PMCID: PMC10659148 DOI: 10.2147/dddt.s427837] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Compound Anoectochilus roxburghii (Wall.) Lindl oral liquid (CAROL) is often as a hepatoprotective agent. The present study aimed to elucidate the protective mechanism of CAROL against alcoholic liver injury in rats by untargeted metabolomics combined with multivariate statistical analysis. Methods An alcoholic liver disease model was established in sprague-dawley (SD) rats by gavage of alcohol, and CAROL treatment was administered. The hepatoprotective effect of CAROL was evaluated by examining liver tissues changes and detecting biochemical index activities and cytokines in serum and liver homogenates. The metabolites in serum samples were examined using ultrahigh-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC-QTOF/MS) and multivariate statistical analysis to screen for differentially expressed metabolites and Kyoto Encyclopedia of Genes and Genomes (KEGG) to assess potential metabolic pathways. Results CAROL has the potential to downregulate inflammation levels and alleviate oxidative stress. The differential metabolites are mainly engaged in riboflavin metabolism, arginine and proline metabolism, phenylalanine, tyrosine and tryptophan biosynthesis metabolism, phenylalanine metabolism, pyrimidine metabolism, and vitamin B6 metabolism to achieve hepatoprotective effects. Conclusion CAROL may exhibit beneficial hepatoprotective effects by reducing inflammation, mitigating oxidative stress, and modulating metabolites and their metabolic pathways.This study has important implications for advancing the clinical application of CAROL.
Collapse
Affiliation(s)
- Tingxuan Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Youjia Wu
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Lingyi Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Renyi Lin
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Zhenyue Li
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Xiaoxiao Wang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Pingping Wu
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Liying Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| |
Collapse
|
18
|
Dubinin MV, Belosludtseva NV, Mikheeva IB, Chelyadnikova YA, Penkina DK, Vafina AB, Starinets VS, Kireev II, Belosludtsev KN. Uridine Administration Promotes Normalization of Heart Mitochondrial Function in Dystrophin-Deficient Mice and Decreases Tissue Fibrosis. Bull Exp Biol Med 2023; 176:54-59. [PMID: 38091140 DOI: 10.1007/s10517-023-05966-0] [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: 03/22/2023] [Indexed: 12/19/2023]
Abstract
The work shows the effect of the metabolic modulator uridine on the functioning and ultrastructure of heart mitochondria in dystrophin-deficient mdx mice. Intraperitoneal administration of uridine (30 mg/kg/day for 28 days) improved K+ transport and increased its content in the heart mitochondria of mdx mice to the level of wild-type animals. This was accompanied by a significant decrease in the level of malondialdehyde and an increase in the number of mitochondria in the heart of mdx mice. At the same time, uridine did not affect the hyperfunctionality of mitochondria in mdx mice, which manifested in an increase in the calcium retention capacity. Nevertheless, we noted that uridine causes a significant decrease in the level of fibrosis in the heart of mdx mice, which attested to a positive effect of therapy.
Collapse
Affiliation(s)
- M V Dubinin
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia.
| | - N V Belosludtseva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | - I B Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | | | - D K Penkina
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia
| | - A B Vafina
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia
| | - V S Starinets
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| | - I I Kireev
- A. N. Belozersky Institute of Physico-Chemical Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - K N Belosludtsev
- Mari State University, Yoshkar-Ola, Mari El Republic, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia
| |
Collapse
|
19
|
Xu F, Chen R, Zhang C, Wang H, Ding Z, Yu L, Tian F, Chen W, Zhou Y, Zhai Q. Cholecystectomy Significantly Alters Gut Microbiota Homeostasis and Metabolic Profiles: A Cross-Sectional Study. Nutrients 2023; 15:4399. [PMID: 37892474 PMCID: PMC10609985 DOI: 10.3390/nu15204399] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Cholecystectomy (CCE) is a standard clinical treatment for conditions like gallstones and cholecystitis. However, its link to post-CCE syndrome, colorectal cancer, and nonalcoholic fatty liver disease has raised concerns. Additionally, studies have demonstrated the disruptive effects of CCE on gut microbiota homeostasis and bile acid (BA) metabolism. Considering the role of gut microbiota in regulating host metabolic and immune pathways, the use of dietary and probiotic intervention strategies to maintain a stable gut ecosystem after CCE could potentially reduce associated disease risks. Inter-study variations have made it challenging to identify consistent gut microbiota patterns after CCE, a prerequisite for targeted interventions. In this study, we first meta-analyzed 218 raw 16S rRNA gene sequencing datasets to determine consistent patterns of structural and functional changes in the gut microbiota after CCE. Our results revealed significant alterations in the gut microbiota's structure and function due to CCE. Furthermore, we identified characteristic gut microbiota changes associated with CCE by constructing a random model classifier. In the validation cohort, this classifier achieved an area under the receiver operating characteristic curve (AUC) of 0.713 and 0.683 when distinguishing between the microbiota of the CCE and healthy groups at the family and genus levels, respectively. Further, fecal metabolomics analysis demonstrated that CCE also substantially modified the metabolic profile, including decreased fecal short-chain fatty acid levels and disrupted BA metabolism. Importantly, dietary patterns, particularly excessive fat and total energy intake, influenced gut microbiota and metabolic profile changes post-CCE. These dietary habits were associated with further enrichment of the microbiota related to BA metabolism and increased levels of intestinal inflammation after CCE. In conclusion, our study identified specific alterations in gut microbiota homeostasis and metabolic profiles associated with CCE. It also revealed a potential link between dietary patterns and gut microbiota changes following CCE. Our study provides a theoretical basis for modulating gut microbiota homeostasis after CCE using long-term dietary strategies and probiotic interventions.
Collapse
Affiliation(s)
- Fusheng Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (F.X.); (R.C.); (C.Z.); (L.Y.); (F.T.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ruimin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (F.X.); (R.C.); (C.Z.); (L.Y.); (F.T.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (F.X.); (R.C.); (C.Z.); (L.Y.); (F.T.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Wang
- Department of Hepatobiliary, Wuxi No. 2 People’s Hospital, Jiangnan University Medical Center, Wuxi 214002, China; (H.W.); (Z.D.)
| | - Zhijie Ding
- Department of Hepatobiliary, Wuxi No. 2 People’s Hospital, Jiangnan University Medical Center, Wuxi 214002, China; (H.W.); (Z.D.)
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (F.X.); (R.C.); (C.Z.); (L.Y.); (F.T.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (F.X.); (R.C.); (C.Z.); (L.Y.); (F.T.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (F.X.); (R.C.); (C.Z.); (L.Y.); (F.T.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Yongping Zhou
- Department of Hepatobiliary, Wuxi No. 2 People’s Hospital, Jiangnan University Medical Center, Wuxi 214002, China; (H.W.); (Z.D.)
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (F.X.); (R.C.); (C.Z.); (L.Y.); (F.T.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
20
|
Rehou S, de Brito Monteiro L, Auger C, Knuth CM, Abdullahi A, Stanojcic M, Jeschke MG. Propranolol Normalizes Metabolomic Signatures Thereby Improving Outcomes After Burn. Ann Surg 2023; 278:519-529. [PMID: 37389480 DOI: 10.1097/sla.0000000000005973] [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: 07/01/2023]
Abstract
OBJECTIVE AND BACKGROUND Propranolol, a nonselective beta-receptor blocker, improves outcomes of severely burned patients. While the clinical and physiological benefits of beta-blockade are well characterized, the underlying metabolic mechanisms are less well defined. We hypothesized that propranolol improves outcomes after burn injury by profoundly modulating metabolic pathways. METHODS In this phase II randomized controlled trial, patients with burns ≥20% of total body surface area were randomly assigned to control or propranolol (dose given to decrease heart rate <100 bpm). Outcomes included clinical markers, inflammatory and lipidomic profiles, untargeted metabolomics, and molecular pathways. RESULTS Fifty-two severely burned patients were enrolled in this trial (propranolol, n=23 and controls, n=29). There were no significant differences in demographics or injury severity between groups. Metabolomic pathway analyses of the adipose tissue showed that propranolol substantially alters several essential metabolic pathways involved in energy and nucleotide metabolism, as well as catecholamine degradation ( P <0.05). Lipidomic analysis revealed that propranolol-treated patients had lower levels of proinflammatory palmitic acid ( P <0.05) and saturated fatty acids ( P <0.05) with an increased ratio of polyunsaturated fatty acids ( P <0.05), thus shifting the lipidomic profile towards an anti-inflammatory phenotype after burn ( P <0.05). These metabolic effects were mediated by decreased activation of hormone-sensitive lipase at serine 660 ( P <0.05) and significantly reduced endoplasmic reticulum stress by decreasing phospho-JNK ( P <0.05). CONCLUSION Propranolol's ability to mitigate pathophysiological changes to essential metabolic pathways results in significantly improved stress responses.
