1
|
Bohnacker S, Henkel FDR, Hartung F, Geerlof A, Riemer S, Prodjinotho UF, Salah EB, Mourão ASD, Bohn S, Teder T, Thomas D, Gurke R, Boeckel C, Ud-Dean M, König AC, Quaranta A, Alessandrini F, Lechner A, Spitzlberger B, Kabat AM, Pearce E, Haeggström JZ, Hauck SM, Wheelock CE, Jakobsson PJ, Sattler M, Voehringer D, Feige MJ, da Costa CP, Esser-von Bieren J. A helminth enzyme subverts macrophage-mediated immunity by epigenetic targeting of prostaglandin synthesis. Sci Immunol 2024; 9:eadl1467. [PMID: 39642243 DOI: 10.1126/sciimmunol.adl1467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/13/2024] [Indexed: 12/08/2024]
Abstract
The molecular mechanisms by which worm parasites evade host immunity are incompletely understood. In a mouse model of intestinal helminth infection using Heligmosomoides polygyrus bakeri (Hpb), we show that helminthic glutamate dehydrogenase (heGDH) drives parasite chronicity by suppressing macrophage-mediated host defense. Combining RNA-seq, ChIP-seq, and targeted lipidomics, we identify prostaglandin E2 (PGE2) as a major immune regulatory mechanism of heGDH. The induction of PGE2 and other immunoregulatory factors, including IL-12 family cytokines and indoleamine 2,3-dioxygenase 1, by heGDH required p300-mediated histone acetylation, whereas the enzyme's catalytic activity suppressed the synthesis of type 2-promoting leukotrienes by macrophages via 2-hydroxyglutarate. By contrast, the induction of immunoregulatory factors involved the heGDH N terminus by potentially mediating interactions with cellular targets (CD64 and GPNMB) identified by proteomics. Type 2 cytokines counteracted suppressive effects of heGDH on host defense, indicating that type 2 immunity can limit helminth-driven immune evasion. Thus, helminths harness a ubiquitous metabolic enzyme to epigenetically target type 2 macrophage activation and establish chronicity.
Collapse
Affiliation(s)
- Sina Bohnacker
- Department of Immunobiology, Université de Lausanne, Epalinges, Switzerland
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Fiona D R Henkel
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Franziska Hartung
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Arie Geerlof
- Protein Expression and Purification Facility (PEPF), Institute of Structural Biology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Sandra Riemer
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Ulrich F Prodjinotho
- Institute for Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
- Center for Global Health, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Eya Ben Salah
- Department of Immunobiology, Université de Lausanne, Epalinges, Switzerland
| | - André Santos Dias Mourão
- Protein Expression and Purification Facility (PEPF), Institute of Structural Biology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefan Bohn
- Department of CryoEM Technology, Max Planck Institute of Biochemistry, Martinsried, Germany
- Cryo-Electron Microscopy Platform and Institute of Structural Biology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Tarvi Teder
- Department of Medical Biochemistry and Biophysics, Division of Chemistry II, Karolinska Institutet, Stockholm, Sweden
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP) and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt am Main, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP) and Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt am Main, Germany
| | - Christiane Boeckel
- Institute of Computational Biology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Minhaz Ud-Dean
- Institute of Computational Biology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ann-Christine König
- Metabolomics and Proteomics Core, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Alessandro Quaranta
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Francesca Alessandrini
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Antonie Lechner
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Benedikt Spitzlberger
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Agnieszka M Kabat
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Edward Pearce
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Jesper Z Haeggström
- Department of Medical Biochemistry and Biophysics, Division of Chemistry II, Karolinska Institutet, Stockholm, Sweden
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Department of Medicine, Division of Rheumatology, Karolinska Institutet and Karolinska University Hospital at Solna, Stockholm, Sweden
| | - Michael Sattler
- Cryo-Electron Microscopy Platform and Institute of Structural Biology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Bavarian NMR-Center, Department Chemie, Technische Universität München, Garching, Germany
| | - David Voehringer
- Infektionsbiologische Abteilung, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Germany
| | - Matthias J Feige
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Clarissa Prazeres da Costa
- Institute for Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
- Center for Global Health, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Julia Esser-von Bieren
- Department of Immunobiology, Université de Lausanne, Epalinges, Switzerland
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| |
Collapse
|
2
|
Chen H, Huang S, Yao S, Wang J, Huang J, Yu Z. Multi-omics analyses of Bacillus amyloliquefaciens treated mice infected with Schistosoma japonicum reveal dynamics change of intestinal microbiome and its associations with host metabolism. PLoS Negl Trop Dis 2024; 18:e0012583. [PMID: 39466852 PMCID: PMC11515987 DOI: 10.1371/journal.pntd.0012583] [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: 04/25/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Schistosomiasis japonica is a serious threat to human health. It causes damage to the intestine and liver. Probiotic therapy has been shown to be effective in alleviating intestinal diseases and improving host health. Previous studies have found that Bacillus amyloliquefaciens could alleviate the pathological symptoms of schistosomiasis japonica, but the regulatory mechanism of alleviating schistosomiasis japonica is still unknown. PRINCIPAL FINDINGS This study analyzed the dynamic changes of intestinal microbiome in mice infected with Schistosoma japonicum after the intervention of B. amyloliquefaciens and its connection to host metabolism by multi-omics sequencing technology. B. amyloliquefaciens was found to significantly regulate the homeostasis of intestinal microbiota by promoting the growth of beneficial bacteria and inhibiting potential pathogenic bacteria and protect the number of core microbes. Meanwhile, the genes related to the metabolism of glycerophospholipids and amino acid from intestinal microbiome changed significantly, and were shown to be significantly positively correlated with the associated metabolites of microbial origin. Moreover, host metabolism (lipid metabolism and steroid hormone biosynthesis) was also found to be significantly regulated. CONCLUSIONS The recovery of intestinal microbial homeostasis and the regulation of host metabolism revealed the potential probiotic properties of B. amyloliquefaciens, which also provided new ideas for the prevention and adjuvant treatment of schistosomiasis japonica.
