1
|
Toczylowska B, Kalinowski P, Kacka-Piotrowska A, Duda P, Grąt M, Zieminska E. Metabolic Pattern of Brain Death-NMR-Based Metabolomics of Cerebrospinal Fluid. Int J Mol Sci 2025; 26:2719. [PMID: 40141360 PMCID: PMC11942502 DOI: 10.3390/ijms26062719] [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: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 03/28/2025] Open
Abstract
The aim of this study was to gain insight into the biochemical status of cerebrospinal fluid in the presence of brain death in life-supported patients. The biochemical status was determined via in vitro NMR spectroscopy of cerebrospinal fluid (CSF) obtained by lumbar puncture from 22 patients with confirmed brain death and compared with that of 34 control patients (without neurological diseases). Forty-one NMR signals from raw CSF samples and 20 signals from lipid extracts were analyzed using univariate and multivariate statistical methods. ANOVA revealed significant differences in all analyzed signals. No single biochemical marker was found to predict brain death. The CSF metabolic profiles of patients who died differed significantly from those of patients in the control group. There were many statistically significantly different compounds, including amino acids, ketone bodies, lactate, pyruvate, citrate, guanidinoacetate, choline, and glycerophosphocholine. Analysis of lipids revealed significant differences in cholesterol, estriol, and phosphoethanolamine. Discriminant analysis allows the analysis of metabolic profiles instead of single biomarkers of cerebrospinal fluid compounds. The results of our analysis allowed us to split the groups-the control group, which consisted of patients with a normal biochemical CSF composition, and the brain death group-with confirmed brain death.
Collapse
Affiliation(s)
- Beata Toczylowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 4 Trojdena Street, 02-109 Warsaw, Poland; (B.T.); (P.D.)
| | - Piotr Kalinowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland; (P.K.); (M.G.)
| | | | - Paulina Duda
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 4 Trojdena Street, 02-109 Warsaw, Poland; (B.T.); (P.D.)
| | - Michał Grąt
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland; (P.K.); (M.G.)
| | - Elzbieta Zieminska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| |
Collapse
|
2
|
Jiang Y, Hu Z, Huang R, Ho K, Wang P, Kang J. Metabolic reprogramming and macrophage expansion define ACPA-negative rheumatoid arthritis: insights from single-cell RNA sequencing. Front Immunol 2025; 15:1512483. [PMID: 39830504 PMCID: PMC11739280 DOI: 10.3389/fimmu.2024.1512483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
Background Anti-citrullinated peptide antibodies (ACPA)-negative (ACPA-) rheumatoid arthritis (RA) presents significant diagnostic and therapeutic challenges due to the absence of specific biomarkers, underscoring the need to elucidate its distinctive cellular and metabolic profiles for more targeted interventions. Methods Single-cell RNA sequencing data from peripheral blood mononuclear cells (PBMCs) and synovial tissues of patients with ACPA- and ACPA+ RA, as well as healthy controls, were analyzed. Immune cell populations were classified based on clustering and marker gene expression, with pseudotime trajectory analysis, weighted gene co-expression network analysis (WGCNA), and transcription factor network inference providing further insights. Cell-cell communication was explored using CellChat and MEBOCOST, while scFEA enabled metabolic flux estimation. A neural network model incorporating key genes was constructed to differentiate patients with ACPA- RA from healthy controls. Results Patients with ACPA- RA demonstrated a pronounced increase in classical monocytes in PBMCs and C1QChigh macrophages (p < 0.001 and p < 0.05). Synovial macrophages exhibited increased heterogeneity and were enriched in distinct metabolic pathways, including complement cascades and glutathione metabolism. The neural network model achieved reliable differentiation between patients with ACPA- RA and healthy controls (AUC = 0.81). CellChat analysis identified CD45 and CCL5 as key pathways facilitating macrophage-monocyte interactions in ACPA- RA, prominently involving iron-mediated metabolite communication. Metabolic flux analysis indicated elevated beta-alanine and glutathione metabolism in ACPA- RA macrophages. Conclusion These findings underscore that ACPA-negative rheumatoid arthritis is marked by elevated classical monocytes in circulation and metabolic reprogramming of synovial macrophages, particularly in complement cascade and glutathione metabolism pathways. By integrating single-cell RNA sequencing with machine learning, this study established a neural network model that robustly differentiates patients with ACPA- RA from healthy controls, highlighting promising diagnostic biomarkers and therapeutic targets centered on immune cell metabolism.
Collapse
Affiliation(s)
- Yafeng Jiang
- Department of Hematology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhaolan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Roujie Huang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaying Ho
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Pengfei Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin Kang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital of Central South University, Changsha, China
- Department of Rheumatology and Immunology, Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, Changsha, China
| |
Collapse
|
3
|
Malla S, Sajeevan KA, Acharya B, Chowdhury R, Saha R. Dissecting metabolic landscape of alveolar macrophage. Sci Rep 2024; 14:30383. [PMID: 39638830 PMCID: PMC11621776 DOI: 10.1038/s41598-024-81253-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: 10/08/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
The highly plastic nature of Alveolar Macrophage (AM) plays a crucial role in the defense against inhaled particulates and pathogens in the lungs. Depending on the signal, AM acquires either the classically activated M1 phenotype or the alternatively activated M2 phenotype. In this study, we investigate the metabolic shift in the activated phases of AM (M1 and M2 phases) by reconstructing context specific Genome-Scale Metabolic (GSM) models. Metabolic pathways such as pyruvate metabolism, arachidonic acid metabolism, chondroitin/heparan sulfate biosynthesis, and heparan sulfate degradation are found to be important driving forces in the development of the M1/M2 phenotypes. Additionally, we formulated a bilevel optimization framework named MetaShiftOptimizer to identify minimal modifications that shift one activated state (M1/M2) to the other. The identified reactions involve metabolites such as glycogenin, L-carnitine, 5-hydroperoxy eicosatetraenoic acid, and leukotriene B4, which show potential to be further investigated as significant factors for developing efficient therapy targets for severe respiratory disorders in the future. Overall, our study contributes to the understanding of the metabolic capabilities of the M1 and M2 phenotype of AM and identifies pathways and reactions that can be potential targets for polarization shift and also be used as therapeutic strategies against respiratory diseases.
Collapse
Affiliation(s)
- Sunayana Malla
- Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | - Bibek Acharya
- Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Ratul Chowdhury
- Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Rajib Saha
- Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
4
|
Lu J, Yao T, Fu S, Ye L. Metabolomic and microbiomic resilience of Hong Kong oysters to dual stressors: Zinc oxide nanoparticles and low salinity. CHEMOSPHERE 2024; 368:143722. [PMID: 39528128 DOI: 10.1016/j.chemosphere.2024.143722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Zinc oxide nanoparticles, increasingly used in industrial and consumer products, and low salinity, exacerbated by climate change-induced alterations in precipitation patterns, represent significant environmental pressures in estuarine and coastal environments. This study advances previous research on their impacts on Hong Kong oysters (Crassostrea hongkongensis) by integrating metabolomics of hepatopancreas and gills with intestinal microbiomics. Employing advanced multi-omics integration methods, our analysis reveals novel insights into metabolic resilience under combined stress conditions. This resilience is characterized by coordinated, organ-specific adjustments in energy metabolism (d-glucose 1-phosphate in hepatopancreas, cytidine in gills), antioxidant defenses (glutathione, meso-2,6-diaminoheptanedioate, pimelic acid in hepatopancreas; indole, 3-(3-hydroxyphenyl)propanoic acid in gills), immune function (l-glutamine, ergocalciferol in hepatopancreas; argininosuccinic acid in gills), and membrane stability (lanosterin in hepatopancreas, allantoin in gills). Notably, under dual stressors, we observed a previously undescribed stabilization of microbial alpha diversity and certain phyla, an absence of distinctive biomarkers, and certain metabolic activity stabilization within the intestinal microbiota. These findings suggest robust compensatory mechanisms that maintain physiological homeostasis and microbial balance under stress, contrasting with primarily negative impacts reported in previous studies. Integration of metabolomic and microbiomic data revealed coordinated responses between microbial community changes and metabolic adjustments, particularly in osmoregulation, energy metabolism and antioxidant defenses, under dual stressors. This comprehensive approach provides a more realistic model of environmental challenges, revealing sophisticated adaptive strategies in Hong Kong oysters. Our study offers critical insights for understanding bivalve resilience, informing conservation strategies, and managing marine ecosystems in the face of increasing anthropogenic pressures.
Collapse
Affiliation(s)
- Jie Lu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China; Sanya Tropical Fisheries Research Institute, Sanya, 572426, China
| | - Tuo Yao
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China; Sanya Tropical Fisheries Research Institute, Sanya, 572426, China; Tropical Fisheries Research and Development Center, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Sanya, 572018, China
| | - Shengli Fu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
| | - Lingtong Ye
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China.
| |
Collapse
|
5
|
Dos Santos K, Bertho G, Baudin M, Giraud N. Glutamine: A key player in human metabolism as revealed by hyperpolarized magnetic resonance. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2024; 144-145:15-39. [PMID: 39645348 DOI: 10.1016/j.pnmrs.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 12/09/2024]
Abstract
In recent years, there has been remarkable progress in the field of dissolution dynamic nuclear polarization (D-DNP). This method has shown significant potential for enhancing nuclear polarization by over 10,000 times, resulting in a substantial increase in sensitivity. The unprecedented signal enhancements achieved with D-DNP have opened new possibilities for in vitro analysis. This method enables the monitoring of structural and enzymatic kinetics with excellent time resolution at low concentrations. Furthermore, these advances can be straightforwardly translated to in vivo magnetic resonance imaging and magnetic resonance spectroscopy (MRI and MRS) experiments. D-DNP studies have used a range of 13C labeled molecules to gain deeper insights into the cellular metabolic pathways and disease hallmarks. Over the last 15 years, D-DNP has been used to analyze glutamine, a key player in the cellular metabolism, involved in many diseases including cancer. Glutamine is the most abundant amino acid in blood plasma and the major carrier of nitrogen, and it is converted to glutamate inside the cell, where the latter is the most abundant amino acid. It has been shown that increased glutamine consumption by cells is a hallmark of tumor cancer metabolism. In this review, we first highlight the significance of glutamine in metabolism, providing an in-depth description of its use at the cellular level as well as its specific roles in various organs. Next, we present a comprehensive overview of the principles of D-DNP. Finally, we review the state of the art in D-DNP glutamine analysis and its application in oncology, neurology, and perfusion marker studies.