Collapse
Affiliation(s)
- Sarah Rehou
- Hamilton Health Sciences, Hamilton, ON, Canada
- School of Medicine, St. George's University, True Blue, Grenada
| | | | - Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Carly M Knuth
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Abdikarim Abdullahi
- Sunnybrook Research Institute, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mile Stanojcic
- Sunnybrook Research Institute, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Marc G Jeschke
- Hamilton Health Sciences, Hamilton, ON, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
He K, Liu M, Wang Q, Chen S, Guo X. Combined analysis of 16S rDNA sequencing and metabolomics to find biomarkers of drug-induced liver injury. Sci Rep 2023; 13:15138. [PMID: 37704684 PMCID: PMC10499917 DOI: 10.1038/s41598-023-42312-w] [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: 04/05/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
Drug induced liver injury (DILI) is a kind of liver dysfunction which caused by drugs, and gut microbiota could affect liver injury. However, the relationship between gut microbiota and its metabolites in DILI patients is not clear. The total gut microbiota DNA was extracted from 28 DILI patient and 28 healthy control volunteers (HC) and 16S rDNA gene were amplified. Next, differentially metabolites were screened. Finally, the correlations between the diagnostic strains and differentially metabolites were studied.The richness and uniformity of the bacterial communities decreased in DILI patients, and the structure of gut microbiota changed obviously. Enterococcus and Veillonella which belong to Firmicutes increased in DILI, and Blautia and Ralstonia which belong to Firmicutes, Dialister which belongs to Proteobacteria increased in HC. In addition, these diagnostic OTUs of DILI were associated with the DILI damage mechanism. On the other hands, there were 66 differentially metabolites between DILI and HC samples, and these metabolites were mainly enriched in pyrimidine metabolism and steroid hormone biosynthesis pathways. Furthermore, the collinear network map of the key microbiota-metabolites were constructed and the results indicated that Cortodoxone, Prostaglandin I1, Bioyclo Prostaglandin E2 and Anacardic acid were positively correlated with Blautia and Ralstonia, and negatively correlated with Veillonella.This study analyzed the changes of DILI from the perspective of gut microbiota and metabolites. Key strains and differentially metabolites of DILI were screened and the correlations between them were studied. This study further illustrated the mechanism of DILI.
Collapse
Affiliation(s)
- Kaini He
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China
| | - Mimi Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China
| | - Qian Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China
| | - Sijie Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China
| | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China.
| |
Collapse
|
22
|
Zhou NN, Wang T, Lin YX, Xu R, Wu HX, Ding FF, Qiao F, Du ZY, Zhang ML. Uridine alleviates high-carbohydrate diet-induced metabolic syndromes by activating sirt1/AMPK signaling pathway and promoting glycogen synthesis in Nile tilapia ( Oreochromis niloticus). ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 14:56-66. [PMID: 37252330 PMCID: PMC10208930 DOI: 10.1016/j.aninu.2023.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/06/2023] [Accepted: 03/21/2023] [Indexed: 05/31/2023]
Abstract
Carbohydrates have a protein sparing effect, but long-term feeding of a high-carbohydrate diet (HCD) leads to metabolic disorders due to the limited utilization efficiency of carbohydrates in fish. How to mitigate the negative effects induced by HCD is crucial for the rapid development of aquaculture. Uridine is a pyrimidine nucleoside that plays a vital role in regulating lipid and glucose metabolism, but whether uridine can alleviate metabolic syndromes induced by HCD remains unknown. In this study, a total of 480 Nile tilapia (Oreochromis niloticus) (average initial weight 5.02 ± 0.03 g) were fed with 4 diets, including a control diet (CON), HCD, HCD + 500 mg/kg uridine (HCUL) and HCD + 5,000 mg/kg uridine (HCUH), for 8 weeks. The results showed that addition of uridine decreased hepatic lipid, serum glucose, triglyceride and cholesterol (P < 0.05). Further analysis indicated that higher concentration of uridine activated the sirtuin1 (sirt1)/adenosine 5-monophosphate-activated protein kinase (AMPK) signaling pathway to increase lipid catabolism and glycolysis while decreasing lipogenesis (P < 0.05). Besides, uridine increased the activity of glycogen synthesis-related enzymes (P < 0.05). This study suggested that uridine could alleviate HCD-induced metabolic syndrome by activating the sirt1/AMPK signaling pathway and promoting glycogen synthesis. This finding reveals the function of uridine in fish metabolism and facilitates the development of new additives in aquatic feeds.
Collapse
|
23
|
Gao S, Sun Y, Chen X, Zhu C, Liu X, Wang W, Gan L, Lu Y, Schaarschmidt F, Herde M, Witte CP, Chen M. Pyrimidine catabolism is required to prevent the accumulation of 5-methyluridine in RNA. Nucleic Acids Res 2023; 51:7451-7464. [PMID: 37334828 PMCID: PMC10415118 DOI: 10.1093/nar/gkad529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023] Open
Abstract
5-Methylated cytosine is a frequent modification in eukaryotic RNA and DNA influencing mRNA stability and gene expression. Here we show that free 5-methylcytidine (5mC) and 5-methyl-2'-deoxycytidine are generated from nucleic acid turnover in Arabidopsis thaliana, and elucidate how these cytidines are degraded, which is unclear in eukaryotes. First CYTIDINE DEAMINASE produces 5-methyluridine (5mU) and thymidine which are subsequently hydrolyzed by NUCLEOSIDE HYDROLASE 1 (NSH1) to thymine and ribose or deoxyribose. Interestingly, far more thymine is generated from RNA than from DNA turnover, and most 5mU is directly released from RNA without a 5mC intermediate, since 5-methylated uridine (m5U) is an abundant RNA modification (m5U/U ∼1%) in Arabidopsis. We show that m5U is introduced mainly by tRNA-SPECIFIC METHYLTRANSFERASE 2A and 2B. Genetic disruption of 5mU degradation in the NSH1 mutant causes m5U to occur in mRNA and results in reduced seedling growth, which is aggravated by external 5mU supplementation, also leading to more m5U in all RNA species. Given the similarities between pyrimidine catabolism in plants, mammals and other eukaryotes, we hypothesize that the removal of 5mU is an important function of pyrimidine degradation in many organisms, which in plants serves to protect RNA from stochastic m5U modification.
Collapse
Affiliation(s)
- Shangyu Gao
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Sun
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoguang Chen
- Department of Molecular Nutrition and Biochemistry of Plants, Institute of Plant Nutrition, Leibniz University Hannover, Herrenhäuser Str. 2, D-30419 Hannover, Germany
| | - Changhua Zhu
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoye Liu
- Department of Criminal Science and Technology, Nanjing Forest Police College, Nanjing 210023, China
| | - Wenlei Wang
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Lijun Gan
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Yanwu Lu
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Frank Schaarschmidt
- Department of Biostatistics, Institute of Cell Biology and Biophysics, Leibniz University Hannover, Herrenhäuser Str. 2, D-30419 Hannover, Germany
| | - Marco Herde
- Department of Molecular Nutrition and Biochemistry of Plants, Institute of Plant Nutrition, Leibniz University Hannover, Herrenhäuser Str. 2, D-30419 Hannover, Germany
| | - Claus-Peter Witte
- Department of Molecular Nutrition and Biochemistry of Plants, Institute of Plant Nutrition, Leibniz University Hannover, Herrenhäuser Str. 2, D-30419 Hannover, Germany
| | - Mingjia Chen
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
24
|
Zi L, Ma W, Zhang L, Qiao B, Qiu Z, Xu J, Zhang J, Ye Y, Yang Y, Dong K, Chen C, Wang W, Zhao Q. Uridine Inhibits Hepatocellular Carcinoma Cell Development by Inducing Ferroptosis. J Clin Med 2023; 12:jcm12103552. [PMID: 37240659 DOI: 10.3390/jcm12103552] [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: 01/27/2023] [Revised: 03/24/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Uridine is a key metabolite used as a substrate for the production of DNA, RNA, and glucose, and it is mainly synthesized in the liver. Currently, it is not known whether uridine levels are altered in the tumor microenvironment of patients with hepatocellular carcinoma (HCC) and whether uridine can be a target for tumor therapy. In this study, the detection of genes associated with de novo uridine synthesis, carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, dihydroorotase (CAD) (n = 115), and dihydroorotate dehydrogenase (DHODH) (n = 115) in HCC tissues through tissue microarrays revealed that the expression of CAD and DHODH was higher in tumor compared with paraneoplastic tissues. Next, we collected tumor tissues from surgically resected HCC patients and the corresponding adjacent non-tumor tissues (n = 46) for LC-MS/MS assays. The results showed that the median and interquartile ranges of uridine content in non-tumor and tumor tissues were 640.36 (504.45-807.43) and 484.22 (311.91-626.73) nmol/g, respectively. These results suggest that uridine metabolism is disturbed in HCC patients. To further investigate whether uridine can be used as a tumor-therapeutic target, a series of high concentrations of uridine were incubated with HCC cells in vitro and in vivo. It was observed that uridine dose-dependently inhibited the proliferation, invasion, and migration of HCC cells by activating the ferroptosis pathway. Overall, these results reveal for the first time the range of uridine content in human HCC tissues and suggest that uridine may be a new target for HCC therapy.