Collapse
Affiliation(s)
- Hao Chen
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Shuaiqin Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Siqi Yao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jingyan Wang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Zheng Yu
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Chiodi D, Ishihara Y. The role of the methoxy group in approved drugs. Eur J Med Chem 2024; 273:116364. [PMID: 38781921 DOI: 10.1016/j.ejmech.2024.116364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 05/25/2024]
Abstract
The methoxy substituent is prevalent in natural products and, consequently, is present in many natural product-derived drugs. It has also been installed in modern drug molecules with no remnant of natural product features because medicinal chemists have been taking advantage of the benefits that this small functional group can bestow on ligand-target binding, physicochemical properties, and ADME parameters. Herein, over 230 methoxy-containing small-molecule drugs, as well as several fluoromethoxy-containing drugs, are presented from the vantage point of the methoxy group. Biochemical mechanisms of action, medicinal chemistry SAR studies, and numerous X-ray cocrystal structures are analyzed to identify the precise role of the methoxy group for many of the drugs and drug classes. Although the methoxy substituent can be considered as the hybridization of a hydroxy and a methyl group, the combination of these functionalities often results in unique effects that can amount to more than the sum of the individual parts.
Collapse
Affiliation(s)
- Debora Chiodi
- Department of Chemistry, Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, CA, 92121, USA
| | - Yoshihiro Ishihara
- Department of Chemistry, Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
4
|
Lu G, Gao D, Jiang W, Yu X, Tong J, Liu X, Qiao T, Wang R, Zhang M, Wang S, Yang J, Li D, Lv Z. Disrupted gut microecology after high-dose 131I therapy and radioprotective effects of arachidonic acid supplementation. Eur J Nucl Med Mol Imaging 2024; 51:2395-2408. [PMID: 38561516 PMCID: PMC11178657 DOI: 10.1007/s00259-024-06688-9] [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: 01/23/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Despite the potential radiotoxicity in differentiated thyroid cancer (DTC) patients with high-dose 131I therapy, the alterations and regulatory mechanisms dependent on intestinal microecology remain poorly understood. We aimed to identify the characteristics of the gut microbiota and metabolites in DTC patients suffering from high-dose 131I therapy and explore the radioprotective mechanisms underlying arachidonic acid (ARA) treatment. METHODS A total of 102 patients with DTC were recruited, with fecal samples collected before and after 131I therapy for microbiome and untargeted and targeted metabolomic analyses. Mice were exposed to total body irradiation with ARA replenishment and antibiotic pretreatment and were subjected to metagenomic, metabolomic, and proteomic analyses. RESULTS 131I therapy significantly changed the structure of gut microbiota and metabolite composition in patients with DTC. Lachnospiraceae were the most dominant bacteria after 131I treatment, and metabolites with decreased levels and pathways related to ARA and linoleic acid were observed. In an irradiation mouse model, ARA supplementation not only improved quality of life and recovered hematopoietic and gastrointestinal systems but also ameliorated oxidative stress and inflammation and preserved enteric microecology composition. Additionally, antibiotic intervention eliminated the radioprotective effects of ARA. Proteomic analysis and ursolic acid pretreatment showed that ARA therapy greatly influenced intestinal lipid metabolism in mice subjected to irradiation by upregulating the expression of hydroxy-3-methylglutaryl-coenzyme A synthase 1. CONCLUSION These findings highlight that ARA, as a key metabolite, substantially contributes to radioprotection. Our study provides novel insights into the pivotal role that the microbiota-metabolite axis plays in radionuclide protection and offers effective biological targets for treating radiation-induced adverse effects.
Collapse
Affiliation(s)
- Ganghua Lu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Dingwei Gao
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wen Jiang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaqing Yu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Junyu Tong
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaoyan Liu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Tingting Qiao
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Ru Wang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Mengyu Zhang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Shaoping Wang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Jianshe Yang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Dan Li
- Department of Nuclear Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510289, China.
| | - Zhongwei Lv
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
5
|
Hartung F, Haimerl P, Schindela S, Mussack V, Kirchner B, Henkel FDR, Bernhardt U, Zissler UM, Santarella-Mellwig R, Pfaffl M, Schmidt-Weber CB, Chaker AM, Esser-von Bieren J. Extracellular vesicle miRNAs drive aberrant macrophage responses in NSAID-exacerbated respiratory disease. Allergy 2024; 79:1893-1907. [PMID: 38573073 DOI: 10.1111/all.16117] [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: 06/29/2023] [Revised: 03/01/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) have been implicated in the pathogenesis of asthma, however, how EVs contribute to immune dysfunction and type 2 airway inflammation remains incompletely understood. We aimed to elucidate roles of airway EVs and their miRNA cargo in the pathogenesis of NSAID-exacerbated respiratory disease (N-ERD), a severe type 2 inflammatory condition. METHODS EVs were isolated from induced sputum or supernatants of cultured nasal polyp or turbinate tissues of N-ERD patients or healthy controls by size-exclusion chromatography and characterized by particle tracking, electron microscopy and miRNA sequencing. Functional effects of EV miRNAs on gene expression and mediator release by human macrophages or normal human bronchial epithelial cells (NHBEs) were studied by RNA sequencing, LC-MS/MS and multiplex cytokine assays. RESULTS EVs were highly abundant in secretions from the upper and lower airways of N-ERD patients. N-ERD airway EVs displayed profoundly altered immunostimulatory capacities and miRNA profiles compared to airway EVs of healthy individuals. Airway EVs of N-ERD patients, but not of healthy individuals induced inflammatory cytokine (GM-CSF and IL-8) production by NHBEs. In macrophages, N-ERD airway EVs exhibited an impaired potential to induce cytokine and prostanoid production, while enhancing M2 macrophage activation. Let-7 family miRNAs were highly enriched in sputum EVs from N-ERD patients and mimicked suppressive effects of N-ERD EVs on macrophage activation. CONCLUSION Aberrant airway EV miRNA profiles may contribute to immune dysfunction and chronic type 2 inflammation in N-ERD. Let-7 family miRNAs represent targets for correcting aberrant macrophage activation and mediator responses in N-ERD.