Collapse
Affiliation(s)
- Karen Dos Santos
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques Université Paris Cité, 45 rue des Saints Pères, 75006 Paris, France
| | - Gildas Bertho
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques Université Paris Cité, 45 rue des Saints Pères, 75006 Paris, France
| | - Mathieu Baudin
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques Université Paris Cité, 45 rue des Saints Pères, 75006 Paris, France; Laboratoire des Biomolécules, LBM, Département de chimie, École Normale Supérieure, PSL Université, Sorbonne Université 45 rue d'Ulm, 75005 Paris, France
| | - Nicolas Giraud
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques Université Paris Cité, 45 rue des Saints Pères, 75006 Paris, France.
| |
Collapse
|
6
|
Leisching G, Yennemadi A, Gogan K, Keane J. Interferon α and β induce differential transcriptional and functional metabolic phenotypes in human macrophages and blunt glycolysis in response to antigenic stimuli. Eur J Immunol 2024; 54:e2451032. [PMID: 38993003 DOI: 10.1002/eji.202451032] [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: 01/25/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024]
Abstract
The impact of chronic exposure to type I interferons (IFN)-α2a, 2b, and β on macrophage metabolism, intimately linked to macrophage function, is not well understood. This study assesses the nuanced host responses induced by type I IFN cytokines, offering insights into potential therapeutic approaches in diseases associated with these cytokines. Employing a combination of transcriptional profiling and real-time functional analysis, we delineated metabolic reprogramming in response to chronic IFN exposure. Our results reveal distinct transcriptional metabolic profiles between macrophages chronically exposed to IFN-α and IFN-β. IFN-β significantly diminishes the oxygen consumption rate and glycolytic proton extrusion rate in macrophages. Conversely, IFN-α2b decreased parameters of mitochondrial fitness and induced a shift toward glutamine oxidation. Assessing the ability of macrophages to induce glycolysis in response to antigenic stimuli (LPS and iH37Rv), we found that chronic exposure to all IFN subtypes limited glycolytic induction. This study addresses a critical oversight in the literature, where individual roles of IFN subtypes are frequently amalgamated and lack distinction. These findings not only provide novel insights into the divergent effects of IFN-α2a, α2b, and β on macrophage metabolism but also highlight their potential implications for developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Gina Leisching
- Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Anjali Yennemadi
- Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Karl Gogan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Zhang Z, Wang Y, Xia L, Zhang Y. Roles of Critical Amino Acids Metabolism in The Interactions Between Intracellular Bacterial Infection and Macrophage Function. Curr Microbiol 2024; 81:280. [PMID: 39031203 DOI: 10.1007/s00284-024-03801-x] [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: 05/03/2024] [Accepted: 07/10/2024] [Indexed: 07/22/2024]
Abstract
Macrophages, as crucial participants in the innate immune system, respond to pathogenic challenges through their dynamic metabolic adjustments, demonstrating the intimate interplay between cellular metabolism and immune function. Bacterial infection of macrophages causes changes in macrophage metabolism, affecting both macrophage function and bacterial virulence and intracellular survival. This review explores the reprogramming of amino acid metabolism in macrophages in response to bacterial infection, with a particular focus on the influence of critical amino acids such as serine, glutamine, and arginine on the immune functions of macrophages; highlights the roles of these metabolic pathways in macrophage functions such as phagocytosis, inflammatory response, immune regulation, and pathogen clearance; reveals how pathogens exploit and manipulate the amino acid metabolism within macrophages to support their own growth and replication, thereby showcasing the intricate interplay between macrophages and pathogens. It provides a foundation for understanding the interactions between macrophages amino acid metabolism and pathogens, offering potential strategies and therapeutic targets for the development of novel anti-infection therapies.
Collapse
Affiliation(s)
- Zuowei Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, 212013, Jiangsu, China
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China
| | - Yurou Wang
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
8
|
Li D, Yao H, Ren Y, Shang J, Han X, Cao X, Song T, Zeng X. Testosterone regulates thymic remodeling by altering metabolic reprogramming in male rats. Gen Comp Endocrinol 2024; 348:114448. [PMID: 38191062 DOI: 10.1016/j.ygcen.2024.114448] [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: 10/11/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024]
Abstract
The thymus is an energy-consuming organ, and its metabolism changes with atrophy. Testosterone regulates thymus remodeling (atrophy and regeneration). However, the characteristics of the energy metabolism during testosterone-mediated thymic atrophy and regeneration remain unclear. In this study, we demonstrated that testosterone ablation (implemented by immunocastration and surgical castration) induced global metabolic changes in the thymus. Kyoto Encyclopedia of Genes and Genomes pathway enrichment for differential metabolites and metabolite set enrichment analysis for total metabolites revealed that testosterone ablation affected thymic glycolysis, glutamate metabolism, and fatty acid β-oxidation. Testosterone ablation-induced thymic regeneration was accompanied by attenuated glycolysis and glutamate metabolism and changed fatty acid composition and content. Testosterone supplementation in immunocastrated and surgically castrated rats enhanced glutaminolysis, reduced the level of unsaturated fatty acids, enhanced the β-oxidation of unsaturated fatty acids in the mitochondria, boosted the tricarboxylic acid (TCA) cycle, and accelerated thymic atrophy. Overall, these results imply that metabolic reprogramming is directly related to thymic remodeling.
Collapse
Affiliation(s)
- Dong Li
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Huan Yao
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Yonghao Ren
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Jiameng Shang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Xinfa Han
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Xiaohan Cao
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Tianzeng Song
- Institute of animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa 850009, Xizang, PR China.
| | - Xianyin Zeng
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China.
| |
Collapse
|
9
|
Baranovicova E, Kalenska D, Kaplan P, Kovalska M, Tatarkova Z, Lehotsky J. Blood and Brain Metabolites after Cerebral Ischemia. Int J Mol Sci 2023; 24:17302. [PMID: 38139131 PMCID: PMC10743907 DOI: 10.3390/ijms242417302] [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: 10/27/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
The study of an organism's response to cerebral ischemia at different levels is essential to understanding the mechanism of the injury and protection. A great interest is devoted to finding the links between quantitative metabolic changes and post-ischemic damage. This work aims to summarize the outcomes of the most studied metabolites in brain tissue-lactate, glutamine, GABA (4-aminobutyric acid), glutamate, and NAA (N-acetyl aspartate)-regarding their biological function in physiological conditions and their role after cerebral ischemia/reperfusion. We focused on ischemic damage and post-ischemic recovery in both experimental-including our results-as well as clinical studies. We discuss the role of blood glucose in view of the diverse impact of hyperglycemia, whether experimentally induced, caused by insulin resistance, or developed as a stress response to the cerebral ischemic event. Additionally, based on our and other studies, we analyze and critically discuss post-ischemic alterations in energy metabolites and the elevation of blood ketone bodies observed in the studies on rodents. To complete the schema, we discuss alterations in blood plasma circulating amino acids after cerebral ischemia. So far, no fundamental brain or blood metabolite(s) has been recognized as a relevant biological marker with the feasibility to determine the post-ischemic outcome or extent of ischemic damage. However, studies from our group on rats subjected to protective ischemic preconditioning showed that these animals did not develop post-ischemic hyperglycemia and manifested a decreased metabolic infringement and faster metabolomic recovery. The metabolomic approach is an additional tool for understanding damaging and/or restorative processes within the affected brain region reflected in the blood to uncover the response of the whole organism via interorgan metabolic communications to the stressful cerebral ischemic challenge.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| |
Collapse
|
10
|
Li XY, Zeng ZX, Cheng ZX, Wang YL, Yuan LJ, Zhai ZY, Gong W. Common pathogenic bacteria-induced reprogramming of the host proteinogenic amino acids metabolism. Amino Acids 2023; 55:1487-1499. [PMID: 37814028 PMCID: PMC10689525 DOI: 10.1007/s00726-023-03334-w] [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: 04/02/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023]
Abstract
Apart from cancer, metabolic reprogramming is also prevalent in other diseases, such as bacterial infections. Bacterial infections can affect a variety of cells, tissues, organs, and bodies, leading to a series of clinical diseases. Common Pathogenic bacteria include Helicobacter pylori, Salmonella enterica, Mycobacterium tuberculosis, Staphylococcus aureus, and so on. Amino acids are important and essential nutrients in bacterial physiology and support not only their proliferation but also their evasion of host immune defenses. Many pathogenic bacteria or opportunistic pathogens infect the host and lead to significant changes in metabolites, especially the proteinogenic amino acids, to inhibit the host's immune mechanism to achieve its immune evasion and pathogenicity. Here, we review the regulation of host metabolism, while host cells are infected by some common pathogenic bacteria, and discuss how amino acids of metabolic reprogramming affect bacterial infections, revealing the potential adjunctive application of amino acids alongside antibiotics.
Collapse
Affiliation(s)
- Xiao-Yue Li
- The First School of Clinical Medicine, Southern Medical University, Guangdong, 510515, China
| | - Zi-Xin Zeng
- The First School of Clinical Medicine, Southern Medical University, Guangdong, 510515, China
| | - Zhi-Xing Cheng
- The First School of Clinical Medicine, Southern Medical University, Guangdong, 510515, China
| | - Yi-Lin Wang
- The First School of Clinical Medicine, Southern Medical University, Guangdong, 510515, China
| | - Liang-Jun Yuan
- The First School of Clinical Medicine, Southern Medical University, Guangdong, 510515, China
| | - Zhi-Yong Zhai
- Shenzhen Hospital, Southern Medical University, Shenzhen Clinical Medical College, Southern Medical University, Guangdong, 518101, China.
| | - Wei Gong
- Shenzhen Hospital, Southern Medical University, Shenzhen Clinical Medical College, Southern Medical University, Guangdong, 518101, China.
| |
Collapse
|
11
|
Gu B, Pan F, Wang H, Zou Z, Song J, Xing J, Tang X, Zhan Y. Untargeted LC-MS metabolomics reveals the metabolic responses in olive flounder subjected to hirame rhabdovirus infection. Front Immunol 2023; 14:1148740. [PMID: 37711614 PMCID: PMC10498126 DOI: 10.3389/fimmu.2023.1148740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 08/02/2023] [Indexed: 09/16/2023] Open
Abstract
Hirame novirhabdovirus (HIRRV), which mainly infects the olive flounder (Paralichthys olivaceus), is considered to be one of the most serious viral pathogens threatening the global fish culture industry. However, little is known about the mechanism of host-pathogen interactions at the metabolomic level. In this study, in order to explore the metabolic response of olive flounder to HIRRV infection, liquid chromatography mass spectrometry (LC-MS) was used to detect the changes of endogenous compounds of the olive flounder after HIRRV infection. A total of 954 unique masses were obtained, including 495 metabolites and 459 lipids. Among them, 7 and 173 qualified differential metabolites were identified at 2 days and 7 days post-infection, respectively. Distinct metabolic profiles were observed along with viral infection. At the early stage of infection, only a few metabolites were perturbed. Among them, the level of inosine and carnosine were increased and the potential antiviral ability of these two metabolites was further confirmed by exogenous addition experiment. At the late stage of HIRRV infection, the metabolic profiles changed remarkably. The changes in amino acids and nucleotides especially the 7-methylguanine also accelerated the amplification of viral particles. And the down-regulation of glutathione (GSH) implied an elevated level of ROS (reactive oxygen species) that attenuated the immune system of flounders. HIRRV also induced the accumulation of purine and reduction of pyrimidine, and elevated LPC and LPE levels. The unbalanced purine/pyrimidine and altered lipid profile may be beneficial for the replication and infection of HIRRV at the late stage of infection. These findings provide new insights into the pathogenic mechanism of HIRRV infection in olive flounder.