Collapse
Affiliation(s)
- Liuliu Zi
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wangbin Ma
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Lilong Zhang
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Boyang Qiao
- School of Pharmaceutical Sciences, Wuhan University, Wuhan 430072, China
| | - Zhendong Qiu
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Junhui Xu
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Jiacheng Zhang
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Yahong Ye
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yueyuan Yang
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Keshuai Dong
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Chen Chen
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Weixing Wang
- Department of Hepatobiliary and Laparoscopic Surgery, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuchang District, Wuhan 430060, China
| | - Qingyan Zhao
- Hubei Key Laboratory of Cardiology, Cardiovascular Research Institute of Wuhan University, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
25
|
Wang Z, Zhang Y, Zhou T, Wu X. N-carbamoyl aspartate reduced body weight by stimulating the thermogenesis of iBAT. Biochem Biophys Res Commun 2023; 665:152-158. [PMID: 37163935 DOI: 10.1016/j.bbrc.2023.04.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Uridine has formerly been shown to alleviate obesity and hepatic lipid accumulation. N-carbamoyl aspartate (NCA) provides carbon atoms to uridine in de novo pyrimidine biosynthesis pathway. However, whether NCA is involved in the lipid metabolism remains elusive. Here we showed that NCA supplementation significantly decreased (P < 0.05) serum cholesterol (CHOL), high-density lipoprotein (HDL), lactate dehydrogenase (LDH), and alkaline phosphatase (ALP) levels of mice, and significantly increased (P < 0.05) relative mRNA expression of genes related to the synthesis of pyrimidine nucleotides and polyunsaturated fatty acids. Besides, supplemented with NCA significantly decreased body weight and area under the curve (AUC), and increased body temperature in the high-fat diet fed mice. For further, relative protein expression of uridine monophosphate synthase (UMPS), sterol regulatory element-binding protein 1(SREBP-1) and phosphorylated hormone-sensitive triglyceride lipase (P-HSL) in the liver, and uncoupling protein 1 (UCP-1) in interscapular brown adipose tissue (iBAT) also showed upregulated in the high-fat diet fed mice. Thus, NCA promoted de novo synthesis of pyrimidine and polyunsaturated fatty acid, and reduced body weight by stimulating high-fat diet-induced thermogenesis of iBAT.
Collapse
Affiliation(s)
- Zhefeng Wang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, PR China; College of Advanced Agricultural Sciences, University of Chinese Academy of Science, Beijing, 100049, PR China
| | - Yumei Zhang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, PR China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Tiantian Zhou
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, PR China; College of Advanced Agricultural Sciences, University of Chinese Academy of Science, Beijing, 100049, PR China
| | - Xin Wu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, PR China; Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China; College of Advanced Agricultural Sciences, University of Chinese Academy of Science, Beijing, 100049, PR China.
| |
Collapse
|
26
|
Skinner OS, Blanco-Fernández J, Goodman RP, Kawakami A, Shen H, Kemény LV, Joesch-Cohen L, Rees MG, Roth JA, Fisher DE, Mootha VK, Jourdain AA. Salvage of ribose from uridine or RNA supports glycolysis in nutrient-limited conditions. Nat Metab 2023; 5:765-776. [PMID: 37198474 PMCID: PMC10229423 DOI: 10.1038/s42255-023-00774-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/03/2023] [Indexed: 05/19/2023]
Abstract
Glucose is vital for life, serving as both a source of energy and carbon building block for growth. When glucose is limiting, alternative nutrients must be harnessed. To identify mechanisms by which cells can tolerate complete loss of glucose, we performed nutrient-sensitized genome-wide genetic screens and a PRISM growth assay across 482 cancer cell lines. We report that catabolism of uridine from the medium enables the growth of cells in the complete absence of glucose. While previous studies have shown that uridine can be salvaged to support pyrimidine synthesis in the setting of mitochondrial oxidative phosphorylation deficiency1, our work demonstrates that the ribose moiety of uridine or RNA can be salvaged to fulfil energy requirements via a pathway based on: (1) the phosphorylytic cleavage of uridine by uridine phosphorylase UPP1/UPP2 into uracil and ribose-1-phosphate (R1P), (2) the conversion of uridine-derived R1P into fructose-6-P and glyceraldehyde-3-P by the non-oxidative branch of the pentose phosphate pathway and (3) their glycolytic utilization to fuel ATP production, biosynthesis and gluconeogenesis. Capacity for glycolysis from uridine-derived ribose appears widespread, and we confirm its activity in cancer lineages, primary macrophages and mice in vivo. An interesting property of this pathway is that R1P enters downstream of the initial, highly regulated steps of glucose transport and upper glycolysis. We anticipate that 'uridine bypass' of upper glycolysis could be important in the context of disease and even exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Owen S Skinner
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Russell P Goodman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Akinori Kawakami
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hongying Shen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Yale Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Lajos V Kemény
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | | | | | | | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Vamsi K Mootha
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Alexis A Jourdain
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
27
|
Lai K, Song C, Gao M, Deng Y, Lu Z, Li N, Geng Q. Uridine Alleviates Sepsis-Induced Acute Lung Injury by Inhibiting Ferroptosis of Macrophage. Int J Mol Sci 2023; 24:ijms24065093. [PMID: 36982166 PMCID: PMC10049139 DOI: 10.3390/ijms24065093] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 03/30/2023] Open
Abstract
Uridine metabolism is extensively reported to be involved in combating oxidative stress. Redox-imbalance-mediated ferroptosis plays a pivotal role in sepsis-induced acute lung injury (ALI). This study aims to explore the role of uridine metabolism in sepsis-induced ALI and the regulatory mechanism of uridine in ferroptosis. The Gene Expression Omnibus (GEO) datasets including lung tissues in lipopolysaccharides (LPS) -induced ALI model or human blood sample of sepsis were collected. In vivo and vitro, LPS was injected into mice or administered to THP-1 cells to generate sepsis or inflammatory models. We identified that uridine phosphorylase 1 (UPP1) was upregulated in lung tissues and septic blood samples and uridine significantly alleviated lung injury, inflammation, tissue iron level and lipid peroxidation. Nonetheless, the expression of ferroptosis biomarkers, including SLC7A11, GPX4 and HO-1, were upregulated, while lipid synthesis gene (ACSL4) expression was greatly restricted by uridine supplementation. Moreover, pretreatment of ferroptosis inducer (Erastin or Era) weakened while inhibitor (Ferrostatin-1 or Fer-1) strengthened the protective effects of uridine. Mechanistically, uridine inhibited macrophage ferroptosis by activating Nrf2 signaling pathway. In conclusion, uridine metabolism dysregulation is a novel accelerator for sepsis-induced ALI and uridine supplementation may offer a potential avenue for ameliorating sepsis-induced ALI by suppressing ferroptosis.
Collapse
Affiliation(s)
- Kai Lai
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Congkuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
28
|
Zhang L, Qin Z, Zhang L, Jiang Y, Zhu J. Dynamic changes of quality and flavor characterization of Zhejiang rosy vinegar during fermentation and aging based on untargeted metabolomics. Food Chem 2023; 404:134702. [DOI: 10.1016/j.foodchem.2022.134702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/24/2022] [Accepted: 10/18/2022] [Indexed: 11/22/2022]
|
29
|
Niu K, Bai P, Zhang J, Feng X, Qiu F. Cytidine Alleviates Dyslipidemia and Modulates the Gut Microbiota Composition in ob/ob Mice. Nutrients 2023; 15:nu15051147. [PMID: 36904146 PMCID: PMC10005144 DOI: 10.3390/nu15051147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Cytidine and uridine are endogenous metabolites in the pyrimidine metabolism pathway, and cytidine is a substrate that can be metabolized into uridine via cytidine deaminase. Uridine has been widely reported to be effective in regulating lipid metabolism. However, whether cytidine could ameliorate lipid metabolism disorder has not yet been investigated. In this research, ob/ob mice were used, and the effect of cytidine (0.4 mg/mL in drinking water for five weeks) on lipid metabolism disorder was evaluated in terms of an oral glucose tolerance test, serum lipid levels, liver histopathological analysis and gut microbiome analysis. Uridine was used as a positive control. Our findings reveal that cytidine could alleviate certain aspects of dyslipidemia and improve hepatic steatosis via modulating the gut microbiota composition in ob/ob mice, especially increasing the abundance of short-chain fatty acids-producing microbiota. These results suggest that cytidine supplementation could be a potential therapeutic approach for dyslipidemia.
Collapse
Affiliation(s)
| | | | | | - Xinchi Feng
- Correspondence: (X.F.); (F.Q.); Tel.: +86-22-595-6223 (X.F.)
| | - Feng Qiu
- Correspondence: (X.F.); (F.Q.); Tel.: +86-22-595-6223 (X.F.)
| |
Collapse
|
30
|
Gong M, Lu H, Li L, Feng M, Zou Z. Integration of transcriptomics and metabonomics revealed the protective effects of hemp seed oil against methionine-choline-deficient diet-induced non-alcoholic steatohepatitis in mice. Food Funct 2023; 14:2096-2111. [PMID: 36734470 DOI: 10.1039/d2fo03054c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic liver disease with few therapeutic options available currently. Hemp seed oil extracted from the seeds of hemp (Cannabis sativa L.) has significant nutritional and biological properties due to the unique composition of polyunsaturated fatty acids and various antioxidant compounds. However, little is known about the beneficial effects and molecular mechanisms of hemp seed oil on NASH. Here, the hepatoprotective effects of hemp seed oil on methionine-choline-deficient (MCD) diet-induced NASH in C57BL/6 mice were explored via integration of transcriptomics and metabolomics. Hemp seed oil could improve hepatic steatosis, inflammation and fibrosis in mice with MCD diet-induced NASH. In a nuclear magnetic resonance (NMR)-based metabonomic study, the hepatic and urinary metabolic profiles of mice supplemented with hemp seed oil showed a tendency to recover to healthy controls compared to those of NASH mice. Eight potential biomarkers associated with NASH in both liver tissue and urine were restored to near normal levels by administration of hemp seed oil. The proposed pathways were mainly involved in pyrimidine metabolism, one-carbon metabolism, amino acid metabolism, glycolysis and the tricarboxylic acid (TCA) cycle. Hepatic transcriptomics based on Illumina RNA-Seq sequencing showed that hemp seed oil exerted anti-NASH activities by regulating multiple signaling pathways, e.g., downregulation of the TNF signaling pathway, the IL-17 signaling pathway, the MAPK signaling pathway and the NF-κB signaling pathway, which played a pivotal role in the pathogenesis of NASH. In particular, integration of metabonomic and transcriptomic results suggested that hemp seed oil could attenuate NASH-related liver fibrosis by inhibition of glutaminolysis. These results provided new insights into the hepatoprotective effects of hemp seed oil against MCD diet-induced NASH and hemp seed oil might have potential as an effective therapy for NASH.