Collapse
Affiliation(s)
- Franziska Hartung
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Pascal Haimerl
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Sonja Schindela
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Veronika Mussack
- Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Fiona D R Henkel
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Ulrike Bernhardt
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | | | - Michael Pfaffl
- Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Adam M Chaker
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, TUM School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Julia Esser-von Bieren
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
6
|
O'Carroll SM, Henkel FDR, O'Neill LAJ. Metabolic regulation of type I interferon production. Immunol Rev 2024; 323:276-287. [PMID: 38465724 DOI: 10.1111/imr.13318] [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] [Indexed: 03/12/2024]
Abstract
Over the past decade, there has been a surge in discoveries of how metabolic pathways regulate immune cell function in health and disease, establishing the field of immunometabolism. Specifically, pathways such as glycolysis, the tricarboxylic acid (TCA) cycle, and those involving lipid metabolism have been implicated in regulating immune cell function. Viral infections cause immunometabolic changes which lead to antiviral immunity, but little is known about how metabolic changes regulate interferon responses. Interferons are critical cytokines in host defense, rapidly induced upon pathogen recognition, but are also involved in autoimmune diseases. This review summarizes how metabolic change impacts interferon production. We describe how glycolysis, lipid metabolism (specifically involving eicosanoids and cholesterol), and the TCA cycle-linked intermediates itaconate and fumarate impact type I interferons. Targeting these metabolic changes presents new therapeutic possibilities to modulate type I interferons during host defense or autoimmune disorders.
Collapse
Affiliation(s)
- Shane M O'Carroll
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Fiona D R Henkel
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
7
|
Garg PK, Guan W, Nomura S, Weir NL, Tintle N, Virtanen JK, Hirakawa Y, Qian F, Sun Q, Rimm E, Lemaitre RN, Jensen PN, Heckbert SR, Imamura F, Steur M, Leander K, Laguzzi F, Voortman T, Ninomiya T, Mozaffarian D, Harris WS, Siscovick DS, Tsai MY. n-6 fatty acid biomarkers and incident atrial fibrillation: an individual participant-level pooled analysis of 11 international prospective studies. Am J Clin Nutr 2023; 118:921-929. [PMID: 37769813 DOI: 10.1016/j.ajcnut.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 09/02/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND The presence of atrial fibrillation (AF) is associated with an over 2-fold increased risk of stroke, heart failure, and cardiovascular mortality. Long chain n-6 PUFAs have been suggested to have a variety of beneficial biologic effects that may reduce AF development; however, prior studies evaluating this relationship are limited. OBJECTIVES We prospectively evaluated the association between circulating levels of linoleic acid (LA) and arachidonic acid (AA) with incident AF. METHODS We used participant-level data from a global consortium of 11 prospective cohort studies with measurements of LA and AA in adults (aged ≥18 y). Participating studies conducted de novo analyses using a prespecified analytical plan with harmonized definitions for exposures, outcomes, covariates, and subgroups. Associations were pooled using inverse-variance weighted meta-analysis. RESULTS Among 41,335 participants, 6173 incident cases of AF were ascertained, with median follow-up time of 14 y. In multivariable analysis, per interquintile range (difference between the 10th and 90th percentiles for each fatty acid), circulating n-6 levels were not associated with incident AF. For LA, the hazard ratio per interquintile range was 0.96 (95% confidence interval [CI]: 0.89, 1.04), and for AA, 1.02 (95% CI: 0.94, 1.10), with little evidence of heterogeneity between cohorts. Associations were similarly nonsignificant across subgroups of age, race, and biomarker fraction. CONCLUSIONS Biomarkers of n-6 fatty acids including LA and AA are not associated with incident AF. These findings suggest that overall effects of n-6 PUFAs on influencing AF development are neutral.
Collapse
Affiliation(s)
- Parveen K Garg
- Division of Cardiology, University of Southern California Keck School of Medicine, Los Angeles, CA, United States
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, United States
| | - Sarah Nomura
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Natalie L Weir
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Nathan Tintle
- Department of Population Health Sciences, College of Nursing, University of Illinois-Chicago, Chicago, IL, United States
| | - Jyrki K Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Yoichiro Hirakawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Frank Qian
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Eric Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Paul N Jensen
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Marinka Steur
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Karin Leander
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Federica Laguzzi
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States; Division of Cardiology, Tufts Medical Center, Boston, MA, United States
| | - William S Harris
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, United States
| | | | - Michael Y Tsai
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
8
|
Huang Y, Wang Y, Su H, Wang H, Xu H, Xu C, Zhou F, Zhang Y. Association between polyunsaturated fatty acid intake and the prevalence of erectile dysfunction: A cross-sectional analysis of the NHANES 2001-2004. Lipids Health Dis 2023; 22:182. [PMID: 37880723 PMCID: PMC10601238 DOI: 10.1186/s12944-023-01950-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/18/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Polyunsaturated fatty acids (PUFAs) have demonstrated significant therapeutic potential across a wide range of disease. The aim of this study was to investigate the potential impact of PUFA intake on the prevalence of erectile dysfunction (ED). METHODS The study included a total of 3730 participants from the National Health and Nutrition Examination Survey (NHANES) 2001-2004. Univariate analysis, multivariate regression analysis, subgroup analysis and machine learning were utilized to explore the relationship of variables to ED. Dose response curves were constructed to observe the linear or nonlinear relationship between PUFA intake and the prevalence of ED. Propensity score matching (PSM) was used for sensitivity analysis. Finally, the potential mechanistic link between PUFA intake and ED was explored. RESULTS Through univariate and multivariate regression analysis results before and after PSM and XGBoost algorithm model results, arachidonic acid (AA) was chosen as the main research object. The consumption of AA was found to be associated with a decreased prevalence of ED under the fully adjusted model [OR = 0.33 (0.20, 0.56), P < 0.001]. The interaction between AA and education was found in the subgroup analysis. Dose-response curves indicated a linear negative correlation between AA intake and the prevalence of ED. The above results were confirmed in the data analysis after 1:1 PSM. In addition, AA intake was associated with a decrease in inflammatory biomarkers and homocysteine. CONCLUSIONS The results suggest that AA intake is negatively correlated with the prevalence of ED. Further, anti-inflammatory and anti-endothelial damage may play a role in this.