Collapse
Affiliation(s)
- Bingyu Gu
- College of Marine Life Science, Ocean University of China, Qingdao, China
- Laboratory of Pathology and Immunology of Aquatic Animals, Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education (KLMME), Fisheries College, Ocean University of China, Qingdao, China
| | - Fenghuang Pan
- Laboratory of Pathology and Immunology of Aquatic Animals, Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education (KLMME), Fisheries College, Ocean University of China, Qingdao, China
| | - Hongxiang Wang
- Laboratory of Pathology and Immunology of Aquatic Animals, Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education (KLMME), Fisheries College, Ocean University of China, Qingdao, China
| | - Zhiyi Zou
- Haide College, Ocean University of China, Qingdao, China
| | - Junya Song
- Haide College, Ocean University of China, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education (KLMME), Fisheries College, Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture, Ministry of Education (KLMME), Fisheries College, Ocean University of China, Qingdao, China
| | - Yuanchao Zhan
- College of Marine Life Science, Ocean University of China, Qingdao, China
| |
Collapse
|
12
|
Radford-Smith DE, Yates AG, Rizvi L, Anthony DC, Probert F. HDL and LDL have distinct, opposing effects on LPS-induced brain inflammation. Lipids Health Dis 2023; 22:54. [PMID: 37095493 PMCID: PMC10124044 DOI: 10.1186/s12944-023-01817-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
Endotoxemia and sepsis induce neuroinflammation and increase the risk of neurodegenerative disorders although the mechanism by which peripheral infection leads to brain inflammation is not well understood. While circulating serum lipoproteins are known immunometabolites with the potential to modulate the acute phase response and cross the blood brain barrier, their contribution to neuroinflammation during systemic infection is unknown. The objective of this study was to elucidate the mechanisms by which lipoprotein subclasses modulate lipopolysaccharide (LPS)-induced neuroinflammation. Adult C57BL/6 mice were divided into 6 treatment groups, including a sterile saline vehicle control group (n = 9), an LPS group (n = 11), a premixed LPS + HDL group (n = 6), a premixed LPS + LDL group (n = 5), a HDL only group (n = 6) and an LDL only group (n = 3). In all cases injections were administered intraperitoneally. LPS was administered at 0.5 mg/kg, and lipoproteins were administered at 20 mg/kg. Behavioural testing and tissue collection was performed 6 h post-injection. The magnitude of peripheral and central inflammation was determined by qPCR of pro-inflammatory genes in fresh liver and brain. Metabolite profiles of liver, plasma and brain were determined by 1H NMR. Endotoxin concentration in the brain was measured by the Limulus Amoebocyte Lysate (LAL) assay. Co-administration of LPS + HDL exacerbated both peripheral and central inflammation, whilst LPS + LDL attenuated this inflammation. Metabolomic analysis identified several metabolites significantly associated with LPS-induced inflammation, which were partially rescued by LDL, but not HDL. Endotoxin was detected at significantly greater concentrations in the brains of animals that received LPS + HDL compared to LPS + saline, but not those that received LPS + LDL. These results suggest that HDL may promote neuroinflammation through direct shuttling of endotoxin to the brain. In contrast, LDL was shown to have anti-neuroinflammatory properties in this study. Our results indicate that lipoproteins may be useful targets in neuroinflammation and neurodegeneration associated with endotoxemia and sepsis.
Collapse
Affiliation(s)
- Daniel E Radford-Smith
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK.
- Department of Chemistry, University of Oxford, Oxford, UK.
| | - Abi G Yates
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Laila Rizvi
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Daniel C Anthony
- Department of Pharmacology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Fay Probert
- Department of Chemistry, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Modulations in human neutrophil metabolome and S-glutathionylation of glycolytic pathway enzymes during the course of extracellular trap formation. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166581. [PMID: 36265832 DOI: 10.1016/j.bbadis.2022.166581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/19/2022]
Abstract
Neutrophil extracellular trap formation (NETosis) has been irrefutably referred to as a distinct and unique form of active cell death with the purpose to counteract invading pathogens or augmenting the inflammatory cascade. Since the discovery, consistent efforts have been made to understand the various aspects of the initiation and sustenance of NETosis. In this study, using a global metabolomics approach during the phorbol 12-myristate 13-acetate (PMA) induced NETosis in human neutrophils, various metabolic pathways were found to be altered which includes intermediates related to, carbohydrate metabolism, and redox related metabolites, nucleic acid metabolism, and amino acids metabolism. Enrichment analysis of the metabolite sets highlighted the importance of the pentose phosphate pathway (PPP) and glutathione metabolism PMA-induced NETotic neutrophils. Further, analysis of the glutathyniolation status of neutrophil proteins by Matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) indicated six different glutathionylated proteins: among them, two metabolically important proteins were α-enolase and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) with MALDI score 166 and 70 respectively. Other proteins were lactoferrin, β-actin, c-myc promoter-binding protein, and uracil DNA glycosylase with MALDI scores of 96, 167, 104, and 68 respectively. Besides, activation of signalling proteins involved in metabolic regulation is also correlated with NETosis. Altogether, a balance between reactive oxygen species-glutathione metabolism seems to regulate the activity of glycolytic enzymes such as GAPDH and α-enolase during PMA-induced NETosis in a time-dependent manner.
Collapse
|
14
|
Batchu P, Naldurtiker A, Kouakou B, Terrill TH, McCommon GW, Kannan G. Metabolomic exploration of the effects of habituation to livestock trailer and extended transportation in goats. Front Mol Biosci 2022; 9:1027069. [PMID: 36465562 PMCID: PMC9714579 DOI: 10.3389/fmolb.2022.1027069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/01/2022] [Indexed: 03/26/2024] Open
Abstract
Goats raised for meat production are often transported long distances. Twelve-month-old male Spanish goats were used to determine the effects of habituation to trailers on plasma metabolomic profiles when transported for extended periods. In a split-plot design, 168 goats were separated into two treatment (TRT; whole plot) groups and maintained on two different paddocks. Concentrate supplement was fed to one group inside two livestock trailers (habituated group, H), while the other group received the same quantity of concentrate, but not inside the trailers (non-habituated, NH). Goats were subjected to a 10-h transportation stress in 4 replicates (n = 21 goats/replicate/TRT) after 4 weeks of habituation period. Blood samples were collected prior to loading, 20 min after loading (0 h), and at 2, 4, 6, 8, and 10 h of transportation (Time; subplot). A targeted quantitative metabolomics approach was employed to analyze the samples. The data were analyzed using R software and MIXED procedures in SAS. Several amino acids (alanine, serine, glycine, histidine, glutamate, trans-hydroxyproline, asparagine, threonine, methylhistidine, ornithine, proline, leucine, tryptophan) were higher (p < 0.05) in the H group compared to the NH group. Six long-chain acylcarnitines were higher (p < 0.05), while free (C0) and short-chain (C3, C5) carnitines were lower (p < 0.05) in the NH goats compared to the H goats. In general, amino acid concentrations decreased and long-chain acylcarnitine (>C10) levels increased with transportation time (p < 0.05). Butyric acid, α-ketoglutaric acid, and α-aminoadipic acid concentrations were lower (p < 0.05) and β-hydroxybutyric acid concentrations were higher in the NH goats compared to the H goats. Plasma glucose, non-esterified fatty acid (NEFA) and urea nitrogen concentrations were significantly influenced by Time (p < 0.01). Plasma NEFA concentrations were significantly lower (p < 0.01) in the H group than the NH group. Habituation to trailers can be beneficial in enhancing stress coping abilities in goats due to higher concentrations of metabolites such as butyrate and certain amino acids that support antioxidant activities and immune function. Plasma long-chain acylcarnitines may be good indicators of stress during long-distance transportation in goats.
Collapse
Affiliation(s)
| | | | | | | | | | - Govind Kannan
- Agricultural Research Station, Fort Valley State University, Fort Valley, GA, United States
| |
Collapse
|
15
|
Javaid A, Wang F, Horst EA, Diaz-Rubio ME, Wang LF, Baumgard LH, McFadden JW. Effects of acute intravenous lipopolysaccharide administration on the plasma lipidome and metabolome in lactating Holstein cows experiencing hyperlipidemia. Metabolomics 2022; 18:75. [PMID: 36125563 DOI: 10.1007/s11306-022-01928-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 08/01/2022] [Indexed: 10/14/2022]
Abstract
INTRODUCTION The effects of lipopolysaccharides (i.e., endotoxin; LPS) on metabolism are poorly defined in lactating dairy cattle experiencing hyperlipidemia. OBJECTIVES Our objective was to explore the effects of acute intravenous LPS administration on metabolism in late-lactation Holstein cows experiencing hyperlipidemia induced by intravenous triglyceride infusion and feed restriction. METHODS Ten non-pregnant lactating Holstein cows (273 ± 35 d in milk) were administered a single bolus of saline (3 mL of saline; n [Formula: see text] 5) or LPS (0.375 [Formula: see text]g of LPS/kg of body weight; n [Formula: see text] 5). Simultaneously, cows were intravenously infused a triglyceride emulsion and feed restricted for 16 h to induce hyperlipidemia in an attempt to model the periparturient period. Blood was sampled at routine intervals. Changes in circulating total fatty acid concentrations and inflammatory parameters were measured. Plasma samples were analyzed using untargeted lipidomics and metabolomics. RESULTS Endotoxin increased circulating serum amyloid A, LPS-binding protein, and cortisol concentrations. Endotoxin administration decreased plasma lysophosphatidylcholine (LPC) concentrations and increased select plasma ceramide concentrations. These outcomes suggest modulation of the immune response and insulin action. Lipopolysaccharide decreased the ratio of phosphatidylcholine to phosphatidylethanomanine, which potentially indicate a decrease in the hepatic activation of phosphatidylethanolamine N-methyltransferase and triglyceride export. Endotoxin administration also increased plasma concentrations of pyruvic and lactic acids, and decreased plasma citric acid concentrations, which implicate the upregulation of glycolysis and downregulation of the citric acid cycle (i.e., the Warburg effect), potentially in leukocytes. CONCLUSION Acute intravenous LPS administration decreased circulating LPC concentrations, modified ceramide and glycerophospholipid concentrations, and influenced intermediary metabolism in dairy cows experiencing hyperlipidemia.
Collapse
Affiliation(s)
- Awais Javaid
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA.
| | - Feiran Wang
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
- China Agricultural University, Beijing, 100193, China
| | - Erin A Horst
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - M Elena Diaz-Rubio
- Cornell Institute of Biotechnology, Cornell University, Ithaca, NY, 14853, USA
| | - Lin F Wang
- Henan Agricultural University, Zhengzhou, 450002, China
| | - Lance H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Joseph W McFadden
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
16
|
Baranovicova E, Kalenska D, Kovalska M, Lehotsky J. Hippocampal metabolic recovery as a manifestation of the protective effect of ischemic preconditioning in rats. Neurochem Int 2022; 160:105419. [PMID: 36113578 DOI: 10.1016/j.neuint.2022.105419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 08/25/2022] [Accepted: 09/04/2022] [Indexed: 10/31/2022]
Abstract
The ever-present risk of brain ischemic events in humans and its full prevention make the detailed studies of an organism's response to ischemia at different levels essential to understanding the mechanism of the injury as well as protection. We used the four-vessel occlusion as an animal model of forebrain ischemia to investigate its impact on the metabolic alterations in both the hippocampus and the blood plasma to see changes on the systemic level. By inducing sublethal ischemic stimuli, we focused on the endogenous phenomena known as ischemic tolerance. NMR spectroscopy was used to analyze relative metabolite levels in tissue extracts from rats' hippocampus and blood plasma in three various ischemic/reperfusion times: 3 h, 24 h, and 72 h. Hippocampal tissues were characterized by postischemically decreased glutamate and GABA (4-aminobutyrate) tissue content balanced with increased glutamine level, with most pronounced changes at 3 h reperfusion time. Glutamate (as well as glutamine) levels recovered towards the control levels on the third day, as if the glutamate re-synthesis would be firstly preferred before GABA. These results are indicating the higher feasibility of re-establishing of glutamatergic transmission three days after an ischemic event, in contrast to GABA-ergic. Tissue levels of N-acetylaspartate (NAA), as well as choline, were decreased without the tendency to recover three days after the ischemic event. Metabolomic analysis of blood plasma revealed that ischemically preconditioned rats, contrary to the non-preconditioned animals, did not show hyperglycemic conditions. Ischemically induced semi-ketotic state, manifested in increased plasma ketone bodies 3-hydroxybutyrate and acetoacetate, seems to be programmed to support the brain tissue revitalization after the ischemic event. These and other metabolites changes found in blood plasma as well as in the hippocampus were observed to a lower extent or recovered faster in preconditioned animals. Some metabolomic changes in hippocampal tissue extract were so strong that even single metabolites were able to differentiate between ischemic, ischemically preconditioned, and control brain tissues.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| |
Collapse
|
17
|
Circulating Metabolites in Relation to the Kidney Allograft Function in Posttransplant Patients. Metabolites 2022; 12:metabo12070661. [PMID: 35888785 PMCID: PMC9318187 DOI: 10.3390/metabo12070661] [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: 06/24/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 12/05/2022] Open
Abstract
End-stage kidney disease is preferably treated by kidney transplantation. The suboptimal function of the allograft often results in misbalances in kidney-controlled processes and requires long-term monitoring of allograft function and viability. As the kidneys are organs with a very high metabolomic rate, a metabolomics approach is suitable to describe systematic changes in post-transplant patients and has great potential for monitoring allograft function, which has not been described yet. In this study, we used blood plasma samples from 55 patients after primary kidney transplantation identically treated with immunosuppressants with follow-up 50 months in the mean after surgery and evaluated relative levels of basal plasma metabolites detectable by NMR spectroscopy. We were looking for the correlations between circulating metabolites levels and allograft performance and allograft rejection features. Our results imply a quantitative relationship between restricted renal function, insufficient hydroxylation of phenylalanine to tyrosine, lowered renal glutamine utilization, shifted nitrogen balance, and other alterations that are not related exclusively to the metabolism of the kidney. No link between allograft function and energy metabolism can be concluded, as no changes were found for glucose, glycolytic intermediates, and 3-hydroxybutyrate as a ketone body representative. The observed changes are to be seen as a superposition of changes in the comprehensive inter-organ metabolic exchange, when the restricted function of one organ may induce compensatory effects or cause secondary alterations. Particular differences in plasma metabolite levels in patients with acute cellular and antibody-mediated allograft rejection were considered rather to be related to the loss of kidney function than to the molecular mechanism of graft rejection since they largely follow the alterations observed by restricted allograft function. In the end, we showed using a simple mathematical model, multilinear regression, that the basal plasmatic metabolites correlated with allograft function expressed by the level of glomerular filtration rate (with creatinine: p-value = 4.0 × 10−26 and r = 0.94, without creatinine: p-value = 3.2 × 10−22 and r = 0.91) make the noninvasive estimation of the allograft function feasible.