Collapse
Affiliation(s)
- Mengjuan Gong
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Hailong Lu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lixi Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Meiqi Feng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhongjie Zou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
31
|
Plasma Metabolite Signatures in Male Carriers of Genetic Variants Associated with Non-Alcoholic Fatty Liver Disease. Metabolites 2023; 13:metabo13020267. [PMID: 36837886 PMCID: PMC9964056 DOI: 10.3390/metabo13020267] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/01/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Both genetic and non-genetic factors are important in the pathophysiology of non-alcoholic fatty liver disease (NAFLD). The aim of our study was to identify novel metabolites and pathways associated with NAFLD by including both genetic and non-genetic factors in statistical analyses. We genotyped six genetic variants in the PNPLA3, TM6SF2, MBOAT7, GCKR, PPP1R3B, and HSD17B13 genes reported to be associated with NAFLD. Non-targeted metabolomic profiling was performed from plasma samples. We applied a previously validated fatty liver index to identify participants with NAFLD. First, we associated the six genetic variants with 1098 metabolites in 2 339 men without NAFLD to determine the effects of the genetic variants on metabolites, and then in 2 535 men with NAFLD to determine the joint effects of genetic variants and non-genetic factors on metabolites. We identified several novel metabolites and metabolic pathways, especially for PNPLA3, GCKR, and PPP1R38 variants relevant to the pathophysiology of NAFLD. Importantly, we showed that each genetic variant for NAFLD had a specific metabolite signature. The plasma metabolite signature was unique for each genetic variant, suggesting that several metabolites and different pathways are involved in the risk of NAFLD. The FLI index reliably identifies metabolites for NAFLD in large population-based studies.
Collapse
|
32
|
Luukkonen PK, Sakuma I, Gaspar RC, Mooring M, Nasiri A, Kahn M, Zhang XM, Zhang D, Sammalkorpi H, Penttilä AK, Orho-Melander M, Arola J, Juuti A, Zhang X, Yimlamai D, Yki-Järvinen H, Petersen KF, Shulman GI. Inhibition of HSD17B13 protects against liver fibrosis by inhibition of pyrimidine catabolism in nonalcoholic steatohepatitis. Proc Natl Acad Sci U S A 2023; 120:e2217543120. [PMID: 36669104 PMCID: PMC9942818 DOI: 10.1073/pnas.2217543120] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/15/2022] [Indexed: 01/21/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, in which prognosis is determined by liver fibrosis. A common variant in hydroxysteroid 17-beta dehydrogenase 13 (HSD17B13, rs72613567-A) is associated with a reduced risk of fibrosis in NAFLD, but the underlying mechanism(s) remains unclear. We investigated the effects of this variant in the human liver and in Hsd17b13 knockdown in mice by using a state-of-the-art metabolomics approach. We demonstrate that protection against liver fibrosis conferred by the HSD17B13 rs72613567-A variant in humans and by the Hsd17b13 knockdown in mice is associated with decreased pyrimidine catabolism at the level of dihydropyrimidine dehydrogenase. Furthermore, we show that hepatic pyrimidines are depleted in two distinct mouse models of NAFLD and that inhibition of pyrimidine catabolism by gimeracil phenocopies the HSD17B13-induced protection against liver fibrosis. Our data suggest pyrimidine catabolism as a therapeutic target against the development of liver fibrosis in NAFLD.
Collapse
Affiliation(s)
- Panu K. Luukkonen
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki00290, Finland
- Minerva Foundation Institute for Medical Research, Helsinki00290, Finland
| | - Ikki Sakuma
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
| | - Rafael C. Gaspar
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
| | - Meghan Mooring
- Department of Pediatrics, The Yale Liver Center, Yale School of Medicine, New Haven06520, CT
| | - Ali Nasiri
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
| | - Mario Kahn
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
| | - Xian-Man Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
| | - Henna Sammalkorpi
- Department of Abdominal Surgery, Helsinki University Hospital and University of Helsinki, Helsinki00290, Finland
| | - Anne K. Penttilä
- Department of Abdominal Surgery, Helsinki University Hospital and University of Helsinki, Helsinki00290, Finland
| | - Marju Orho-Melander
- Department of Clinical Sciences, Diabetes and Endocrinology, University Hospital Malmö, Lund University, Malmö21428, Sweden
| | - Johanna Arola
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki00290, Finland
| | - Anne Juuti
- Department of Abdominal Surgery, Helsinki University Hospital and University of Helsinki, Helsinki00290, Finland
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven06520, CT
| | - Dean Yimlamai
- Department of Pediatrics, The Yale Liver Center, Yale School of Medicine, New Haven06520, CT
| | - Hannele Yki-Järvinen
- Minerva Foundation Institute for Medical Research, Helsinki00290, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki00290, Finland
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
| | - Gerald I. Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven06520, CT
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven06520, CT
| |
Collapse
|
33
|
Liébana-García R, Olivares M, Francés-Cuesta C, Rubio T, Rossini V, Quintas G, Sanz Y. Intestinal group 1 innate lymphoid cells drive macrophage-induced inflammation and endocrine defects in obesity and promote insulinemia. Gut Microbes 2023; 15:2181928. [PMID: 36823075 PMCID: PMC9980552 DOI: 10.1080/19490976.2023.2181928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Hypercaloric diets overactivate the intestinal immune system and disrupt the microbiome and epithelial cell functions, impairing glucose metabolism. The origins of this inflammatory cascade are poorly characterized. We investigated the involvement of intestinal proinflammatory group 1 innate lymphoid cells (ILC1s) in obesity progression and metabolic disruption. In obese mice, we studied longitudinally the ILC1s response to the diet and ILC1s depletion to address its role in obesity. ILC1s are required for the expansion of pro-inflammatory macrophages and ILC2s. ILC1s depletion induced the ILC3-IL-22 pathway, increasing mucin production, antimicrobial peptides, and neuroendocrine cells. These changes were translated into higher gut hormones and reduced insulinemia and adiposity. ILC1s depletion was also associated with a bloom in Akkermansia muciniphila and decreases in Bilophila spp. Intestinal-ILC1s are upstream activators of inflammatory signals, connecting immunity with the microbiome, the enteroendocrine system, and the intestinal barrier in the control of glucose metabolism and adiposity.
Collapse
Affiliation(s)
- Rebeca Liébana-García
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Marta Olivares
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain,CONTACT Marta Olivares Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Carlos Francés-Cuesta
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Teresa Rubio
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Valerio Rossini
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Guillermo Quintas
- Health and Biomedicine, Leitat Technological Center, Terrassa, Spain,Analytical Unit, Health Research Institute La Fe, Valencia, Spain
| | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain,Yolanda Sanz Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
34
|
Gwynne WD, Suk Y, Custers S, Mikolajewicz N, Chan JK, Zador Z, Chafe SC, Zhai K, Escudero L, Zhang C, Zaslaver O, Chokshi C, Shaikh MV, Bakhshinyan D, Burns I, Chaudhry I, Nachmani O, Mobilio D, Maich WT, Mero P, Brown KR, Quaile AT, Venugopal C, Moffat J, Montenegro-Burke JR, Singh SK. Cancer-selective metabolic vulnerabilities in MYC-amplified medulloblastoma. Cancer Cell 2022; 40:1488-1502.e7. [PMID: 36368321 DOI: 10.1016/j.ccell.2022.10.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/09/2022] [Accepted: 10/06/2022] [Indexed: 11/12/2022]
Abstract
MYC-driven medulloblastoma (MB) is an aggressive pediatric brain tumor characterized by therapy resistance and disease recurrence. Here, we integrated data from unbiased genetic screening and metabolomic profiling to identify multiple cancer-selective metabolic vulnerabilities in MYC-driven MB tumor cells, which are amenable to therapeutic targeting. Among these targets, dihydroorotate dehydrogenase (DHODH), an enzyme that catalyzes de novo pyrimidine biosynthesis, emerged as a favorable candidate for therapeutic targeting. Mechanistically, DHODH inhibition acts on target, leading to uridine metabolite scarcity and hyperlipidemia, accompanied by reduced protein O-GlcNAcylation and c-Myc degradation. Pyrimidine starvation evokes a metabolic stress response that leads to cell-cycle arrest and apoptosis. We further show that an orally available small-molecule DHODH inhibitor demonstrates potent mono-therapeutic efficacy against patient-derived MB xenografts in vivo. The reprogramming of pyrimidine metabolism in MYC-driven medulloblastoma represents an unappreciated therapeutic strategy and a potential new class of treatments with stronger cancer selectivity and fewer neurotoxic sequelae.