Collapse
Affiliation(s)
- Yong Huang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yingying Wang
- Department of Oncology, People Hospital of Jiulongpo District, Chongqing, 400050, China
| | - Huiyi Su
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Hexi Wang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haoyu Xu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengwei Xu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fulin Zhou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yao Zhang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
9
|
Skrzydlewska E, Łuczaj W, Biernacki M, Wójcik P, Jarocka-Karpowicz I, Orehovec B, Baršić B, Tarle M, Kmet M, Lukšić I, Marušić Z, Bauer G, Žarković N. Preliminary Comparison of Molecular Antioxidant and Inflammatory Mechanisms Determined in the Peripheral Blood Granulocytes of COVID-19 Patients. Int J Mol Sci 2023; 24:13574. [PMID: 37686388 PMCID: PMC10488240 DOI: 10.3390/ijms241713574] [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: 07/19/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
The aim of this study was to evaluate selected parameters of redox signaling and inflammation in the granulocytes of COVID-19 patients who recovered and those who died. Upon admission, the patients did not differ in terms of any relevant clinical parameter apart from the percentage of granulocytes, which was 6% higher on average in those patients who died. Granulocytes were isolated from the blood of 15 healthy people and survivors and 15 patients who died within a week, and who were selected post hoc for analysis according to their matching gender and age. They differed only in the lethal outcome, which could not be predicted upon arrival at the hospital. The proteins level (respective ELISA), antioxidant activity (spectrophotometry), and lipid mediators (UPUPLC-MS) were measured in the peripheral blood granulocytes obtained via gradient centrifugation. The levels of Nrf2, HO-1, NFκB, and IL-6 were higher in the granulocytes of COVID-19 patients who died within a week, while the activity of cytoplasmic Cu,Zn-SOD and mitochondrial Mn-SOD and IL-2/IL-10 were lower in comparison to the levels observed in survivors. Furthermore, in the granulocytes of those patients who died, an increase in pro-inflammatory eicosanoids (PGE2 and TXB2), together with elevated cannabinoid receptors 1 and 2 (associated with a decrease in the anti-inflammatory 15d-PGJ2), were found. Hence, this study suggests that by triggering transcription factors, granulocytes activate inflammatory and redox signaling, leading to the production of pro-inflammatory eicosanoids while reducing cellular antioxidant capacity through SOD, thus expressing an altered response to COVID-19, which may result in the onset of systemic oxidative stress, ARDS, and the death of the patient.
Collapse
Affiliation(s)
- Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Michał Biernacki
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Piotr Wójcik
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Iwona Jarocka-Karpowicz
- Department of Analytical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.Ł.); (M.B.); (P.W.); (I.J.-K.)
| | - Biserka Orehovec
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Bruno Baršić
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Marko Tarle
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Marta Kmet
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
| | - Ivica Lukšić
- Clinical Hospital Dubrava, HR-10000 Zagreb, Croatia; (B.O.); (B.B.); (M.T.); (M.K.); (I.L.)
- School of Medicine, University of Zagreb, HR-10000 Zagreb, Croatia
| | - Zlatko Marušić
- Division of Pathology, Clinical Hospital Centre Zagreb, HR-10000 Zagreb, Croatia;
| | - Georg Bauer
- Institute of Virology, Medical Center–University of Freiburg, 79104 Freiburg, Germany;
| | - Neven Žarković
- Laboratory for Oxidative Stress (LabOS), Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| |
Collapse
|
10
|
Villain E, Chanson A, Mainka M, Kampschulte N, Le Faouder P, Bertrand-Michel J, Brandolini-Bulon M, Charbit B, Musvosvi M, Bilek N, Scriba TJ, Quintana-Murci L, Schebb NH, Duffy D, Gladine C. Integrated analysis of whole blood oxylipin and cytokine responses after bacterial, viral, and T cell stimulation reveals new immune networks. iScience 2023; 26:107422. [PMID: 37575177 PMCID: PMC10415927 DOI: 10.1016/j.isci.2023.107422] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/24/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Oxylipins are major immunomodulating mediators, yet studies of inflammation focus mainly on cytokines. Here, using a standardized whole-blood stimulation system, we characterized the oxylipin-driven inflammatory responses to various stimuli and their relationships with cytokine responses. We performed a pilot study in 25 healthy individuals using 6 different stimuli: 2 bacterial stimuli (LPS and live BCG), 2 viral stimuli (vaccine-grade poly I:C and live H1N1 attenuated influenza), an enterotoxin superantigen and a Null control. All stimuli induced a strong production of oxylipins but most importantly, bacterial, viral, and T cell immune responses show distinct oxylipin signatures. Integration of the oxylipin and cytokine responses for each condition revealed new immune networks improving our understanding of inflammation regulation. Finally, the oxylipin responses and oxylipin-cytokine networks were compared in patients with active tuberculosis or with latent infection. This revealed different responses to BCG but not LPS stimulation highlighting new regulatory pathways for further investigations.
Collapse
Affiliation(s)
- Etienne Villain
- Institut Pasteur, Université Paris Cité, Translational Immunology Unit, Paris, France
| | - Aurélie Chanson
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Malwina Mainka
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nadja Kampschulte
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Pauline Le Faouder
- MetaToul, MetaboHUB, Inserm/UPS UMR 1048-I2MC, Institut des Maladies Métaboliques et Cardiovasculaires, 31400 Toulouse, France
| | - Justine Bertrand-Michel
- MetaToul, MetaboHUB, Inserm/UPS UMR 1048-I2MC, Institut des Maladies Métaboliques et Cardiovasculaires, 31400 Toulouse, France
| | - Marion Brandolini-Bulon
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
- Université Clermont Auvergne, INRAE, UNH, Plateforme D’Exploration Du Métabolisme, MetaboHUB Clermont, Clermont-Ferrand, France
| | - Bruno Charbit
- Institut Pasteur, Université Paris Cité, CBUTechS, Paris, France
| | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Lluis Quintana-Murci
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
- Collège de France, 75005 Paris, France
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Darragh Duffy
- Institut Pasteur, Université Paris Cité, Translational Immunology Unit, Paris, France
- Institut Pasteur, Université Paris Cité, CBUTechS, Paris, France
| | - Cécile Gladine
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| |
Collapse
|
11
|
Park S, Cathey AL, Hao W, Zeng L, Pennathur S, Aung MT, Rosario-Pabón Z, Vélez-Vega CM, Cordero JF, Alshawabkeh A, Watkins DJ, Meeker JD. Associations of phthalates, phthalate replacements, and their mixtures with eicosanoid biomarkers during pregnancy. ENVIRONMENT INTERNATIONAL 2023; 178:108101. [PMID: 37487376 PMCID: PMC10733973 DOI: 10.1016/j.envint.2023.108101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/25/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Humans are exposed to complex mixtures of phthalates. Gestational exposure to phthalates has been linked to preeclampsia and preterm birth through potential pathways such as endocrine disruption, oxidative stress, and inflammation. Eicosanoids are bioactive signaling lipids that are related to a variety of homeostatic and inflammatory processes. We investigated associations between urinary phthalates and their mixtures with plasma eicosanoid levels during pregnancy using the PROTECT cohort in Puerto Rico (N = 655). After adjusting for covariates, we estimated pair-wise associations between the geometric mean of individual phthalate metabolite concentrations across pregnancy and eicosanoid biomarkers using multivariable linear regression. We used bootstrapping of adaptive elastic net regression (adENET) to evaluate phthalate mixtures associated with eicosanoids and subsequently create environmental risk scores (ERS) to represent weighted sums of phthalate exposure for each individual. After adjusting for false-discovery, in single-pollutant analysis, 14 of 20 phthalate metabolites or parent compound indices showed significant and primarily negative associations with multiple eicosanoids. In our mixture analysis, associations with several metabolites of low molecular weight phthalates - DEP, DBP, and DIBP - became prominent. Additionally, MEHHTP and MECPTP, metabolites of a new phthalate replacement, DEHTP, were selected as important predictors for determining the concentrations of multiple eicosanoids from different pathway groups. A unit increase in phthalate ERS derived from bootstrapping of adENET was positively associated with several eicosanoids mainly from Cytochrome P450 pathway. For example, an increase in ERS was associated with 11(S)-HETE (β = 1.6, 95% CI: 0.020, 3.180), (±)11,12-DHET (β = 2.045, 95% CI: 0.250, 3.840), 20(S)-HETE (β = 0.813, 95% CI: 0.147, 1.479), and 9 s-HODE (β = 2.381, 95% CI: 0.657, 4.104). Gestational exposure to phthalates and phthalate mixtures were associated with eicosanoid levels during pregnancy. Results from the mixture analyses underscore the complexity of physiological impacts of phthalate exposure and call for further in-depth studies to examine these relationships.