Collapse
|
18
|
Amino acid sensor GCN2 promotes SARS-CoV-2 receptor ACE2 expression in response to amino acid deprivation. Commun Biol 2022; 5:651. [PMID: 35778545 PMCID: PMC9249868 DOI: 10.1038/s42003-022-03609-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 06/21/2022] [Indexed: 12/14/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been identified as a primary receptor for severe acute respiratory syndrome coronaviruses 2 (SARS-CoV-2). Here, we investigated the expression regulation of ACE2 in enterocytes under amino acid deprivation conditions. In this study, we found that ACE2 expression was upregulated upon all or single essential amino acid deprivation in human colonic epithelial CCD841 cells. Furthermore, we found that knockdown of general control nonderepressible 2 (GCN2) reduced intestinal ACE2 mRNA and protein levels in vitro and in vivo. Consistently, we revealed two GCN2 inhibitors, GCN2iB and GCN2-IN-1, downregulated ACE2 protein expression in CCD841 cells. Moreover, we found that increased ACE2 expression in response to leucine deprivation was GCN2 dependent. Through RNA-sequencing analysis, we identified two transcription factors, MAFB and MAFF, positively regulated ACE2 expression under leucine deprivation in CCD841 cells. These findings demonstrate that amino acid deficiency increases ACE2 expression and thereby likely aggravates intestinal SARS-CoV-2 infection. Amino acid deprivation increases ACE2 expression in the gut, potentially aggravating SARS-CoV-2 infection.
Collapse
|
19
|
Tosato M, Ciciarello F, Zazzara MB, Pais C, Savera G, Picca A, Galluzzo V, Coelho-Júnior HJ, Calvani R, Marzetti E, Landi F. Nutraceuticals and Dietary Supplements for Older Adults with Long COVID. Clin Geriatr Med 2022; 38:565-591. [PMID: 35868674 PMCID: PMC9212635 DOI: 10.1016/j.cger.2022.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matteo Tosato
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy
| | - Francesca Ciciarello
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy
| | - Maria Beatrice Zazzara
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy
| | - Cristina Pais
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy
| | - Giulia Savera
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy
| | - Vincenzo Galluzzo
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy
| | - Hélio José Coelho-Júnior
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, L.go F. Vito 8, Rome 00168, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy.
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, L.go F. Vito 8, Rome 00168, Italy
| | - Francesco Landi
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, Rome 00168, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, L.go F. Vito 8, Rome 00168, Italy
| |
Collapse
|
20
|
Dai YW, Wen ZK, Wu ZX, Wu HD, Lv LX, Yan CZ, Liu CH, Wang ZQ, Zheng C. Amino Acid Metabolism-Related lncRNA Signature Predicts the Prognosis of Breast Cancer. Front Genet 2022; 13:880387. [PMID: 35646057 PMCID: PMC9136175 DOI: 10.3389/fgene.2022.880387] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/25/2022] [Indexed: 12/16/2022] Open
Abstract
Background and Purpose: Breast cancer (BRCA) is the most frequent female malignancy and is potentially life threatening. The amino acid metabolism (AAM) has been shown to be strongly associated with the development and progression of human malignancies. In turn, long noncoding RNAs (lncRNAs) exert an important influence on the regulation of metabolism. Therefore, we attempted to build an AAM-related lncRNA prognostic model for BRCA and illustrate its immune characteristics and molecular mechanism. Experimental Design: The RNA-seq data for BRCA from the TCGA-BRCA datasets were stochastically split into training and validation cohorts at a 3:1 ratio, to construct and validate the model, respectively. The amino acid metabolism-related genes were obtained from the Molecular Signature Database. A univariate Cox analysis, least absolute shrinkage and selection operator (LASSO) regression, and a multivariate Cox analysis were applied to create a predictive risk signature. Subsequently, the immune and molecular characteristics and the benefits of chemotherapeutic drugs in the high-risk and low-risk subgroups were examined. Results: The prognostic model was developed based on the lncRNA group including LIPE-AS1, AC124067.4, LINC01655, AP005131.3, AC015802.3, USP30-AS1, SNHG26, and AL589765.4. Low-risk patients had a more favorable overall survival than did high-risk patients, in accordance with the results obtained for the validation cohort and the complete TCGA cohort. The elaborate results illustrated that a low-risk index was correlated with DNA-repair–associated pathways; a low TP53 and PIK3CA mutation rate; high infiltration of CD4+ T cells, CD8+ T cells, and M1 macrophages; active immunity; and less-aggressive phenotypes. In contrast, a high-risk index was correlated with cancer and metastasis-related pathways; a high PIK3CA and TP53 mutation rate; high infiltration of M0 macrophages, fibroblasts, and M2 macrophages; inhibition of the immune response; and more invasive phenotypes. Conclusion: In conclusion, we attempted to shed light on the importance of AAM-associated lncRNAs in BRCA. The prognostic model built here might be acknowledged as an indispensable reference for predicting the outcome of patients with BRCA and help identify immune and molecular characteristics.
Collapse
Affiliation(s)
- Yin-wei Dai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-kai Wen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi-xuan Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hao-dong Wu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin-xi Lv
- Wenzhou Medical University, Wenzhou, China
| | - Cong-zhi Yan
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cong-hui Liu
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zi-qiong Wang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chen Zheng
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Chen Zheng,
| |
Collapse
|
21
|
Sorto P, Mäyränpää MI, Saksi J, Nuotio K, Ijäs P, Tuimala J, Vikatmaa P, Soinne L, Kovanen PT, Lindsberg PJ. Glutamine synthetase in human carotid plaque macrophages associates with features of plaque vulnerability: An immunohistological study. Atherosclerosis 2022; 352:18-26. [DOI: 10.1016/j.atherosclerosis.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
|
22
|
Baranovicova E, Hnilicova P, Kalenska D, Kaplan P, Kovalska M, Tatarkova Z, Tomascova A, Lehotsky J. Metabolic Changes Induced by Cerebral Ischemia, the Effect of Ischemic Preconditioning, and Hyperhomocysteinemia. Biomolecules 2022; 12:554. [PMID: 35454143 PMCID: PMC9032340 DOI: 10.3390/biom12040554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Abstract
1H Nuclear Magnetic Resonance (NMR) metabolomics is one of the fundamental tools in the fast-developing metabolomics field. It identifies and quantifies the most abundant metabolites, alterations of which can describe energy metabolism, activated immune response, protein synthesis and catabolism, neurotransmission, and many other factors. This paper summarizes our results of the 1H NMR metabolomics approach to characterize the distribution of relevant metabolites and their alterations induced by cerebral ischemic injury or its combination with hyperhomocysteinemia in the affected tissue and blood plasma in rodents. A decrease in the neurotransmitter pool in the brain tissue likely follows the disordered feasibility of post-ischemic neurotransmission. This decline is balanced by the increased tissue glutamine level with the detected impact on neuronal health. The ischemic injury was also manifested in the metabolomic alterations in blood plasma with the decreased levels of glycolytic intermediates, as well as a post-ischemically induced ketosis-like state with increased plasma ketone bodies. As the 3-hydroxybutyrate can act as a likely neuroprotectant, its post-ischemic increase can suggest its supporting role in balancing ischemic metabolic dysregulation. Furthermore, the 1H NMR approach revealed post-ischemically increased 3-hydroxybutyrate in the remote organs, such as the liver and heart, as well as decreased myocardial glutamate. Ischemic preconditioning, as a proposed protective strategy, was manifested in a lower extent of metabolomic changes and/or their faster recovery in a longitudinal study. The paper also summarizes the pre- and post-ischemic metabolomic changes in the rat hyperhomocysteinemic models. Animals are challenged with hyperglycemia and ketosis-like state. A decrease in several amino acids in plasma follows the onset and progression of hippocampal neuropathology when combined with ischemic injury. The 1H NMR metabolomics approach also offers a high potential for metabolites in discriminatory analysis in the search for potential biomarkers of ischemic injury. Based on our results and the literature data, this paper presents valuable findings applicable in clinical studies and suggests the precaution of a high protein diet, especially foods which are high in Met content and low in B vitamins, in the possible risk of human cerebrovascular neuropathology.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (E.B.); (P.H.); (A.T.)
| | - Petra Hnilicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (E.B.); (P.H.); (A.T.)
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.K.); (Z.T.)
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.K.); (Z.T.)
| | - Anna Tomascova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (E.B.); (P.H.); (A.T.)
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.K.); (Z.T.)
| |
Collapse
|
23
|
Zhu D, Wu S, Li Y, Zhang Y, Chen J, Ma J, Cao L, Lyu Z, Hou T. Ferroptosis-related gene SLC1A5 is a novel prognostic biomarker and correlates with immune infiltrates in stomach adenocarcinoma. Cancer Cell Int 2022; 22:124. [PMID: 35305616 PMCID: PMC8933927 DOI: 10.1186/s12935-022-02544-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is associated with high morbidity and mortality rates. Ferroptosis is an iron-dependent form of cell death, which plays an important role in the development of many cancers. Tumor-associated competing endogenous RNAs (ceRNAs) regulate tumorigenesis and development. Our study aimed to construct ceRNA networks and explore the relationship between ferroptosis-related genes in the ceRNA network and immune infiltration in STAD. METHODS Based on the interactions among long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs), a ceRNA network was constructed to illustrate the relationships among lncRNAs, miRNAs, and mRNAs. Subsequently, gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) functional enrichment analyses were carried out to explore the functions and interactions of the differentially expressed (DE) mRNAs related to the ceRNA network. Differential expression and prognostic analysis of ferroptosis-related genes in the ceRNA network were performed using the R package "limma" and "survminer." The correlation between ferroptosis-related genes and tumor-infiltrating immune cells was analyzed using Spearman correlation analysis and CIBERSORT. Quantitative real-time PCR (qRT-PCR) was used to validate the expression of ferroptosis-related genes in STAD cells lines. RESULTS A ceRNA network consisting of 29 DElncRNAs, 31 DEmiRNAs, and 182 DEmRNAs was constructed. These DEmRNAs were significantly enriched in pathways related to the occurrence and development of STAD. The ferroptosis-related gene SLC1A5 was upregulated in STAD (P < 0.001) and was associated with better prognosis (P = 0.049). The CIBERSORT database and Spearman correlation analysis indicated that SLC1A5 was correlated with eight types of tumor-infiltrating immune cells and immune checkpoints, including PD-L1(CD-274) and PD-1(PDCD1). The SLC1A5 mRNA was found to be highly expressed in STAD cells lines. CONCLUSIONS Our study provides insights into the function of ceRNAs in STAD and identifies biomarkers for the development of therapies for STAD. The ferroptosis-related gene SLC1A5 in the ceRNA network was associated with both tumor-infiltrating immune cells and immune checkpoints in the tumor microenvironment, suggesting that SLC1A5 may be a novel prognostic marker and a potential target for STAD immunotherapy in the future.