Collapse
Affiliation(s)
- William D Gwynne
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Yujin Suk
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Michael G DeGroote School of Medicine, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Stefan Custers
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Nicholas Mikolajewicz
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada
| | - Jeremy K Chan
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zsolt Zador
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Shawn C Chafe
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Kui Zhai
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Laura Escudero
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Cunjie Zhang
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Olga Zaslaver
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chirayu Chokshi
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Muhammad Vaseem Shaikh
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - David Bakhshinyan
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Ian Burns
- Michael G DeGroote School of Medicine, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Iqra Chaudhry
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Omri Nachmani
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada
| | - Daniel Mobilio
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - William T Maich
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Patricia Mero
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kevin R Brown
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada
| | - Andrew T Quaile
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chitra Venugopal
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Jason Moffat
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada; Institute for Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - J Rafael Montenegro-Burke
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College St, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Sheila K Singh
- Department of Surgery, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada; Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
35
|
Darekar S, Laín S. Asymmetric inheritance of cytoophidia could contribute to determine cell fate and plasticity: The onset of alternative differentiation patterns in daughter cells may rely on the acquisition of either CTPS or IMPDH cytoophidia: The onset of alternative differentiation patterns in daughter cells may rely on the acquisition of either CTPS or IMPDH cytoophidia. Bioessays 2022; 44:e2200128. [PMID: 36209393 DOI: 10.1002/bies.202200128] [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: 07/01/2022] [Revised: 08/26/2022] [Accepted: 09/21/2022] [Indexed: 12/20/2022]
Abstract
Two enzymes involved in the synthesis of pyrimidine and purine nucleotides, CTP synthase (CTPS) and IMP dehydrogenase (IMPDH), can assemble into a single or very few large filaments called rods and rings (RR) or cytoophidia. Most recently, asymmetric cytoplasmic distribution of organelles during cell division has been described as a decisive event in hematopoietic stem cell fate. We propose that cytoophidia, which could be considered as membrane-less organelles, may also be distributed asymmetrically during mammalian cell division as previously described for Schizosaccharomyces pombe. Furthermore, because each type of nucleotide intervenes in distinct processes (e.g., membrane synthesis, glycosylation, and G protein-signaling), alterations in the rate of synthesis of specific nucleotide types could influence cell differentiation in multiple ways. Therefore, we hypothesize that whether a daughter cell inherits or not CTPS or IMPDH filaments determines its fate and that this asymmetric inheritance, together with the dynamic nature of these structures enables plasticity in a cell population.
Collapse
Affiliation(s)
- Suhas Darekar
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Sonia Laín
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Dubinin MV, Starinets VS, Belosludtseva NV, Mikheeva IB, Chelyadnikova YA, Penkina DK, Vedernikov AA, Belosludtsev KN. The Effect of Uridine on the State of Skeletal Muscles and the Functioning of Mitochondria in Duchenne Dystrophy. Int J Mol Sci 2022; 23:10660. [PMID: 36142572 PMCID: PMC9500747 DOI: 10.3390/ijms231810660] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/06/2022] [Accepted: 09/10/2022] [Indexed: 12/16/2022] Open
Abstract
Duchenne muscular dystrophy is caused by the loss of functional dystrophin that secondarily causes systemic metabolic impairment in skeletal muscles and cardiomyocytes. The nutraceutical approach is considered as a possible complementary therapy for this pathology. In this work, we have studied the effect of pyrimidine nucleoside uridine (30 mg/kg/day for 28 days, i.p.), which plays an important role in cellular metabolism, on the development of DMD in the skeletal muscles of dystrophin deficient mdx mice, as well as its effect on the mitochondrial dysfunction that accompanies this pathology. We found that chronic uridine administration reduced fibrosis in the skeletal muscles of mdx mice, but it had no effect on the intensity of degeneration/regeneration cycles and inflammation, pseudohypetrophy, and muscle strength of the animals. Analysis of TEM micrographs showed that uridine also had no effect on the impaired mitochondrial ultrastructure of mdx mouse skeletal muscle. The administration of uridine was found to lead to an increase in the expression of the Drp1 and Parkin genes, which may indicate an increase in the intensity of organelle fission and the normalization of mitophagy. Uridine had little effect on OXPHOS dysfunction in mdx mouse mitochondria, and moreover, it was suppressed in the mitochondria of wild type animals. At the same time, uridine restored the transport of potassium ions and reduced the production of reactive oxygen species; however, this had no effect on the impaired calcium retention capacity of mdx mouse mitochondria. The obtained results demonstrate that the used dose of uridine only partially prevents mitochondrial dysfunction in skeletal muscles during Duchenne dystrophy, though it mitigates the development of destructive processes in skeletal muscles.
Collapse
Affiliation(s)
- Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Vlada S. Starinets
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Natalia V. Belosludtseva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Irina B. Mikheeva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Yuliya A. Chelyadnikova
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Daria K. Penkina
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Alexander A. Vedernikov
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| |
Collapse
|
37
|
Belosludtseva NV, Starinets VS, Mikheeva IB, Belosludtsev MN, Dubinin MV, Mironova GD, Belosludtsev KN. Effect of Chronic Treatment with Uridine on Cardiac Mitochondrial Dysfunction in the C57BL/6 Mouse Model of High-Fat Diet-Streptozotocin-Induced Diabetes. Int J Mol Sci 2022; 23:10633. [PMID: 36142532 PMCID: PMC9502122 DOI: 10.3390/ijms231810633] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022] Open
Abstract
Long-term hyperglycemia in diabetes mellitus is associated with complex damage to cardiomyocytes and the development of mitochondrial dysfunction in the myocardium. Uridine, a pyrimidine nucleoside, plays an important role in cellular metabolism and is used to improve cardiac function. Herein, the antidiabetic potential of uridine (30 mg/kg/day for 21 days, i.p.) and its effect on mitochondrial homeostasis in the heart tissue were examined in a high-fat diet-streptozotocin-induced model of diabetes in C57BL/6 mice. We found that chronic administration of uridine to diabetic mice normalized plasma glucose and triglyceride levels and the heart weight/body weight ratio and increased the rate of glucose utilization during the intraperitoneal glucose tolerance test. Analysis of TEM revealed that uridine prevented diabetes-induced ultrastructural abnormalities in mitochondria and sarcomeres in ventricular cardiomyocytes. In diabetic heart tissue, the mRNA level of Ppargc1a decreased and Drp1 and Parkin gene expression increased, suggesting the disturbances of mitochondrial biogenesis, fission, and mitophagy, respectively. Uridine treatment of diabetic mice restored the mRNA level of Ppargc1a and enhanced Pink1 gene expression, which may indicate an increase in the intensity of mitochondrial biogenesis and mitophagy, and as a consequence, mitochondrial turnover. Uridine also reduced oxidative phosphorylation dysfunction and suppressed lipid peroxidation, but it had no significant effect on the impaired calcium retention capacity and potassium transport in the heart mitochondria of diabetic mice. Altogether, these findings suggest that, along with its hypoglycemic effect, uridine has a protective action against diabetes-mediated functional and structural damage to cardiac mitochondria and disruption of mitochondrial quality-control systems in the diabetic heart.
Collapse
Affiliation(s)
- Natalia V. Belosludtseva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Vlada S. Starinets
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Irina B. Mikheeva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
| | - Maxim N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| | - Galina D. Mironova
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
| | - Konstantin N. Belosludtsev
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Russia
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, Yoshkar-Ola 424001, Russia
| |
Collapse
|
38
|
Chen W, Yao M, Chen M, Ou Z, Yang Q, He Y, Zhang N, Deng M, Wu Y, Chen R, Tan X, Kong Z. Using an untargeted metabolomics approach to analyze serum metabolites in COVID-19 patients with nucleic acid turning negative. Front Pharmacol 2022; 13:964037. [PMID: 36091834 PMCID: PMC9449332 DOI: 10.3389/fphar.2022.964037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The coronavirus disease of 2019 (COVID-19) is a severe public health issue that has infected millions of people. The effective prevention and control of COVID-19 has resulted in a considerable increase in the number of cured cases. However, little research has been done on a complete metabonomic examination of metabolic alterations in COVID-19 patients following treatment. The current project pursues rigorously to characterize the variation of serum metabolites between healthy controls and COVID-19 patients with nucleic acid turning negative via untargeted metabolomics. Methods: The metabolic difference between 20 COVID-19 patients (CT ≥ 35) and 20 healthy controls were investigated utilizing untargeted metabolomics analysis employing High-resolution UHPLC-MS/MS. COVID-19 patients’ fundamental clinical indicators, as well as health controls, were also collected. Results: Out of the 714 metabolites identified, 203 still significantly differed between COVID-19 patients and healthy controls, including multiple amino acids, fatty acids, and glycerophospholipids. The clinical indexes including monocytes, lymphocytes, albumin concentration, total bilirubin and direct bilirubin have also differed between our two groups of participators. Conclusion: Our results clearly showed that in COVID-19 patients with nucleic acid turning negative, their metabolism was still dysregulated in amino acid metabolism and lipid metabolism, which could be the mechanism of long-COVID and calls for specific post-treatment care to help COVID-19 patients recover.