Collapse
Affiliation(s)
- Seonyoung Park
- Department of Environmental Health Sciences, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Amber L Cathey
- Department of Environmental Health Sciences, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Wei Hao
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Lixia Zeng
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Subramaniam Pennathur
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Max T Aung
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Zaira Rosario-Pabón
- Graduate School of Public Health, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| | - Carmen M Vélez-Vega
- Graduate School of Public Health, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| | - José F Cordero
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, GA, USA
| | | | - Deborah J Watkins
- Department of Environmental Health Sciences, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - John D Meeker
- Department of Environmental Health Sciences, University of Michigan, School of Public Health, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Mehta H, Tasin I, Hackstein CP, Willberg C, Klenerman P. Prostaglandins differentially modulate mucosal-associated invariant T-cell activation and function according to stimulus. Immunol Cell Biol 2023; 101:262-272. [PMID: 36541521 PMCID: PMC10152717 DOI: 10.1111/imcb.12617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/29/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Mucosal-associated invariant T (MAIT) cells are an innate-like T-cell type conserved in many mammals and especially abundant in humans. Their semi-invariant T-cell receptor (TCR) recognizes the major histocompatibility complex-like molecule MR1 presenting riboflavin intermediates associated with microbial metabolism. Full MAIT cell triggering requires costimulation via cytokines, and the cells can also be effectively triggered in a TCR-independent manner by cytokines [e.g. interleukin (IL)-12 and IL-18 in combination]. Thus, triggering of MAIT cells is highly sensitive to local soluble mediators. Suppression of MAIT cell activation has not been well explored and could be very relevant to their roles in infection, inflammation and cancer. Prostaglandins (PG) are major local mediators of these microenvironments which can have regulatory roles for T cells. Here, we explored whether prostaglandins suppressed MAIT cell activation in response to TCR-dependent and TCR-independent signals. We found that protaglandin E2 (PGE2 ) and to a lesser extent protaglandin D2 (PGD2 ), but not leukotrienes, suppressed MAIT cell responses to Escherichia coli or TCR triggers. However, there was no impact on cytokine-induced triggering. The inhibition was blocked by targeting the signaling mediated via PG receptor 2 (PTGER2) and 4 (PTGER4) receptors in combination. These data indicate that prostaglandins can potentially modulate local MAIT cell functions in vivo and indicate distinct regulation of the TCR-dependent and TCR-independent pathways of MAIT cell activation.
Collapse
Affiliation(s)
- Hema Mehta
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
| | - Irene Tasin
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
| | | | - Christian Willberg
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
| | - Paul Klenerman
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
- NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
| |
Collapse
|
13
|
Cui S, Ye J. A protein-lipid complex that detoxifies free fatty acids. Bioessays 2023; 45:e2200210. [PMID: 36585363 PMCID: PMC9974861 DOI: 10.1002/bies.202200210] [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: 10/28/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023]
Abstract
Fatty acids (FAs) are well known to serve as substrates for reactions that provide cells with membranes and energy. In contrast to these metabolic reactions, the physiological importance of FAs themselves known as free FAs (FFAs) in cells remains obscure. Since accumulation of FFAs in cells is toxic, cells must develop mechanisms to detoxify FFAs. One such mechanism is to sequester free polyunsaturated FAs (PUFAs) into a droplet-like structure assembled by Fas-Associated Factor 1 (FAF1), a cytosolic protein. This sequestration limits access of PUFAs to Fe2+ , thereby preventing Fe2+ -catalyzed PUFA peroxidation. Consequently, assembly of the FAF1-FFA complex is critical to protect cells from ferroptosis, a cell death pathway triggered by PUFA peroxidation. The observations that free PUFAs in cytosol are not randomly diffused but rather sequestered into a membraneless complex should open new directions to explore signaling pathways by which FFAs regulate cellular physiology.
Collapse
Affiliation(s)
- Shaojie Cui
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jin Ye
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
14
|
Bagayoko S, Meunier E. Emerging roles of ferroptosis in infectious diseases. FEBS J 2022; 289:7869-7890. [PMID: 34670020 DOI: 10.1111/febs.16244] [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: 05/31/2021] [Revised: 09/06/2021] [Accepted: 10/20/2021] [Indexed: 01/14/2023]
Abstract
In living organisms, lipid peroxidation is a continuously occurring cellular process and therefore involved in various physiological and pathological contexts. Among the broad variety of lipids, polyunsaturated fatty acids (PUFA) constitute a major target of oxygenation either when released as mediators by phospholipases or when present in membranous phospholipids. The last decade has seen the characterization of an iron- and lipid peroxidation-dependent cell necrosis, namely, ferroptosis, that involves the accumulation of peroxidized PUFA-containing phospholipids. Further studies could link ferroptosis in a very large body of (physio)-pathological processes, including cancer, neurodegenerative, and metabolic diseases. In this review, we mostly focus on the emerging involvement of lipid peroxidation-driven ferroptosis in infectious diseases, and the immune consequences. We also discuss the putative ability of microbial virulence factors to exploit or to dampen ferroptosis regulatory pathways to their own benefit.