Collapse
Affiliation(s)
- Dandan Zhu
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Guangdong Clinical Laboratory Center, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Sifan Wu
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Yafang Li
- Guangdong Clinical Laboratory Center, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Yu Zhang
- Medical Department, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Jierong Chen
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Jianhong Ma
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Lixue Cao
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| | - Zejian Lyu
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| | - Tieying Hou
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
- Guangdong Clinical Laboratory Center, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Medical Department, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
24
|
Singh D, Khan MA, Siddique HR. Specific targeting of cancer stem cells by immunotherapy: A possible stratagem to restrain cancer recurrence and metastasis. Biochem Pharmacol 2022; 198:114955. [PMID: 35181312 DOI: 10.1016/j.bcp.2022.114955] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs), the tumor-initiating cells playing a crucial role in cancer progression, recurrence, and metastasis, have the intrinsic property of self-renewal and therapy resistance. The tumorigenic properties of these cells include generation of cellular heterogeneity and immuno-suppressive tumor microenvironment (TME), conferring them the capability to resist a variety of anti-cancer therapeutics. Further, CSCs possess several unique immunological properties that help them escape recognition by the innate and adaptive immune system and shape a TME into a pro-tumorigenic and immunosuppressive landscape. In this context, immunotherapy is considered one of the best therapeutic options for eliminating CSCs to halt cancer recurrence and metastasis. In this review, we discuss the various immunomodulatory properties of CSCs and the interaction of CSCs with the immune system enabling immune evasion. In addition, we also highlight the present research update on immunotherapeutic targeting of CSCs and the possible further scope of research on this topic. We believe that a deeper understanding of CSCs' immunological properties and the crosstalk between CSCs and the immune system can develop better innovative immune-therapeutics and enhance the efficacy of current therapy-resistant cancer treatments.
Collapse
Affiliation(s)
- Deepti Singh
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammad Afsar Khan
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
25
|
Svetikienė M, Trybė D, Strioga M, Veželienė J, Isajevas V, Malickaitė R, Jurgauskienė L, Ringaitienė D, Šerpytis M, Šipylaitė J. Impact of Immunonutrition on T Cell Activation: A Randomized Control Study in Cardiac Surgery Patients. Acta Med Litu 2021; 28:240-252. [PMID: 35637935 PMCID: PMC9133614 DOI: 10.15388/amed.2021.28.2.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/01/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022] Open
Abstract
Background. Cardiac surgery provokes an intense inflammatory response that can cause an immunosuppressive state and adverse postoperative outcomes. We recently showed that postoperative immunonutrition with glutamine in “fragile” low-risk cardiac surgery patients was associated with a significantly increased level of CD3+ and CD4+ T cells. In order to clarify the biological relevance and clinical importance of these findings, we investigated whether an increase in the CD4+ T cell level was caused by changes in the systemic inflammatory response (caused by surgery or infection) and if it was associated with their activation status. Methods. A randomized control study of low operative risk but “fragile” cardiac surgery patients was performed. Patients were randomized into immunonutrition (IN) and control groups (C). The IN group received normal daily meals plus special immune nutrients for 5 days postoperatively, while the C group received only normal daily meals. Laboratory parameters were investigated before surgery and on the sixth postoperative day and the groups were compared accordingly. The expression of the CD69+ marker was investigated to determine T cell activation status. Serum concentrations of cytokines (interleukin-10 (IL-10), tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6)) and C-reactive protein (CRP) were determined to assess the systemic inflammatory response, while procalcitonin (PCT) levels were evaluated to confirm or deny possible bacterial infection. Results. Fifty-five patients were enrolled in the study. Twenty-seven (49.1%) were randomized in the IN group. Results show that on the sixth postoperative day, the CD4+CD69+ and CD8+CD69+ counts did not differ between the IN and C groups, accordingly 0.25 [0.16–0.50] vs 0.22 [0.13-0.41], p=0.578 and 0.13 [0.06–0.3] vs 0.09 [0.05–0.14], p=0.178. Also, statistically significant differences were not observed in the cytokine levels (IN and C groups: TNF-α 8.13 [7.32–10.31] vs 8.78 [7.65–11.2], p=0.300; IL-6 14.65 [9.28–18.95] vs 12.25 [8.55–22.50], p=0.786; IL-10 5.0 [5.0–5.0] vs 5.0 [5.0–5.0], p=0.343 respectively), which imply that an elevated T cell count is not associated with the systemic inflammatory response. Also, PCT (IN and C groups: 0.03 [0.01–0.09] vs 0.05 [0.03–0.08], p=0.352) and CRP (IN and C groups 62.7 [34.2–106.0] vs 63.7 [32.9–91.0], p=0.840) levels did not differ between the two groups. Moreover, low levels of PCT indicated that the increase in T cell count was not determined by bacterial infection. Conclusions. Our findings showed that CD4+ T cell levels were associated with neither the systemic inflammatory response nor bacterial infection. Secondly, increases in T cells are not accompanied by their activation status. These results suggest a hypothesis that a higher postoperative T cell concentration may be associated with postoperative immunonutrition in low-risk cardiac surgery patients with intact cellular vitality, i.e. “fragile”. However, immunonutrition alone did not affect T cell activation status.
Collapse
|
26
|
Atila A, Alay H, Yaman ME, Akman TC, Cadirci E, Bayrak B, Celik S, Atila NE, Yaganoglu AM, Kadioglu Y, Halıcı Z, Parlak E, Bayraktutan Z. The serum amino acid profile in COVID-19. Amino Acids 2021; 53:1569-1588. [PMID: 34605988 PMCID: PMC8487804 DOI: 10.1007/s00726-021-03081-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
The pandemic of the coronavirus disease (COVID-19) caused by SARS-CoV-2 affects millions of people worldwide. There are still many unknown aspects to this infection which affects the whole world. In addition, the potential impacts caused by this infection are still unclear. Amino acid metabolism, in particular, contains significant clues in terms of the development and prevention of many diseases. Therefore, this study aimed to compare amino acid profile of COVID-19 and healthy subject. In this study, the amino acid profiles of patients with asymptomatic, mild, moderate, and severe/critical SARS-CoV-2 infection were scanned with LC–MS/MS. The amino acid profile encompassing 30 amino acids in 142 people including 30 control and 112 COVID-19 patients was examined. 20 amino acids showed significant differences when compared to the control group in COVID-19 patient groups with different levels of severity in the statistical analyses conducted. It was detected that the branched-chain amino acids (BCAAs) changed in correlation with one another, and l-2-aminobutyric acid and l-phenylalanine had biomarker potential for COVID-19. Moreover, it was concluded that l-2-aminobutyric acid could provide prognostic information about the course of the disease. We believe that a new viewpoint will develop regarding the diagnosis, treatment, and prognosis as a result of the evaluation of the serum amino acid profiles of COVID-19 patients. Determining l-phenylalanine and l-2-aminobutyric levels can be used in laboratories as a COVID-19-biomarker. Also, supplementing COVID patients with taurine and BCAAs can be beneficial for treatment protocols.
Collapse
Affiliation(s)
- Alptug Atila
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Handan Alay
- Department of Infectious Diseases and Clinical Microbiology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Mehmet Emrah Yaman
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Tugrul Cagri Akman
- Department of Analytical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, 25240 Erzurum, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Burak Bayrak
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Saffet Celik
- Technology Research and Development Application and Research Center, Trakya University, 22030 Edirne, Turkey
| | - Nihal Efe Atila
- Department of Otorhinolaryngology, Erzurum Regional Training and Research Hospital, 25240 Erzurum, Turkey
| | - Aycan Mutlu Yaganoglu
- Department of Animal Science, College of Agriculture, Ataturk University, 25240 Erzurum, Turkey
| | - Yucel Kadioglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Ataturk University, 25240 Erzurum, Turkey
| | - Zekai Halıcı
- Department of Pharmacology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Emine Parlak
- Department of Infectious Diseases and Clinical Microbiology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Zafer Bayraktutan
- Department of Biochemistry, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| |
Collapse
|
27
|
Harnessing Metabolic Reprogramming to Improve Cancer Immunotherapy. Int J Mol Sci 2021; 22:ijms221910268. [PMID: 34638609 PMCID: PMC8508898 DOI: 10.3390/ijms221910268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 02/06/2023] Open
Abstract
Immune escape is one of the hallmarks of cancer. While metabolic reprogramming provides survival advantage to tumor cancer cells, accumulating data also suggest such metabolic rewiring directly affects the activation, differentiation and function of immune cells, particularly in the tumor microenvironment. Understanding how metabolic reprogramming affects both tumor and immune cells, as well as their interplay, is therefore critical to better modulate tumor immune microenvironment in the era of cancer immunotherapy. In this review, we discuss alterations in several essential metabolic pathways in both tumor and key immune cells, provide evidence on their dynamic interaction, and propose innovative strategies to improve cancer immunotherapy via the modulation of metabolic pathways.
Collapse
|
28
|
Purkerson JM, Corley JL, Schwartz GJ. Metabolic acidosis exacerbates pyelonephritis in mice prone to vesicoureteral reflux. Physiol Rep 2021; 8:e14525. [PMID: 33030238 PMCID: PMC7543054 DOI: 10.14814/phy2.14525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022] Open
Abstract
Acute pyelonephritis is a common, serious bacterial infection in children. The prevalence of acute pyelonephritis is due at least in part to vesicoureteral reflux (VUR). Although an association between abnormalities in electrolyte and acid–base balance and pyelonephritis is common in young children, the impact of metabolic acidosis (MA) on progression of acute pyelonephritis is not fully understood. In this study, the effect of MA on pyelonephritis was studied in C3H mouse strains prone to VUR. MA induced by ammonium chloride supplementation in food specifically impaired clearance of urinary tract infection with uropathogenic Escherichia. coli (UPEC‐UTI) in innate immune competent C3H strains (HeOuJ, HeN), whereas kidney UPEC burden in Tlr‐4‐deficient HeJ mice was unaffected. Antibody‐mediated depletion of myeloid cells (monocytes, neutrophil) markedly increased UPEC burden in the bladder and kidney confirming the pivotal role of neutrophils and tissue‐resident macrophages in clearance of UPEC‐UTI. MA concurrent with UPEC‐UTI markedly increased expression of cytokine (TNFα, IL‐1β, IL‐6) and chemokine (CXCL 1, 2, and 5) mRNA in isolated kidney CD cells and kidney neutrophil infiltrates were increased four‐ to fivefold compared to normal, UPEC‐infected mice. Thus, MA intensified pyelonephritis and increased the risk of kidney injury by impairing clearance of UPEC‐UTI and potentiating renal inflammation characterized by an elevated kidney neutrophil infiltrate.