Collapse
Affiliation(s)
- Wenyu Chen
- Department of Respiration, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Ming Yao
- Department of Anesthesiology and Pain Research Center, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Miaomiao Chen
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
| | - Zhao Ou
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
| | - Qi Yang
- Department of Respiration, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yanbin He
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, China
| | - Ning Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Min Deng
- Department of Infection, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yuqi Wu
- Calibra Lab at DIAN Diagnostics, Hangzhou, China
| | | | - Xiaoli Tan
- Department of Respiration, Affiliated Hospital of Jiaxing University, Jiaxing, China
- *Correspondence: Xiaoli Tan, ; Ziqing Kong,
| | - Ziqing Kong
- Calibra Lab at DIAN Diagnostics, Hangzhou, China
- *Correspondence: Xiaoli Tan, ; Ziqing Kong,
| |
Collapse
|
39
|
Sullivan KE, Kumar S, Liu X, Zhang Y, de Koning E, Li Y, Yuan J, Fan F. Uncovering the roles of dihydropyrimidine dehydrogenase in fatty-acid induced steatosis using human cellular models. Sci Rep 2022; 12:14109. [PMID: 35982095 PMCID: PMC9388600 DOI: 10.1038/s41598-022-17860-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 08/02/2022] [Indexed: 12/03/2022] Open
Abstract
Pyrimidine catabolism is implicated in hepatic steatosis. Dihydropyrimidine dehydrogenase (DPYD) is an enzyme responsible for uracil and thymine catabolism, and DPYD human genetic variability affects clinically observed toxicity following 5-Fluorouracil administration. In an in vitro model of fatty acid-induced steatosis, the pharmacologic inhibition of DPYD resulted in protection from lipid accumulation. Additionally, a gain-of-function mutation of DPYD, created through clustered regularly interspaced short palindromic repeats associated protein 9 (CRISPR-Cas9) engineering, led to an increased lipid burden, which was associated with altered mitochondrial functionality in a hepatocarcionma cell line. The studies presented herein describe a novel role for DPYD in hepatocyte metabolic regulation as a modulator of hepatic steatosis.
Collapse
Affiliation(s)
- Kelly E Sullivan
- Translational Systems Biology Group, Amgen Inc., Cambridge, MA, 02141, USA.,Vertex Pharmaceuticals, Boston, MA, 02210, USA
| | - Sheetal Kumar
- Translational Systems Biology Group, Amgen Inc., Cambridge, MA, 02141, USA.,Nimbus Therapeutics, Cambridge, MA, 02139, USA
| | - Xin Liu
- Translational Systems Biology Group, Amgen Inc., Cambridge, MA, 02141, USA.,Novartis Institutes for Biomedical Research, Cambridge, MA, 02139, USA
| | - Ye Zhang
- Translational Systems Biology Group, Amgen Inc., Cambridge, MA, 02141, USA.,Novartis Institutes for Biomedical Research, Cambridge, MA, 02139, USA
| | - Emily de Koning
- Translational Systems Biology Group, Amgen Inc., Cambridge, MA, 02141, USA.,Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Yanfei Li
- Amgen Inc., South San Francisco, CA, 90408, USA
| | - Jing Yuan
- Translational Systems Biology Group, Amgen Inc., Cambridge, MA, 02141, USA.,Pfizer Inc., Cambridge, MA, 02139, USA
| | - Fan Fan
- Translational Systems Biology Group, Amgen Inc., Cambridge, MA, 02141, USA. .,Janssen Pharmaceutical Companies of Johnson & Johnson, La Jolla, CA, 92037, USA.
| |
Collapse
|
40
|
Gu X, Li S, Lu M, Li Y, Wang Q, Chen L, Jia Y, Cao S, Zhang T, Zhou M, Gou X. Investigation of Gynura segetum root extract (GSrE) induced hepatotoxicity based on metabolomic signatures and microbial community profiling in rats. Front Microbiol 2022; 13:947757. [PMID: 36016788 PMCID: PMC9396145 DOI: 10.3389/fmicb.2022.947757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/14/2022] [Indexed: 11/26/2022] Open
Abstract
In recent years, many reports focus on the hepatotoxicity of Gynura segetum root extract (GSrE), but the interaction between GSrE and the gut microbiota is still unclear. This study investigated the mechanism of GSrE-induced hepatotoxicity of different doses and exposure durations by combining metabolomics and gut microbiota analysis. SD rats were divided into 3 groups: blank, low-dose (7.5 g/kg), and high-dose (15 g/kg) groups. Urine and feces samples were collected on day 0, day 10, and day 21. Metabolomics based on gas chromatography-mass spectrometry (GC-MS) was carried out to identify metabolites and metabolic pathways. 16S rDNA gene sequencing was applied to investigate the composition of gut microbiota before and after GSrE-induced hepatotoxicity. Finally, a correlation analysis of metabolites and gut microbiota was performed. Differential metabolites in urine and feces involved amino acids, carbohydrates, lipids, organic acids, and short chain fatty acids. Among them, L-valine, L-proline, DL-arabinose, pentanoic acid, D-allose, and D-glucose in urine and D-lactic acid and glycerol in fecal metabolites depended on the exposure of time and dose. In addition, 16S rDNA sequencing analysis revealed that GSrE-induced hepatotoxicity significantly altered the composition of gut microbiota, namely, f_Muribaculaceae_Unclassified, Lactobacillus, Bacteroides, Lachnospiraceae_NK4A136_group, f_Ruminococcaceae_Unclassified, Prevotellaceae_Ga6A1_group, and Escherichia-Shigella. The correlation analysis between gut microbiota and differential metabolites showed the crosstalk between the gut microbiota and metabolism in host involving energy, lipid, and amino acid metabolisms. In summary, our findings revealed that peripheral metabolism and gut microbiota disorders were time- and dose-related and the correlation between gut microbiota and metabolites in GSrE-induced hepatotoxicity.
Collapse
Affiliation(s)
- Xinyi Gu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuwei Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengna Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qixue Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Long Chen
- Experiment Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqun Jia
- Experiment Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shan Cao
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai, China
| | - Ting Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Mingmei Zhou
| | - Xiaojun Gou
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai, China
- Xiaojun Gou
| |
Collapse
|
41
|
Shah HS, Moreno LO, Morieri ML, Tang Y, Mendonca C, Jobe JM, Thacker JB, Mitri J, Monti S, Niewczas MA, Pennathur S, Doria A. Serum Orotidine: A Novel Biomarker of Increased CVD Risk in Type 2 Diabetes Discovered Through Metabolomics Studies. Diabetes Care 2022; 45:1882-1892. [PMID: 35696261 PMCID: PMC9346986 DOI: 10.2337/dc21-1789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 04/26/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To identify novel biomarkers of cardiovascular disease (CVD) risk in type 2 diabetes (T2D) via a hypothesis-free global metabolomics study, while taking into account renal function, an important confounder often overlooked in previous metabolomics studies of CVD. RESEARCH DESIGN AND METHODS We conducted a global serum metabolomics analysis using the Metabolon platform in a discovery set from the Joslin Kidney Study having a nested case-control design comprising 409 individuals with T2D. Logistic regression was applied to evaluate the association between incident CVD events and each of the 671 metabolites detected by the Metabolon platform, before and after adjustment for renal function and other CVD risk factors. Significant metabolites were followed up with absolute quantification assays in a validation set from the Joslin Heart Study including 599 individuals with T2D with and without clinical evidence of significant coronary heart disease (CHD). RESULTS In the discovery set, serum orotidine and 2-piperidinone were significantly associated with increased odds of incident CVD after adjustment for glomerular filtration rate (GFR) (odds ratio [OR] per SD increment 1.94 [95% CI 1.39-2.72], P = 0.0001, and 1.62 [1.26-2.08], P = 0.0001, respectively). Orotidine was also associated with increased odds of CHD in the validation set (OR 1.39 [1.11-1.75]), while 2-piperidinone did not replicate. Furthermore, orotidine, being inversely associated with GFR, mediated 60% of the effects of declining renal function on CVD risk. Addition of orotidine to established clinical predictors improved (P < 0.05) C statistics and discrimination indices for CVD risk (ΔAUC 0.053, rIDI 0.48, NRI 0.42) compared with the clinical predictors alone. CONCLUSIONS Through a robust metabolomics approach, with independent validation, we have discovered serum orotidine as a novel biomarker of increased odds of CVD in T2D, independent of renal function. Additionally, orotidine may be a biological mediator of the increased CVD risk associated with poor kidney function and may help improve CVD risk prediction in T2D.
Collapse
Affiliation(s)
- Hetal S. Shah
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Lorena Ortega Moreno
- Department of Basic Health Sciences, Universidad Rey Juan Carlos, Alcorcón, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), Universidad Rey Juan Carlos, Alcorcón, Spain
| | | | - Yaling Tang
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Christine Mendonca
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
| | - Jenny Marie Jobe
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
| | - Jonathan B. Thacker
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Joanna Mitri
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Stefano Monti
- Computational Biomedicine, Department of Medicine, Boston University, Boston, MA
| | - Monika A. Niewczas
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
42
|
Xu R, Wang T, Ding FF, Zhou NN, Qiao F, Chen LQ, Du ZY, Zhang ML. Lactobacillus plantarum Ameliorates High-Carbohydrate Diet-Induced Hepatic Lipid Accumulation and Oxidative Stress by Upregulating Uridine Synthesis. Antioxidants (Basel) 2022; 11:antiox11071238. [PMID: 35883730 PMCID: PMC9312134 DOI: 10.3390/antiox11071238] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
The overconsumption of carbohydrates induces oxidative stress and lipid accumulation in the liver, which can be alleviated by modulation of intestinal microbiota; however, the underlying mechanism remains unclear. Here, we demonstrated that a strain affiliated with Lactobacillus plantarum (designed as MR1) efficiently attenuated lipid deposition, oxidative stress, as well as inflammatory response, which are caused by high-carbohydrate diet (HC) in fish with poor utilization ability of carbohydrates. Serum untargeted metabolome analysis indicated that pyrimidine metabolism was the significantly changed pathway among the groups. In addition, the content of serum uridine was significantly decreased in the HC group compared with the control group, while it increased by supplementation with L. plantarum MR1. Further analysis showed that addition of L. plantarum MR1 reshaped the composition of gut microbiota and increased the content of intestinal acetate. In vitro experiment showed that sodium acetate could induce the synthesis of uridine in hepatocytes. Furthermore, we proved that uridine could directly ameliorate oxidative stress and decrease liver lipid accumulation in the hepatocytes. In conclusion, this study indicated that probiotic L. plantarum MR1 ameliorated high-carbohydrate diet-induced hepatic lipid accumulation and oxidative stress by increasing the circulating uridine, suggesting that intestinal microbiota can regulate the metabolism of nucleotides to maintain host physiological homeostasis.