Collapse
Affiliation(s)
- Salimata Bagayoko
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, France
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, France
| |
Collapse
|
15
|
Attanzio A, Restivo I, Tutone M, Tesoriere L, Allegra M, Livrea MA. Redox Properties, Bioactivity and Health Effects of Indicaxanthin, a Bioavailable Phytochemical from Opuntia ficus indica, L.: A Critical Review of Accumulated Evidence and Perspectives. Antioxidants (Basel) 2022; 11:antiox11122364. [PMID: 36552572 PMCID: PMC9774763 DOI: 10.3390/antiox11122364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Phytochemicals from plant foods are considered essential to human health. Known for their role in the adaptation of plants to their environment, these compounds can induce adaptive responses in cells, many of which are directed at maintaining the redox tone. Indicaxanthin is a long-known betalain pigment found in the genus Opuntia of cactus pear and highly concentrated in the edible fruits of O. ficus indica, L. whose bioactivity has been overlooked until recently. This review summarizes studies conducted so far in vitro and in vivo, most of which have been performed in our laboratory. The chemical and physicochemical characteristics of Indicaxanthin are reflected in the molecule's reducing properties and antioxidant effects and help explain its ability to interact with membranes, modulate redox-regulated cellular pathways, and possibly bind to protein molecules. Measurement of bioavailability in volunteers has been key to exploring its bioactivity; amounts consistent with dietary intake, or plasma concentration after dietary consumption of cactus pear fruit, have been used in experimental setups mimicking physiological or pathophysiological conditions, in cells and in animals, finally suggesting pharmacological potential and relevance of Indicaxanthin as a nutraceutical. In reporting experimental results, this review also aimed to raise questions and seek insights for further basic research and health promotion applications.
Collapse
|
16
|
Welch BM, McNell EE, Edin ML, Ferguson KK. Inflammation and oxidative stress as mediators of the impacts of environmental exposures on human pregnancy: Evidence from oxylipins. Pharmacol Ther 2022; 239:108181. [PMID: 35367517 PMCID: PMC9525454 DOI: 10.1016/j.pharmthera.2022.108181] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023]
Abstract
Inflammation and oxidative stress play major roles in healthy and pathological pregnancy. Environmental exposure to chemical pollutants may adversely affect maternal and fetal health in pregnancy by dysregulating these critical underlying processes of inflammation and oxidative stress. Oxylipins are bioactive lipids that play a major role in regulating inflammation and increasing lines of evidence point towards an importance in pregnancy. The biosynthetic production of oxylipins requires oxygenation of polyunsaturated fatty acids, which can occur through several well-characterized enzymatic and nonenzymatic pathways. This review describes the state of the science of epidemiologic evidence on oxylipin production in pregnancy and its association with 1) key pregnancy outcomes and 2) environmental exposures. We searched PubMed for studies of pregnancy that measured one or more oxylipin analytes during pregnancy or delivery. We evaluated oxylipin associations with three categories of adverse pregnancy outcomes, including preeclampsia, preterm birth, and fetal growth restriction, along with several categories of environmental pollutants. The majority of studies evaluated one to two oxylipins, most of which focused on oxylipins produced from nonenzymatic processes of oxidative stress. However, an increasing number of recent studies have leveraged technological advancements to profile a large number of oxylipins produced from distinct biosynthetic pathways. Although the literature indicated robust evidence that oxylipins produced via nonenzymatic pathways are associated with pregnancy outcomes and environmental exposures, evidence for enzymatically produced oxylipins showed that associations may differ between biosynthetic pathways. Along with summarizing this evidence, we review promising therapeutic options to regulate oxylipin production and provide a set of recommendations for future epidemiologic studies in these research areas. Further evidence is needed to improve our understanding of how oxylipins may act as key biological mediators for the adverse effects of environmental pollutants on pregnancy outcomes.
Collapse
Affiliation(s)
- Barrett M Welch
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | - Erin E McNell
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Matthew L Edin
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Kelly K Ferguson
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
17
|
Deme P, Moniruzzaman M, Moore D, Heaton R, Ellis R, Letendre S, Haughey N. Association of Plasma Eicosanoid Levels With Immune, Viral, and Cognitive Outcomes in People With HIV. Neurology 2022; 99:e1251-e1264. [PMID: 35851253 PMCID: PMC9576290 DOI: 10.1212/wnl.0000000000200945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/20/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To determine whether plasma eicosanoid levels are associated with immune, viral, and cognitive outcomes in people with HIV (PWH). METHODS We measured 42 eicosanoids in a longitudinal study of 95 PWH and 25 demographically comparable uninfected participants. Routine clinical chemistry, virologic, immune markers, and a neuropsychological test battery assessing 7 cognitive domains were administered to all participants at 2 study visits over an average of 6.5 months. RESULTS Plasma eicosanoid concentrations were elevated in PWH (n = 95) compared with seronegative controls (n = 25) (100% prediction power at 5% false discovery rate [FDR], α = 0.0531) and were negatively associated with lower current and nadir CD4 lymphocyte counts. Higher levels of eicosanoids were associated with impairments in working memory, verbal fluency, and executive function. Higher plasma viral load was associated with elevated proinflammatory eicosanoids (24% prediction power at 5% FDR and 42.4% prediction power at 10% FDR, α = 0.10). Longitudinal analyses showed that eicosanoid levels were correlated with viral load and with plasma creatinine. Despite associations of eicosanoids with viral loads, elevated plasma eicosanoids were similar in virally suppressed and not fully suppressed PWH. DISCUSSION These data show that HIV infection is associated with a robust production of eicosanoids that are not substantially reduced by antiretroviral therapy (ART). The sustained elevation of these oxylipins in PWH despite ART may contribute to an accelerated aging phenotype that includes earlier than expected brain and peripheral organ damage.