Collapse
Affiliation(s)
- Jeffrey M Purkerson
- Pediatric Nephrology, University of Rochester Medical Center, Rochester, NY, USA.,Strong Children's Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Janine L Corley
- Pediatric Nephrology, University of Rochester Medical Center, Rochester, NY, USA.,Strong Children's Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - George J Schwartz
- Pediatric Nephrology, University of Rochester Medical Center, Rochester, NY, USA.,Strong Children's Research Center, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
29
|
Horst EA, Kvidera SK, Baumgard LH. Invited review: The influence of immune activation on transition cow health and performance-A critical evaluation of traditional dogmas. J Dairy Sci 2021; 104:8380-8410. [PMID: 34053763 DOI: 10.3168/jds.2021-20330] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
The progression from gestation into lactation represents the transition period, and it is accompanied by marked physiological, metabolic, and inflammatory adjustments. The entire lactation and a cow's opportunity to have an additional lactation are heavily dependent on how successfully she adapts during the periparturient period. Additionally, a disproportionate amount of health care and culling occurs early following parturition. Thus, lactation maladaptation has been a heavily researched area of dairy science for more than 50 yr. It was traditionally thought that excessive adipose tissue mobilization in large part dictated transition period success. Further, the magnitude of hypocalcemia has also been assumed to partly control whether a cow effectively navigates the first few months of lactation. The canon became that adipose tissue released nonesterified fatty acids (NEFA) and the resulting hepatic-derived ketones coupled with hypocalcemia lead to immune suppression, which is responsible for transition disorders (e.g., mastitis, metritis, retained placenta, poor fertility). In other words, the dogma evolved that these metabolites and hypocalcemia were causal to transition cow problems and that large efforts should be enlisted to prevent increased NEFA, hyperketonemia, and subclinical hypocalcemia. However, despite intensive academic and industry focus, the periparturient period remains a large hurdle to animal welfare, farm profitability, and dairy sustainability. Thus, it stands to reason that there are alternative explanations to periparturient failures. Recently, it has become firmly established that immune activation and the ipso facto inflammatory response are a normal component of transition cow biology. The origin of immune activation likely stems from the mammary gland, tissue trauma during parturition, and the gastrointestinal tract. If inflammation becomes pathological, it reduces feed intake and causes hypocalcemia. Our tenet is that immune system utilization of glucose and its induction of hypophagia are responsible for the extensive increase in NEFA and ketones, and this explains why they (and the severity of hypocalcemia) are correlated with poor health, production, and reproduction outcomes. In this review, we argue that changes in circulating NEFA, ketones, and calcium are simply reflective of either (1) normal homeorhetic adjustments that healthy, high-producing cows use to prioritize milk synthesis or (2) the consequence of immune activation and its sequelae.
Collapse
Affiliation(s)
- E A Horst
- Department of Animal Science, Iowa State University, Ames 50011
| | - S K Kvidera
- Department of Animal Science, Iowa State University, Ames 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames 50011.
| |
Collapse
|
30
|
Recomendaciones para la terapia nutricional de pacientes con COVID-19. ACTA COLOMBIANA DE CUIDADO INTENSIVO 2021. [PMCID: PMC7843081 DOI: 10.1016/j.acci.2021.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Host cell glutamine metabolism as a potential antiviral target. Clin Sci (Lond) 2021; 135:305-325. [PMID: 33480424 DOI: 10.1042/cs20201042] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022]
Abstract
A virus minimally contains a nucleic acid genome packaged by a protein coat. The genome and capsid together are known as the nucleocapsid, which has an envelope containing a lipid bilayer (mainly phospholipids) originating from host cell membranes. The viral envelope has transmembrane proteins that are usually glycoproteins. The proteins in the envelope bind to host cell receptors, promoting membrane fusion and viral entry into the cell. Virus-infected host cells exhibit marked increases in glutamine utilization and metabolism. Glutamine metabolism generates ATP and precursors for the synthesis of macromolecules to assemble progeny viruses. Some compounds derived from glutamine are used in the synthesis of purines and pyrimidines. These latter compounds are precursors for the synthesis of nucleotides. Inhibitors of glutamine transport and metabolism are potential candidate antiviral drugs. Glutamine is also an essential nutrient for the functions of leukocytes (lymphocyte, macrophage, and neutrophil), including those in virus-infected patients. The increased glutamine requirement for immune cell functions occurs concomitantly with the high glutamine utilization by host cells in virus-infected patients. The development of antiviral drugs that target glutamine metabolism must then be specifically directed at virus-infected host cells to avoid negative effects on immune functions. Therefore, the aim of this review was to describe the landscape of cellular glutamine metabolism to search for potential candidates to inhibit glutamine transport or glutamine metabolism.
Collapse
|
32
|
TGF-β in Cancer: Metabolic Driver of the Tolerogenic Crosstalk in the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13030401. [PMID: 33499083 PMCID: PMC7865468 DOI: 10.3390/cancers13030401] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Overcoming tumor immunosuppression still represents one ambitious achievement for cancer immunotherapy. Of note, the cytokine TGF-β contributes to immune evasion in multiple cancer types, by feeding the establishment of a tolerogenic environment in the host. Indeed, it fosters the expansion and accumulation of immunosuppressive regulatory cell populations within the tumor microenvironment (TME), where it also activates resident stromal cells and enhances angiogenesis programs. More recently, TGF-β has also turned out as a key metabolic adjuster in tumors orchestrating metabolic pathways in the TME. In this review, we will scrutinize TGF-β-mediated immune and stromal cell crosstalk within the TME, with a primary focus on metabolic programs.
Collapse
|
33
|
The metabolic importance of the glutaminase II pathway in normal and cancerous cells. Anal Biochem 2020; 644:114083. [PMID: 33352190 DOI: 10.1016/j.ab.2020.114083] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023]
Abstract
In rapidly dividing cells, including many cancer cells, l-glutamine is a major energy source. Utilization of glutamine is usually depicted as: l-glutamine → l-glutamate (catalyzed by glutaminase isozymes; GLS1 and GLS2), followed by l-glutamate → α-ketoglutarate [catalyzed by glutamate-linked aminotransferases or by glutamate dehydrogenase (GDH)]. α-Ketoglutarate is a major anaplerotic component of the tricarboxylic acid (TCA) cycle. However, the glutaminase II pathway also converts l-glutamine to α-ketoglutarate. This pathway consists of a glutamine transaminase coupled to ω-amidase [Net reaction: l-Glutamine + α-keto acid + H2O → α-ketoglutarate + l-amino acid + NH4+]. This review focuses on the biological importance of the glutaminase II pathway, especially in relation to metabolism of cancer cells. Our studies suggest a component enzyme of the glutaminase II pathway, ω-amidase, is utilized by tumor cells to provide anaplerotic carbon. Inhibitors of GLS1 are currently in clinical trials as anti-cancer agents. However, this treatment will not prevent the glutaminase II pathway from providing anaplerotic carbon derived from glutamine. Specific inhibitors of ω-amidase, perhaps in combination with a GLS1 inhibitor, may provide greater therapeutic efficacy.
Collapse
|
34
|
Zhang X, Zink F, Hezel F, Vogt J, Wachter U, Wepler M, Loconte M, Kranz C, Hellmann A, Mizaikoff B, Radermacher P, Hartmann C. Metabolic substrate utilization in stress-induced immune cells. Intensive Care Med Exp 2020; 8:28. [PMID: 33336295 PMCID: PMC7746792 DOI: 10.1186/s40635-020-00316-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 06/11/2020] [Indexed: 12/21/2022] Open
Abstract
Immune cell activation leads to the acquisition of new functions, such as proliferation, chemotaxis, and cytokine production. These functional changes require continuous metabolic adaption in order to sustain ATP homeostasis for sufficient host defense. The bioenergetic demands are usually met by the interconnected metabolic pathways glycolysis, TCA cycle, and oxidative phosphorylation. Apart from glucose, other sources, such as fatty acids and glutamine, are able to fuel the TCA cycle.Rising evidence has shown that cellular metabolism has a direct effect on the regulation of immune cell functions. Thus, quiescent immune cells maintain a basal metabolic state, which shifts to an accelerated metabolic level upon immune cell activation in order to promote key effector functions.This review article summarizes distinct metabolic signatures of key immune cell subsets from quiescence to activation and demonstrates a methodical concept of how to assess cellular metabolic pathways. It further discusses why metabolic functions are of rising interest for translational research and how they can be affected by the underlying pathophysiological condition and/or therapeutic interventions.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Fabian Zink
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Felix Hezel
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Josef Vogt
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Ulrich Wachter
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Martin Wepler
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum Ulm, Ulm, Germany
| | - Maurizio Loconte
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncolocy and Neuroscience, Genoa, Italy
| | - Christine Kranz
- Institut für Analytische und Bioanalytische Chemie, Universität Ulm, Ulm, Germany
| | - Andreas Hellmann
- Institut für Analytische und Bioanalytische Chemie, Universität Ulm, Ulm, Germany
| | - Boris Mizaikoff
- Institut für Analytische und Bioanalytische Chemie, Universität Ulm, Ulm, Germany
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Clair Hartmann
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum Ulm, Ulm, Germany
| |
Collapse
|
35
|
Mezzetti M, Bionaz M, Trevisi E. Interaction between inflammation and metabolism in periparturient dairy cows. J Anim Sci 2020; 98:S155-S174. [PMID: 32810244 DOI: 10.1093/jas/skaa134] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Matteo Mezzetti
- Department of Animal Sciences, Food and Nutrition (DIANA), Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR
| | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition (DIANA), Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, Piacenza, Italy
| |
Collapse
|
36
|
Coleman DN, Lopreiato V, Alharthi A, Loor JJ. Amino acids and the regulation of oxidative stress and immune function in dairy cattle. J Anim Sci 2020; 98:S175-S193. [PMID: 32810243 PMCID: PMC7433927 DOI: 10.1093/jas/skaa138] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Vincenzo Lopreiato
- Department of Health Science, Interdepartmental Services Centre of Veterinary for Human and Animal Health, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Abdulrahman Alharthi
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Juan J Loor
- Department of Animal Sciences, University of Illinois, Urbana, IL.,Division of Nutritional Sciences, University of Illinois, Urbana, IL
| |
Collapse
|
37
|
Kelly B, Pearce EL. Amino Assets: How Amino Acids Support Immunity. Cell Metab 2020; 32:154-175. [PMID: 32649859 DOI: 10.1016/j.cmet.2020.06.010] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/06/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022]
Abstract
Amino acids are fundamental building blocks supporting life. Their role in protein synthesis is well defined, but they contribute to a host of other intracellular metabolic pathways, including ATP generation, nucleotide synthesis, and redox balance, to support cellular and organismal function. Immune cells critically depend on such pathways to acquire energy and biomass and to reprogram their metabolism upon activation to support growth, proliferation, and effector functions. Amino acid metabolism plays a key role in this metabolic rewiring, and it supports various immune cell functions beyond increased protein synthesis. Here, we review the mechanisms by which amino acid metabolism promotes immune cell function, and how these processes could be targeted to improve immunity in pathological conditions.
Collapse
Affiliation(s)
- Beth Kelly
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Erika L Pearce
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.
| |
Collapse
|
38
|
Abstract
Immune checkpoint therapies aiming to enhance T cell responses have revolutionized cancer immunotherapy. However, although a small fraction of patients develops durable anti-tumor responses, the majority of patients display only transient responses, underlying the need for finding auxiliary approaches. Tumor microenvironment poses a major metabolic barrier to efficient anti-tumor T cell activity. As it is now well accepted that metabolism regulates T cell fate and function, harnessing metabolism may be a new strategy to potentiate T cell-based immunotherapies.