Collapse
|
43
|
Mizuno R, Hojo H, Takahashi M, Kashio S, Enya S, Nakao M, Konishi R, Yoda M, Harata A, Hamanishi J, Kawamoto H, Mandai M, Suzuki Y, Miura M, Bamba T, Izumi Y, Kawaoka S. Remote solid cancers rewire hepatic nitrogen metabolism via host nicotinamide-N-methyltransferase. Nat Commun 2022; 13:3346. [PMID: 35705545 PMCID: PMC9200709 DOI: 10.1038/s41467-022-30926-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 05/20/2022] [Indexed: 12/14/2022] Open
Abstract
Cancers disrupt host homeostasis in various manners but the identity of host factors underlying such disruption remains largely unknown. Here we show that nicotinamide-N-methyltransferase (NNMT) is a host factor that mediates metabolic dysfunction in the livers of cancer-bearing mice. Multiple solid cancers distantly increase expression of Nnmt and its product 1-methylnicotinamide (MNAM) in the liver. Multi-omics analyses reveal suppression of the urea cycle accompanied by accumulation of amino acids, and enhancement of uracil biogenesis in the livers of cancer-bearing mice. Importantly, genetic deletion of Nnmt leads to alleviation of these metabolic abnormalities, and buffers cancer-dependent weight loss and reduction of the voluntary wheel-running activity. Our data also demonstrate that MNAM is capable of affecting urea cycle metabolites in the liver. These results suggest that cancers up-regulate the hepatic NNMT pathway to rewire liver metabolism towards uracil biogenesis rather than nitrogen disposal via the urea cycle, thereby disrupting host homeostasis. The presence of cancer can induce systemic disruption of the host homeostasis. Here, the authors show that different solid tumours remotely increase hepatic nicotinamide-Nmethyltransferase disrupting the host urea cycle metabolism in the liver.
Collapse
Affiliation(s)
- Rin Mizuno
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Hiroaki Hojo
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International (ATR), Kyoto, 619-0237, Japan.,ERATO Sato Live Bio-forecasting Project, Japan Science and Technology Agency (JST), Kyoto, 619-0237, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Soshiro Kashio
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Sora Enya
- The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International (ATR), Kyoto, 619-0237, Japan.,ERATO Sato Live Bio-forecasting Project, Japan Science and Technology Agency (JST), Kyoto, 619-0237, Japan
| | - Motonao Nakao
- Division of Metabolomics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Riyo Konishi
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Mayuko Yoda
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Ayano Harata
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Hiroshi Kawamoto
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Yutaka Suzuki
- Graduate School of Frontier Science, The University of Tokyo, Chiba, 277-8562, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shinpei Kawaoka
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan. .,The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International (ATR), Kyoto, 619-0237, Japan. .,ERATO Sato Live Bio-forecasting Project, Japan Science and Technology Agency (JST), Kyoto, 619-0237, Japan. .,Department of Integrative Bioanalytics, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, 980-8575, Japan.
| |
Collapse
|
44
|
Niu K, Bai P, Yang B, Feng X, Qiu F. Asiatic acid alleviates metabolism disorders in ob/ob mice: mechanistic insights. Food Funct 2022; 13:6934-6946. [PMID: 35696250 DOI: 10.1039/d2fo01069k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glucolipid metabolism disorders pose a serious and global health problem, and more effective prevention and treatment methods are urgently needed. In this study, ob/ob mice were used to explore the potential mechanism explaining how asiatic acid (AA) regulates glucolipid metabolism disorders. Five-week AA treatment (30 mg kg-1) significantly improved a host of metabolic factors in ob/ob mice, including hyperglycemia, hyperlipidemia, insulin resistance, and liver histopathology. Combined analysis of untargeted liver metabolomics, liver transcriptomics, and the gut microbiome was conducted, and the results showed that AA alleviates metabolic disorders in ob/ob mice through regulating pyrimidine metabolism, activating PPAR-γ, and modulating gut microbiota. AA treatment remarkedly increased the levels of cytosine and cytidine, two crucial endogenous metabolites related to pyrimidine metabolism, which were significantly decreased in ob/ob mice. AA treatment also affected the levels of 13-S-hydroxyoctadecadienoic acid, an endogenous PPAR-γ agonist. The abundances of Lachnospiraceae_NK4A136_group and norank_f__norank_o__Clostridia_UCG-014 were increased after AA treatment. Meanwhile, correlation analysis showed that endogenous metabolites and gut microbiota were strongly correlated. These findings indicated that AA supplements might be beneficial for the prevention of metabolic disorders.
Collapse
Affiliation(s)
- Kaixia Niu
- School of Chinese Materia Medica, and State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Pengpeng Bai
- School of Chinese Materia Medica, and State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Bingbing Yang
- School of Chinese Materia Medica, and State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xinchi Feng
- School of Chinese Materia Medica, and State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Feng Qiu
- School of Chinese Materia Medica, and State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
45
|
Wang X, Wang Z, Huang R, Lu Z, Chen X, Huang D. UPP1 Promotes Lung Adenocarcinoma Progression through Epigenetic Regulation of Glycolysis. Aging Dis 2022; 13:1488-1503. [PMID: 36186123 PMCID: PMC9466982 DOI: 10.14336/ad.2022.0218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/18/2022] [Indexed: 11/01/2022] Open
Affiliation(s)
- Xuan Wang
- Department of Thoracic Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China.
- Correspondence should be addressed to: Dr. Dayu Huang (), Dr. Xiaofeng Chen (); Dr. Xuan Wang ().Department of Thoracic Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Renhong Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhouyi Lu
- Department of Thoracic Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China.
| | - Xiaofeng Chen
- Department of Thoracic Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China.
- Correspondence should be addressed to: Dr. Dayu Huang (), Dr. Xiaofeng Chen (); Dr. Xuan Wang ().Department of Thoracic Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Dayu Huang
- Department of Thoracic Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China.
- Correspondence should be addressed to: Dr. Dayu Huang (), Dr. Xiaofeng Chen (); Dr. Xuan Wang ().Department of Thoracic Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| |
Collapse
|
46
|
Kang J, Kim AH, Jeon I, Oh J, Jang IJ, Lee S, Cho JY. Endogenous metabolic markers for predicting the activity of dihydropyrimidine dehydrogenase. Clin Transl Sci 2021; 15:1104-1111. [PMID: 34863048 PMCID: PMC9099117 DOI: 10.1111/cts.13203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/09/2021] [Accepted: 11/02/2021] [Indexed: 12/01/2022] Open
Abstract
Five‐fluorouracil (5‐FU) is a chemotherapeutic agent that is mainly metabolized by the rate‐limiting enzyme dihydropyrimidine dehydrogenase (DPD). The DPD enzyme activity deficiency involves a wide range of severities. Previous studies have demonstrated the effect of a DPYD single nucleotide polymorphism on 5‐FU efficacy and highlighted the importance of studying such genes for cancer treatment. Common polymorphisms of DPYD in European ancestry populations are less frequently present in Koreans. DPD is also responsible for the conversion of endogenous uracil (U) into dihydrouracil (DHU). We quantified U and DHU in plasma samples of healthy male Korean subjects, and samples were classified into two groups based on DHU/U ratio. The calculated DHU/U ratios ranged from 0.52 to 7.12, and the two groups were classified into the 10th percentile and 90th percentile for untargeted metabolomics analysis using liquid chromatography‐quantitative time‐of‐flight‐mass spectrometry. A total of 4440 compounds were detected and filtered out based on a coefficient of variation below 30%. Our results revealed that six metabolites differed significantly between the high activity group and low activity group (false discovery rate q‐value < 0.05). Uridine was significantly higher in the low DPD activity group and is a precursor of U involved in pyrimidine metabolism; therefore, we speculated that DPD deficiency can influence uridine levels in plasma. Furthermore, the cutoff values for detecting DPD deficient patients from previous studies were unsuitable for Koreans. Our metabolomics approach is the first study that reported the DHU/U ratio distribution in healthy Korean subjects and identified a new biomarker of DPD deficiency.