Collapse
Affiliation(s)
- Pragney Deme
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mohammed Moniruzzaman
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - David Moore
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Robert Heaton
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ronald Ellis
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Scott Letendre
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Norman Haughey
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
18
|
Pérez MM, Pimentel VE, Fuzo CA, da Silva-Neto PV, Toro DM, Fraga-Silva TFC, Gardinassi LG, Oliveira CNS, Souza COS, Torre-Neto NT, de Carvalho JCS, De Leo TC, Nardini V, Feitosa MR, Parra RS, da Rocha JJR, Feres O, Vilar FC, Gaspar GG, Constant LF, Ostini FM, Degiovani AM, Amorim AP, Viana AL, Fernandes APM, Maruyama SR, Russo EMS, Santos IKFM, Bonato VLD, Cardoso CRB, Sorgi CA, Dias-Baruffi M, Faccioli LH. Acetylcholine, Fatty Acids, and Lipid Mediators Are Linked to COVID-19 Severity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:250-261. [PMID: 35768148 DOI: 10.4049/jimmunol.2200079] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
Abstract
Lipid and cholinergic mediators are inflammatory regulators, but their role in the immunopathology of COVID-19 is still unclear. Here, we used human blood and tracheal aspirate (TA) to investigate whether acetylcholine (Ach), fatty acids (FAs), and their derived lipid mediators (LMs) are associated with COVID-19 severity. First, we analyzed the perturbation profile induced by SARS-CoV-2 infection in the transcriptional profile of genes related to the ACh and FA/LM pathways. Blood and TA were used for metabolomic and lipidomic analyses and for quantification of leukocytes, cytokines, and ACh. Differential expression and coexpression gene network data revealed a unique transcriptional profile associated with ACh and FA/LM production, release, and cellular signaling. Transcriptomic data were corroborated by laboratory findings: SARS-CoV-2 infection increased plasma and TA levels of arachidonic acid, 5-hydroxy-6E,8Z,11Z,14Z-eicosatetraenoic acid, 11-hydroxy-5Z,8Z,12E,14Z-eicosatetraenoic acid, and ACh. TA samples also exhibited high levels of PGE2, thromboxane B2, 12-oxo-5Z,8Z,10E,14Z-eicosatetraenoic acid, and 6-trans-leukotriene B4 Bioinformatics and experimental approaches demonstrated robust correlation between transcriptional profile in Ach and FA/LM pathways and parameters of severe COVID-19. As expected, the increased neutrophil-to-lymphocyte ratio, neutrophil counts, and cytokine levels (IL-6, IL-10, IL-1β, and IL-8) correlated with worse clinical scores. Glucocorticoids protected severe and critical patients and correlated with reduced Ach levels in plasma and TA samples. We demonstrated that pulmonary and systemic hyperinflammation in severe COVID-19 are associated with high levels of Ach and FA/LM. Glucocorticoids favored the survival of patients with severe/critical disease, and this effect was associated with a reduction in ACh levels.
Collapse
Affiliation(s)
- Malena M Pérez
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Vinícius E Pimentel
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto São Paulo, Brazil
| | - Carlos A Fuzo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Pedro V da Silva-Neto
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Programa de Pós-graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Diana M Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Programa de Pós-Graduação em Biociências e Biotecnologia Aplicadas à Farmácia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Programa de Pós-graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas, Manaus, Amazonas, Brazil
| | - Thais F C Fraga-Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz G Gardinassi
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Camilla N S Oliveira
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto São Paulo, Brazil
| | - Camila O S Souza
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto São Paulo, Brazil
| | - Nicola T Torre-Neto
- Departamento de Química. Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jonatan C S de Carvalho
- Departamento de Química. Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thais C De Leo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Viviani Nardini
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marley R Feitosa
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rogerio S Parra
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José J R da Rocha
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Omar Feres
- Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.,Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando C Vilar
- Hospital São Paulo, Ribeirão Preto, São Paulo, Brazil.,Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Gilberto G Gaspar
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Leticia F Constant
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Fátima M Ostini
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Augusto M Degiovani
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Alessandro P Amorim
- Hospital Santa Casa de Misericórdia de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Angelina L Viana
- Departamento de Enfermagem Materno-Infantil e Saúde Pública, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana P M Fernandes
- Departamento de Enfermagem Geral e Especializada, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sandra R Maruyama
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Elisa M S Russo
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Isabel K F M Santos
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Vânia L D Bonato
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Cristina R B Cardoso
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carlos A Sorgi
- Departamento de Química. Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil;
| | | | | |
Collapse
|
19
|
Novel Unspecific Peroxygenase from Truncatella angustata Catalyzes the Synthesis of Bioactive Lipid Mediators. Microorganisms 2022; 10:microorganisms10071267. [PMID: 35888989 PMCID: PMC9322767 DOI: 10.3390/microorganisms10071267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/10/2022] [Accepted: 06/16/2022] [Indexed: 02/01/2023] Open
Abstract
Lipid mediators, such as epoxidized or hydroxylated eicosanoids (EETs, HETEs) of arachidonic acid (AA), are important signaling molecules and play diverse roles at different physiological and pathophysiological levels. The EETs and HETEs formed by the cytochrome P450 enzymes are still not fully explored, but show interesting anti-inflammatory properties, which make them attractive as potential therapeutic target or even as therapeutic agents. Conventional methods of chemical synthesis require several steps and complex separation techniques and lead only to low yields. Using the newly discovered unspecific peroxygenase TanUPO from the ascomycetous fungus Truncatella angustata, 90% regioselective conversion of AA to 14,15-EET could be achieved. Selective conversion of AA to 18-HETE, 19-HETE as well as to 11,12-EET and 14,15-EET was also demonstrated with known peroxygenases, i.e., AaeUPO, CraUPO, MroUPO, MweUPO and CglUPO. The metabolites were confirmed by HPLC-ELSD, MS1 and MS2 spectrometry as well as by comparing their analytical data with authentic standards. Protein structure simulations of TanUPO provided insights into its substrate access channel and give an explanation for the selective oxyfunctionalization of AA. The present study expands the scope of UPOs as they can now be used for selective syntheses of AA metabolites that serve as reference material for diagnostics, for structure-function elucidation as well as for therapeutic and pharmacological purposes.