Collapse
|
39
|
Cassim S, Pouyssegur J. Tumor Microenvironment: A Metabolic Player that Shapes the Immune Response. Int J Mol Sci 2019; 21:E157. [PMID: 31881671 PMCID: PMC6982275 DOI: 10.3390/ijms21010157] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
Immune cells survey and patrol throughout the body and sometimes take residence in niche environments with distinct cellular subtypes and nutrients that may fluctuate from those in which they matured. Rooted in immune cell physiology are metabolic pathways and metabolites that not only deliver substrates and energy for growth and survival, but also instruct effector functions and cell differentiation. Unlike cancer cells, immune cells are not subject to a "Darwinian evolutionary pressure" that would allow them to adapt to developing tumors but are often irrevocably affected to local nutrient deprivation. Thus, immune cells must metabolically adapt to these changing conditions in order to perform their necessary functions. On the other hand, there is now a growing appreciation that metabolic changes occurring in cancer cells can impact on immune cell functionality and contribute to tumor immune evasion, and as such, there is a considerable and growing interest in developing techniques that target metabolism for immunotherapy. In this review, we discuss the metabolic plasticity displayed by innate and adaptive immune cells and highlight how tumor-derived lactate and tumor acidity restrict immunity. To our knowledge, this review outlines the most recent insights on how tumor microenvironment metabolically instructs immune responsiveness.
Collapse
Affiliation(s)
- Shamir Cassim
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
| | - Jacques Pouyssegur
- Department of Medical Biology, Centre Scientifique de Monaco, CSM, 98000 Monaco, Monaco;
- University Côte d’Azur, IRCAN, CNRS, Centre A. Lacassagne, 06189 Nice, France
| |
Collapse
|
40
|
Causes and Consequences of A Glutamine Induced Normoxic HIF1 Activity for the Tumor Metabolism. Int J Mol Sci 2019; 20:ijms20194742. [PMID: 31554283 PMCID: PMC6802203 DOI: 10.3390/ijms20194742] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/01/2019] [Accepted: 09/15/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factor hypoxia-inducible factor 1 (HIF1) is the crucial regulator of genes that are involved in metabolism under hypoxic conditions, but information regarding the transcriptional activity of HIF1 in normoxic metabolism is limited. Different tumor cells were treated under normoxic and hypoxic conditions with various drugs that affect cellular metabolism. HIF1α was silenced by siRNA in normoxic/hypoxic tumor cells, before RNA sequencing and bioinformatics analyses were performed while using the breast cancer cell line MDA-MB-231 as a model. Differentially expressed genes were further analyzed and validated by qPCR, while the activity of the metabolites was determined by enzyme assays. Under normoxic conditions, HIF1 activity was significantly increased by (i) glutamine metabolism, which was associated with the release of ammonium, and it was decreased by (ii) acetylation via acetyl CoA synthetase (ACSS2) or ATP citrate lyase (ACLY), respectively, and (iii) the presence of L-ascorbic acid, citrate, or acetyl-CoA. Interestingly, acetylsalicylic acid, ibuprofen, L-ascorbic acid, and citrate each significantly destabilized HIF1α only under normoxia. The results from the deep sequence analyses indicated that, in HIF1-siRNA silenced MDA-MB-231 cells, 231 genes under normoxia and 1384 genes under hypoxia were transcriptionally significant deregulated in a HIF1-dependent manner. Focusing on glycolysis genes, it was confirmed that HIF1 significantly regulated six normoxic and 16 hypoxic glycolysis-associated gene transcripts. However, the results from the targeted metabolome analyses revealed that HIF1 activity affected neither the consumption of glucose nor the release of ammonium or lactate; however, it significantly inhibited the release of the amino acid alanine. This study comprehensively investigated, for the first time, how normoxic HIF1 is stabilized, and it analyzed the possible function of normoxic HIF1 in the transcriptome and metabolic processes of tumor cells in a breast cancer cell model. Furthermore, these data imply that HIF1 compensates for the metabolic outcomes of glutaminolysis and, subsequently, the Warburg effect might be a direct consequence of the altered amino acid metabolism in tumor cells.
Collapse
|
41
|
Wnt Signaling in Cancer Metabolism and Immunity. Cancers (Basel) 2019; 11:cancers11070904. [PMID: 31261718 PMCID: PMC6678221 DOI: 10.3390/cancers11070904] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/22/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022] Open
Abstract
The Wingless (Wnt)/β-catenin pathway has long been associated with tumorigenesis, tumor plasticity, and tumor-initiating cells called cancer stem cells (CSCs). Wnt signaling has recently been implicated in the metabolic reprogramming of cancer cells. Aberrant Wnt signaling is considered to be a driver of metabolic alterations of glycolysis, glutaminolysis, and lipogenesis, processes essential to the survival of bulk and CSC populations. Over the past decade, the Wnt pathway has also been shown to regulate the tumor microenvironment (TME) and anti-cancer immunity. Wnt ligands released by tumor cells in the TME facilitate the immune evasion of cancer cells and hamper immunotherapy. In this review, we illustrate the role of the canonical Wnt/β-catenin pathway in cancer metabolism and immunity to explore the potential therapeutic approach of targeting Wnt signaling from a metabolic and immunological perspective.
Collapse
|
42
|
Hu X, Deng J, Yu T, Chen S, Ge Y, Zhou Z, Guo Y, Ying H, Zhai Q, Chen Y, Yuan F, Niu Y, Shu W, Chen H, Ma C, Liu Z, Guo F. ATF4 Deficiency Promotes Intestinal Inflammation in Mice by Reducing Uptake of Glutamine and Expression of Antimicrobial Peptides. Gastroenterology 2019; 156:1098-1111. [PMID: 30452920 DOI: 10.1053/j.gastro.2018.11.033] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Activating transcription factor 4 (ATF4) regulates genes involved in the inflammatory response, amino acid metabolism, autophagy, and endoplasmic reticulum stress. We investigated whether its activity is altered in patients with inflammatory bowel diseases (IBDs) and mice with enterocolitis. METHODS We obtained biopsy samples during endoscopy from inflamed and/or uninflamed regions of the colon from 21 patients with active Crohn's disease (CD), 22 patients with active ulcerative colitis (UC), and 38 control individuals without IBD and of the ileum from 19 patients with active CD and 8 individuals without IBD in China. Mice with disruption of Atf4 specifically in intestinal epithelial cells (Atf4ΔIEC mice) and Atf4-floxed mice (controls) were given dextran sodium sulfate (DSS) to induce colitis. Some mice were given injections of recombinant defensin α1 (DEFA1) and supplementation of l-alanyl-glutamine or glutamine in drinking water. Human and mouse ileal and colon tissues were analyzed by quantitative real-time polymerase chain reaction, immunoblots, and immunohistochemistry. Serum and intestinal epithelial cell (IEC) amino acids were measured by high-performance liquid chromatography-tandem mass spectrometry. Levels of ATF4 were knocked down in IEC-18 cells with small interfering RNAs. Microbiomes were analyzed in ileal feces from mice by using 16S ribosomal DNA sequencing. RESULTS Levels of ATF4 were significantly decreased in inflamed intestinal mucosa from patients with active CD or active UC compared with those from uninflamed regions or intestinal mucosa from control individuals. ATF4 was also decreased in colonic epithelia from mice with colitis vs mice without colitis. Atf4ΔIEC mice developed spontaneous enterocolitis and colitis of greater severity than control mice after administration of DSS. Atf4ΔIEC mice had decreased serum levels of glutamine and reduced levels of antimicrobial peptides, such as Defa1, Defa4, Defa5, Camp, and Lyz1, in ileal Paneth cells. Atf4ΔIEC mice had alterations in ileal microbiomes compared with control mice; these changes were reversed by administration of glutamine. Injections of DEFA1 reduced the severity of spontaneous enteritis and DSS-induced colitis in Atf4ΔIEC mice. We found that expression of solute carrier family 1 member 5 (SLC1A5), a glutamine transporter, was directly regulated by ATF4 in cell lines. Overexpression of SLC1A5 in IEC-18 or primary IEC cells increased glutamine uptake and expression of antimicrobial peptides. Knockdown of ATF4 in IEC-18 cells increased expression of inflammatory cytokines, whereas overexpression of SLC1A5 in the knockdown cells reduced cytokine expression. Levels of SLC1A5 were decreased in inflamed intestinal mucosa of patients with CD and UC and correlated with levels of ATF4. CONCLUSIONS Levels of ATF4 are decreased in inflamed intestinal mucosa from patients with active CD or UC. In mice, ATF4 deficiency reduces glutamine uptake by intestinal epithelial cells and expression of antimicrobial peptides by decreasing transcription of Slc1a5. ATF4 might therefore be a target for the treatment of IBD.
Collapse
Affiliation(s)
- Xiaoming Hu
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiali Deng
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tianming Yu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Shanghai Chen
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yadong Ge
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Ziheng Zhou
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yajie Guo
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hao Ying
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Chen
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feixiang Yuan
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuguo Niu
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weigang Shu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Huimin Chen
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Caiyun Ma
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| | - Feifan Guo
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
43
|
Glutamine Addiction and Therapeutic Strategies in Lung Cancer. Int J Mol Sci 2019; 20:ijms20020252. [PMID: 30634602 PMCID: PMC6359540 DOI: 10.3390/ijms20020252] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/05/2019] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
Lung cancer cells are well-documented to rewire their metabolism and energy production networks to support rapid survival and proliferation. This metabolic reorganization has been recognized as a hallmark of cancer. The increased uptake of glucose and the increased activity of the glycolytic pathway have been extensively described. However, over the past years, increasing evidence has shown that lung cancer cells also require glutamine to fulfill their metabolic needs. As a nitrogen source, glutamine contributes directly (or indirectly upon conversion to glutamate) to many anabolic processes in cancer, such as the biosynthesis of amino acids, nucleobases, and hexosamines. It plays also an important role in the redox homeostasis, and last but not least, upon conversion to α-ketoglutarate, glutamine is an energy and anaplerotic carbon source that replenishes tricarboxylic acid cycle intermediates. The latter is generally indicated as glutaminolysis. In this review, we explore the role of glutamine metabolism in lung cancer. Because lung cancer is the leading cause of cancer death with limited curative treatment options, we focus on the potential therapeutic approaches targeting the glutamine metabolism in cancer.
Collapse
|
44
|
Carvalho PLPF, Yamamoto FY, Barros MM, Gatlin Iii DM. l-glutamine in vitro supplementation enhances Nile tilapia Oreochromis niloticus (Linnaeus, 1758) leukocyte function. FISH & SHELLFISH IMMUNOLOGY 2018; 80:592-599. [PMID: 29960065 DOI: 10.1016/j.fsi.2018.06.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/22/2018] [Accepted: 06/26/2018] [Indexed: 06/08/2023]
Abstract
Under appropriate conditions, glutamine (Gln) is an essential nutrient for immunological responses, acting as a metabolic substrate for proliferation of enterocytes and lymphocytes, and having positive effects on the function of stimulated immune cells. Thus, specific components of both innate and adaptive immune systems of Nile tilapia were evaluated after supplementing Gln to cell culture media. Primary cell cultures of kidney leukocytes were used for respiratory burst and phagocytic activity assessment. The ability of macrophages to kill Streptococcus iniae also was evaluated. Additionally, a proliferation assay was conducted with peripheral blood lymphocytes (PBL) exposed to non-specific mitogens. Results showed that macrophage phagocytosis, anion superoxide production, and bactericidal capacity were significantly (P < 0.05) enhanced by Gln supplementation to the culture media. The proliferation of lymphocytes upon mitogenic exposure also was significantly (P < 0.05) enhanced by Gln supplementation to the media. Our results suggest that in vitro, different levels of Gln were necessary for optimal immunological responses of leukocytes and lymphocytes. As such, Gln supplementation was able to enhance and modulate both innate and adaptive responses of Nile tilapia leukocytes, highlighting its potential application as an immunonutrient.