Collapse
Affiliation(s)
- Jihyun Kang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Andrew HyoungJin Kim
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Inseung Jeon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Jaeseong Oh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - In-Jin Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
47
|
Xue B, Hong Q, Li X, Lu M, Zhou J, Yue S, Wang Z, Wang L, Peng Q, Xue B. Hepatic Injury Induced by Dietary Energy Level via Lipid Accumulation and Changed Metabolites in Growing Semi-Fine Wool Sheep. Front Vet Sci 2021; 8:745078. [PMID: 34631866 PMCID: PMC8494768 DOI: 10.3389/fvets.2021.745078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/17/2021] [Indexed: 01/22/2023] Open
Abstract
Liver injury threatens the overall health of an organism, as it is the core organ of the animal body. Liver metabolism is affected by numerous factors, with dietary energy level being a crucial one. Therefore, the present study aimed to evaluate hepatic injury and to describe its metabolic mechanism in ruminants fed diets with different dietary energy levels. A total of 25 Yunnan semi-fine wool sheep were fed diets with five dietary metabolic energy levels and were randomly assigned to five groups as follows: low energy (LE), medium–low energy (MLE), medium energy (ME), medium–high energy (MHE), and high energy (HE). The results revealed that the average optical density (AOD) of lipid droplets in the LE, MLE, and HE groups was higher than that in the ME and MHE groups. The enzyme activity of alanine aminotransferase (ALT) was the lowest in the ME group. An increase in dietary energy level promoted the superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activity and altered the malondialdehyde (MDA) and protein carbonyl (PCO) concentration quadratically. In addition, both high and low dietary energy levels upregulated the mRNA abundance of proinflammatory cytokine interleukin (IL)-1β, nuclear factor-kappa B (NF-κB), and tumor necrosis factor (TNF)-α. Metabonomic analysis revealed that 142, 77, 65, and 108 differential metabolites were detected in the LE, MLE, MHE, and HE groups, compared with ME group respectively. These metabolites were involved in various biochemical pathways, such as glycolipid, bile acid, and lipid metabolism. In conclusion, both high and low dietary energy levels caused hepatic injury. Section staining and metabonomic results revealed that hepatic injury might be caused by altered metabolism and lipid accumulation induced by lipid mobilization.
Collapse
Affiliation(s)
- Benchu Xue
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Qionghua Hong
- Yunna Academy of Animal Science and Vetarinary Medicine, Kunming, China
| | - Xiang Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Mingli Lu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jia Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shuangming Yue
- Department of Bioengineering, Sichuan Water Conservancy College, Chengdu, China
| | - Zhisheng Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Lizhi Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Quanhui Peng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bai Xue
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
48
|
Liu Y, Xie C, Zhai Z, Deng ZY, De Jonge HR, Wu X, Ruan Z. Uridine attenuates obesity, ameliorates hepatic lipid accumulation and modifies the gut microbiota composition in mice fed with a high-fat diet. Food Funct 2021; 12:1829-1840. [PMID: 33527946 DOI: 10.1039/d0fo02533j] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Uridine (UR) is a pyrimidine nucleoside that plays an important role in regulating glucose and lipid metabolism. The aim of this study was to investigate the effect of UR on obesity, fat accumulation in liver, and gut microbiota composition in high-fat diet (HFD)-fed mice. ICR mice were, respectively, divided into 3 groups for 8 weeks, that is, control (CON, n = 12), high fat diet (HFD, n = 16), and HFD + UR groups (0.4 mg mL-1 in drinking water, n = 16). UR supplementation significantly reduced the body weight and suppressed the accumulation of subcutaneous, epididymal, and mesenteric WAT in HFD-fed mice (P < 0.05). Meanwhile, UR also decreased the lipid droplet accumulation in the liver and liver organoids (P < 0.05). In addition, UR supplementation increased bacterial diversity and Bacteroidetes abundance, and decreased the Firmicutes-to-Bacteroidetes ratio in HFD-fed mice significantly (P < 0.05). UR promoted the growth of butyrate-producing bacteria of Odoribacter, unidentified-Ruminococcaceae, Intestinimonas, Ruminiclostridium, and unidentified-Lachnospiraceae. A close correlation between several specific bacterial phyla or genera and the levels of WAT weight, hepatic TC, or hepatic TG genera was revealed through Spearman's correlation analysis. These results demonstrated that UR supplementation could be beneficial by attenuating HFD-induced obesity and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Yilin Liu
- School of Food Science and Technology, State Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China. and Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha 410125, China.
| | - Chunyan Xie
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China and Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Zhenya Zhai
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha 410125, China.
| | - Ze-Yuan Deng
- School of Food Science and Technology, State Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Hugo R De Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Xin Wu
- School of Food Science and Technology, State Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China. and Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha 410125, China. and Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Zheng Ruan
- School of Food Science and Technology, State Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| |
Collapse
|
49
|
Zhang Z, Chen X, Cui B. Modulation of the fecal microbiome and metabolome by resistant dextrin ameliorates hepatic steatosis and mitochondrial abnormalities in mice. Food Funct 2021; 12:4504-4518. [PMID: 33885128 DOI: 10.1039/d1fo00249j] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Targeting the gut-liver axis by manipulating the intestinal microbiome is a promising therapy for nonalcoholic fatty liver disease (NAFLD). This study modulated the intestinal microbiota to explore whether resistant dextrin, as a potential prebiotic, could ameliorate high-fat diet (HFD)-induced hepatic steatosis in C57BL/6J mice. After two months of feeding, significant hepatic steatosis with mitochondrial dysfunction was observed in the HFD-fed mice. However, the concentrations of triglycerides and malondialdehyde in liver tissue and the levels of alanine aminotransferase and aspartate aminotransferase in the serum of mice fed an HFD plus resistant dextrin diet (HFID) were significantly decreased compared to the HFD-fed mice. Additionally, hepatic mitochondrial integrity and reactive oxygen species accumulation were improved in HFID-fed mice, ameliorating hepatic steatosis. The fecal microbiome of HFD-fed mice was enriched in Bifidobacterium, Lactobacillus, and Globicatella, while resistant dextrin increased the abundance of Parabacteroides, Blautia, and Dubosiella. Major changes in fecal metabolites were confirmed for HFID-fed mice, including those related to entero-hepatic circulation (i.e., bile acids), tryptophan metabolism (e.g., indole derivatives), and lipid metabolism (e.g., lipoic acid), as well as increased antioxidants including isorhapontigenin. Furthermore, resistant dextrin decreased inflammatory cytokine levels and intestinal permeability and ameliorated intestinal damage. Together, these findings augmented current knowledge on prebiotic treatment for NAFLD.
Collapse
Affiliation(s)
- Zheng Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | | | | |
Collapse
|
50
|
de Mello VD, Sehgal R, Männistö V, Klåvus A, Nilsson E, Perfilyev A, Kaminska D, Miao Z, Pajukanta P, Ling C, Hanhineva K, Pihlajamäki J. Serum aromatic and branched-chain amino acids associated with NASH demonstrate divergent associations with serum lipids. Liver Int 2021; 41:754-763. [PMID: 33219609 PMCID: PMC8048463 DOI: 10.1111/liv.14743] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/04/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) has been associated with multiple metabolic abnormalities. By applying a non-targeted metabolomics approach, we aimed at investigating whether serum metabolite profile that associates with NAFLD would differ in its association with NAFLD-related metabolic risk factors. METHODS & RESULTS A total of 233 subjects (mean ± SD: 48.3 ± 9.3 years old; BMI: 43.1 ± 5.4 kg/m2 ; 64 male) undergoing bariatric surgery were studied. Of these participants, 164 with liver histology could be classified as normal liver (n = 79), simple steatosis (SS, n = 40) or non-alcoholic steatohepatitis (NASH, n = 45). Among the identified fasting serum metabolites with higher levels in those with NASH when compared to those with normal phenotype were the aromatic amino acids (AAAs: tryptophan, tyrosine and phenylalanine), the branched-chain amino acids (BCAAs: leucine and isoleucine), a phosphatidylcholine (PC(16:0/16:1)) and uridine (all FDRp < 0.05). Only tryptophan was significantly higher in those with NASH compared to those with SS (FDRp < 0.05). Only the AAAs tryptophan and tyrosine correlated positively with serum total and LDL cholesterol (FDRp < 0.1), and accordingly, with liver LDLR at mRNA expression level. In addition, tryptophan was the single AA associated with liver DNA methylation of CpG sites known to be differentially methylated in those with NASH. CONCLUSIONS We found that serum levels of the NASH-related AAAs and BCAAs demonstrate divergent associations with serum lipids. The specific correlation of tryptophan with LDL-c may result from the molecular events affecting LDLR mRNA expression and NASH-associated methylation of genes in the liver.
Collapse
Affiliation(s)
- Vanessa D. de Mello
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Ratika Sehgal
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Ville Männistö
- Department of MedicineUniversity of Eastern Finland and Kuopio University HospitalKuopioFinland
| | - Anton Klåvus
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Emma Nilsson
- Epigenetics and Diabetes UnitDepartment of Clinical SciencesLund University Diabetes CentreMalmöSweden
| | - Alexander Perfilyev
- Epigenetics and Diabetes UnitDepartment of Clinical SciencesLund University Diabetes CentreMalmöSweden
| | - Dorota Kaminska
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Zong Miao
- Department of Human GeneticsDavid Geffen School of Medicine at University of California Los Angeles (UCLA)Los AngelesCAUSA
| | - Päivi Pajukanta
- Department of Human GeneticsDavid Geffen School of Medicine at University of California Los Angeles (UCLA)Los AngelesCAUSA,Institute for Precision HealthSchool of MedicineUCLALos AngelesCAUSA
| | - Charlotte Ling
- Epigenetics and Diabetes UnitDepartment of Clinical SciencesLund University Diabetes CentreMalmöSweden
| | - Kati Hanhineva
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland,Department of BiochemistryFood Chemistry and Food Development UnitUniversity of TurkuTurkuFinland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical NutritionDepartment of Clinical NutritionUniversity of Eastern FinlandKuopioFinland,Department of Medicine, Endocrinology and Clinical NutritionKuopio University HospitalKuopioFinland
| |
Collapse
|