Collapse
Affiliation(s)
- Pedro L P F Carvalho
- Faculdade de Medicina Veterinária e Zootecnia, Departamento de Melhoramento e Nutrição Animal, Universidade Estadual Paulista "Júlio de Mesquita Filho", São Paulo, Brazil; Department of Wildlife and Fisheries Sciences, Texas A&M University System, College Station, TX, 77843-2258, USA.
| | - Fernando Y Yamamoto
- Department of Wildlife and Fisheries Sciences, Texas A&M University System, College Station, TX, 77843-2258, USA.
| | - Margarida M Barros
- Faculdade de Medicina Veterinária e Zootecnia, Departamento de Melhoramento e Nutrição Animal, Universidade Estadual Paulista "Júlio de Mesquita Filho", São Paulo, Brazil.
| | - Delbert M Gatlin Iii
- Department of Wildlife and Fisheries Sciences, Texas A&M University System, College Station, TX, 77843-2258, USA.
| |
Collapse
|
45
|
Ratter JM, Tack CJ, Netea MG, Stienstra R. Environmental Signals Influencing Myeloid Cell Metabolism and Function in Diabetes. Trends Endocrinol Metab 2018; 29:468-480. [PMID: 29789206 DOI: 10.1016/j.tem.2018.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022]
Abstract
The environment induces metabolic reprogramming of immune cells via specific signaling pathways. Recent studies have revealed that changes in cell metabolism affect key immune cell functions including cytokine production and migration. In diabetes, these functions are either insufficiently or excessively activated, translating into diabetes-associated complications, including increased susceptibility to infection and accelerated cardiovascular disease. Diabetes alters the abundance of environmental signals, including glucose, insulin, and lipids. Subsequently, changes in environmental signals drive metabolic reprogramming, impair immune cell function, and ultimately contribute to diabetes-associated complications. We review here recent studies on changes in innate immune cell metabolism, especially in myeloid cells, that are driven by environmental signals relevant to diabetes, and discuss therapeutic perspectives of targeting metabolism of immune cells in diabetes.
Collapse
Affiliation(s)
- Jacqueline M Ratter
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
46
|
Porter L, Toepfner N, Bashant KR, Guck J, Ashcroft M, Farahi N, Chilvers ER. Metabolic Profiling of Human Eosinophils. Front Immunol 2018; 9:1404. [PMID: 30013547 PMCID: PMC6036296 DOI: 10.3389/fimmu.2018.01404] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/06/2018] [Indexed: 12/18/2022] Open
Abstract
Immune cells face constant changes in their microenvironment, which requires rapid metabolic adaptation. In contrast to neutrophils, which are known to rely near exclusively on glycolysis, the metabolic profile of human eosinophils has not been characterized. Here, we assess the key metabolic parameters of peripheral blood-derived human eosinophils using real-time extracellular flux analysis to measure extracellular acidification rate and oxygen consumption rate, and compare these parameters to human neutrophils. Using this methodology, we demonstrate that eosinophils and neutrophils have a similar glycolytic capacity, albeit with a minimal glycolytic reserve. However, compared to neutrophils, eosinophils exhibit significantly greater basal mitochondrial respiration, ATP-linked respiration, maximum respiratory capacity, and spare respiratory capacity. Of note, the glucose oxidation pathway is also utilized by eosinophils, something not evident in neutrophils. Furthermore, using a colorimetric enzymatic assay, we show that eosinophils have much reduced glycogen stores compared to neutrophils. We also show that physiologically relevant levels of hypoxia (PO2 3 kPa), by suppressing oxygen consumption rates, have a profound effect on basal and phorbol-myristate-acetate-stimulated eosinophil and neutrophil metabolism. Finally, we compared the metabolic profile of eosinophils purified from atopic and non-atopic subjects and show that, despite a difference in the activation status of eosinophils derived from atopic subjects, these cells exhibit comparable oxygen consumption rates upon priming with IL-5 and stimulation with fMLP. In summary, our findings show that eosinophils display far greater metabolic flexibility compared to neutrophils, with the potential to use glycolysis, glucose oxidation, and oxidative phosphorylation. This flexibility may allow eosinophils to adapt better to diverse roles in host defense, homeostasis, and immunomodulation.
Collapse
Affiliation(s)
- Linsey Porter
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Nicole Toepfner
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Kathleen R Bashant
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Margaret Ashcroft
- Division of Renal Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Neda Farahi
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Edwin R Chilvers
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| |
Collapse
|
47
|
Abstract
Systemic sclerosis (SSc) is an autoimmune disease of unknown aetiology characterized by vascular lesions, immunological alterations and diffuse fibrosis of the skin and internal organs. Since recent evidence suggests that there is a link between metabolomics and immune mediated disease, serum metabolic profile of SSc patients and healthy controls was investigated by 1H-NMR and GC-MS techniques. The results indicated a lower level of aspartate, alanine, choline, glutamate, and glutarate in SSc patients compared with healthy controls. Moreover, comparing patients affected by limited SSc (lcSSc) and diffuse SSc (dcSSc), 6 discriminant metabolites were identified. The multivariate analysis performed using all the metabolites significantly different revealed glycolysis, gluconeogenesis, energetic pathways, glutamate metabolism, degradation of ketone bodies and pyruvate metabolism as the most important networks. Aspartate, alanine and citrate yielded a high area under receiver-operating characteristic (ROC) curves (AUC of 0.81; CI 0.726–0.93) for discriminating SSc patients from controls, whereas ROC curve generated with acetate, fructose, glutamate, glutamine, glycerol and glutarate (AUC of 0.84; CI 0.7–0.98) discriminated between lcSSc and dcSSc. These results indicated that serum NMR-based metabolomics profiling method is sensitive and specific enough to distinguish SSc from healthy controls and provided a feasible diagnostic tool for the diagnosis and classification of the disease.
Collapse
|
48
|
Wang Y, Liu Y, Cao Q, Shi X, Lu H, Gao S, Yang R. Metabolomic analysis for the protective effects of mangiferin on sepsis-induced lung injury in mice. Biomed Chromatogr 2018; 32:e4208. [PMID: 29431198 DOI: 10.1002/bmc.4208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/09/2018] [Accepted: 01/29/2018] [Indexed: 12/11/2022]
Abstract
This study aimed to investigate the efficacy of mangiferin, including its known antioxidant and anti-inflammatory effects on sepsis-induced lung injury induced by a classical cecal ligation and puncture (CLP) models in mouse using a metabolomics approach. A total of 24 mice were randomly divided into four groups: the sham group was given saline before sham operation. The CLP group received the CLP operation only. HMF and LMF groups were given mangiferin treatment of high dose and low dose of mangiferin, respectively, before the CLP operation. One week after treatment, the mice were sacrificed and their lungs were collected for metabolomics analysis. We developed ultra-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry to perform lung metabolic profiling analysis. With the methods of principal component analysis and partial least squares discriminant analysis, 58 potential metabolites associated with amino acid metabolism, purine metabolism, lipid metabolism and energy regulation were observed to be increased or reduced in HMF and LMF groups compared with the CLP group. Conclusively, our results suggest that mangiferin plays a protective role in the moderation of sepsis-induced lung injury through reducing oxidative stress, regulating lipid metabolism and energy biosynthesis.
Collapse
Affiliation(s)
- Yilin Wang
- Student Unit, Navy Medical University, Shanghai, China
| | - Yang Liu
- Student Unit, Navy Medical University, Shanghai, China
| | - Qiqi Cao
- Student Unit, Navy Medical University, Shanghai, China
| | - Xuan Shi
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongtao Lu
- Department of Navy Aeromedicine, Navy Medical University, Shanghai, China
| | - Songyan Gao
- School of Pharmacy, Navy Medical University, Shanghai, China
| | - Rui Yang
- Department of Anesthesiology, Changzheng Hospital, Navy Medical University, Shanghai, China
| |
Collapse
|
49
|
C de Oliveira D, Santos EW, Nogueira-Pedro A, Xavier JG, Borelli P, Fock RA. Effects of short-term dietary restriction and glutamine supplementation in vitro on the modulation of inflammatory properties. Nutrition 2018; 48:96-104. [PMID: 29469028 DOI: 10.1016/j.nut.2017.11.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 10/21/2017] [Accepted: 11/04/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Dietary restriction (DR) is a nutritional intervention that exerts profound effects on biochemical and immunologic parameters, modulating some inflammatory properties. Glutamine (GLN) is a conditionally essential amino acid that can modulate inflammatory properties. However, there is a lack of data evaluating the effects of DR and GLN supplementation, especially in relation to inflammatory cytokine production and the expression of transcription factors such as nuclear factor (NF)-κB. METHODS We subjected 3-mo-old male Balb/c mice to DR by reducing their food intake by 30%. DR animals lost weight and showed reduced levels of serum triacylglycerols, glucose, cholesterol, and calcium as well as a reduction in bone density. Additionally, blood, peritoneal, and spleen cellularity were reduced, lowering the number of peritoneal F4/80- and CD86-positive cells and the total number of splenic CD4- and CD8-positive cells. RESULTS The production of interleukin (IL)-10 and the expression of NF-κB in splenic cells were not affected by DR or by GLN supplementation. However, peritoneal macrophages from DR animals showed reduced IL-12 and tumor necrosis factor-α production and increased IL-10 production with reduced phosphorylation of NF-κB expression. Additionally, GLN was able to modulate cytokine production by peritoneal cells from the control group, although no effects were observed in cells from the DR group. CONCLUSION DR induces biochemical and immunologic changes, in particular by reducing IL-12 and tumor necrosis factor-α production by macrophages and clearly upregulating IL-10 production, whereas GLN supplementation did not modify these parameters in cells from DR animals.
Collapse
Affiliation(s)
- Dalila C de Oliveira
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Ed Wilson Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Amanda Nogueira-Pedro
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - José Guilherme Xavier
- School of Veterinary Medicine, Institute of Health Science Paulista University, Sao Paulo, Brazil
| | - Primavera Borelli
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
50
|
A past and present overview of macrophage metabolism and functional outcomes. Clin Sci (Lond) 2017; 131:1329-1342. [PMID: 28592702 DOI: 10.1042/cs20170220] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 03/16/2017] [Accepted: 03/22/2017] [Indexed: 12/11/2022]
Abstract
In 1986 and 1987, Philip Newsholme et al. reported macrophages utilize glutamine, as well as glucose, at high rates. These authors measured key enzyme activities and consumption and production levels of metabolites in incubated or cultured macrophages isolated from the mouse or rat intraperitoneal cavity. Metabolic pathways essential for macrophage function were then determined. Macrophages utilize glucose to generate (i) ATP in the pathways of glycolysis and mitochondrial oxidative phosphorylation, (ii) glycerol 3-phosphate for the synthesis of phospholipids and triacylglycerols, (iii) NADPH for the production of reactive oxygen species (ROS) and (iv) ribose for the synthesis of RNA and subsequently production and secretion of protein mediators (e.g. cytokines). Glutamine plays an essential role in macrophage metabolism and function, as it is required for energy production but also provides nitrogen for synthesis of purines, pyrimidines and thus RNA. Macrophages also utilize fatty acids for both energy production in the mitochondria and lipid synthesis essential to plasma membrane turnover and lipid meditator production. Recent studies utilizing metabolomic approaches, transcriptional and metabolite tracking technologies have detailed mitochondrial release of tricarboxylic acid (TCA) intermediates (e.g. citrate and succinate) to the cytosol, which then regulate pro-inflammatory responses. Macrophages can reprogramme their metabolism and function according to environmental conditions and stimuli in order to polarize phenotype so generating pro- or anti-inflammatory cells. Changes in macrophage metabolism result in modified function/phenotype and vice versa. The plasticity of macrophage metabolism allows the cell to quickly respond to changes in environmental conditions such as those induced by hormones and/or inflammation. A past and present overview of macrophage metabolism and impact of endocrine regulation and the relevance to human disease are described in this review.
Collapse
|