Collapse
|
20
|
Zhang XY, Li B, Huang BC, Wang FB, Zhang YQ, Zhao SG, Li M, Wang HY, Yu XJ, Liu XY, Jiang J, Wang ZP. Production, Biosynthesis, and Commercial Applications of Fatty Acids From Oleaginous Fungi. Front Nutr 2022; 9:873657. [PMID: 35694158 PMCID: PMC9176664 DOI: 10.3389/fnut.2022.873657] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/31/2022] [Indexed: 12/18/2022] Open
Abstract
Oleaginous fungi (including fungus-like protists) are attractive in lipid production due to their short growth cycle, large biomass and high yield of lipids. Some typical oleaginous fungi including Galactomyces geotrichum, Thraustochytrids, Mortierella isabellina, and Mucor circinelloides, have been well studied for the ability to accumulate fatty acids with commercial application. Here, we review recent progress toward fermentation, extraction, of fungal fatty acids. To reduce cost of the fatty acids, fatty acid productions from raw materials were also summarized. Then, the synthesis mechanism of fatty acids was introduced. We also review recent studies of the metabolic engineering strategies have been developed as efficient tools in oleaginous fungi to overcome the biochemical limit and to improve production efficiency of the special fatty acids. It also can be predictable that metabolic engineering can further enhance biosynthesis of fatty acids and change the storage mode of fatty acids.
Collapse
Affiliation(s)
- Xin-Yue Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Bing Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Bei-Chen Huang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Feng-Biao Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Yue-Qi Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Shao-Geng Zhao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Min Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Hai-Ying Wang
- Key Laboratory of Sustainable Development of Polar Fishery, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Xin-Jun Yu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Xiao-Yan Liu
- Jiangsu Key Laboratory for Biomass-Based Energy and Enzyme Technology, Huaiyin Normal University, Huaian, China
| | - Jing Jiang
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Zhi-Peng Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
21
|
Saber S, Alomar SY, Yahya G. Blocking prostanoid receptors switches on multiple immune responses and cascades of inflammatory signaling against larval stages in snail fever. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43546-43555. [PMID: 35396684 PMCID: PMC9200668 DOI: 10.1007/s11356-022-20108-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 04/01/2022] [Indexed: 05/27/2023]
Abstract
Schistosomiasis, also known as snail fever or bilharziasis, is a worm infection caused by trematode called schistosomes that affects humans and animals worldwide. Schistosomiasis endemically exists in developing countries. Inflammatory responses elicited in the early phase of infection represent the rate limiting step for parasite migration and pathogenesis and could be a valuable target for therapeutic interventions. Prostaglandin E2 (PGE2) and interleukin (IL)-10 were found to be differentially affected in case of immune-modulation studies and cytokine analysis of hosts infected with either normal or radiation-attenuated parasite (RA) which switches off the development of an effective immune response against the migrating parasite in the early phase of schistosomiasis. Normal parasites induce predominantly a T helper 2 (Th2)-type cytokine response (IL-4 and IL-5) which is essential for parasite survival; here, we discuss in detail the downstream effects and cascades of inflammatory signaling of PGE2 and IL10 induced by normal parasites and the effect of blocking PGE2 receptors. We suggest that by selectively constraining the production of PGE2 during vaccination or therapy of susceptible persons or infected patients of schistosomiasis, this would boost IL-12 and reduce IL-10 production leading to a polarization toward the anti-worm Thl cytokine synthesis (IL-2 and Interferon (IFN)-γ).
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Suliman Y. Alomar
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451 Saudi Arabia
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Al Sharkia, 44519 Egypt
| |
Collapse
|
22
|
Adamson AL, Jeffus D, Davis A, Greengrove E. Epstein-Barr virus lytic replication activates and is dependent upon MAPK-interacting kinase 1/2 in a cell-type dependent manner. Virology 2022; 572:72-85. [DOI: 10.1016/j.virol.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/17/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022]
|
23
|
Bohnacker S, Hartung F, Henkel F, Quaranta A, Kolmert J, Priller A, Ud-Dean M, Giglberger J, Kugler LM, Pechtold L, Yazici S, Lechner A, Erber J, Protzer U, Lingor P, Knolle P, Chaker AM, Schmidt-Weber CB, Wheelock CE, Esser-von Bieren J. Mild COVID-19 imprints a long-term inflammatory eicosanoid- and chemokine memory in monocyte-derived macrophages. Mucosal Immunol 2022; 15:515-524. [PMID: 35288643 PMCID: PMC9038526 DOI: 10.1038/s41385-021-00482-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 02/08/2023]
Abstract
Monocyte-derived macrophages (MDM) drive the inflammatory response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and they are a major source of eicosanoids in airway inflammation. Here we report that MDM from SARS-CoV-2-infected individuals with mild disease show an inflammatory transcriptional and metabolic imprint that lasts for at least 5 months after SARS-CoV-2 infection. MDM from convalescent SARS-CoV-2-infected individuals showed a downregulation of pro-resolving factors and an increased production of pro-inflammatory eicosanoids, particularly 5-lipoxygenase-derived leukotrienes. Leukotriene synthesis was further enhanced by glucocorticoids and remained elevated at 3–5 months, but had returned to baseline at 12 months post SARS-CoV-2 infection. Stimulation with SARS-CoV-2 spike protein or LPS triggered exaggerated prostanoid-, type I IFN-, and chemokine responses in post COVID-19 MDM. Thus, SARS-CoV-2 infection leaves an inflammatory imprint in the monocyte/ macrophage compartment that drives aberrant macrophage effector functions and eicosanoid metabolism, resulting in long-term immune aberrations in patients recovering from mild COVID-19.
Collapse
Affiliation(s)
- Sina Bohnacker
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Franziska Hartung
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Fiona Henkel
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Alessandro Quaranta
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Johan Kolmert
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- The Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Alina Priller
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Minhaz Ud-Dean
- Institute of Computational Biology, Helmholtz Center Munich, 85764, Neuherberg, Germany
| | - Johanna Giglberger
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Luisa M Kugler
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Lisa Pechtold
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Sarah Yazici
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Antonie Lechner
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Johanna Erber
- Department of Internal Medicine II, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich (TUM), School of Medicine and Helmholtz Zentrum München, 81675, Munich, Germany
- German Center for Infection Research (DZIF), Munich partner site, Munich, Germany
| | - Paul Lingor
- Department of Neurology, University Hospital rechts der Isar, Technical University Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Percy Knolle
- Institute of Molecular Immunology and Experimental Oncology, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
- German Center for Infection Research (DZIF), Munich partner site, Munich, Germany
| | - Adam M Chaker
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital rechts der Isar, Technical University of Munich (TUM), School of Medicine, 81675, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
- German Center of Lung Research (DZL), Munich partner site, Munich, Germany
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 141-86, Stockholm, Sweden
- Gunma Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Japan
| | - Julia Esser-von Bieren
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany.
| |
Collapse
|
24
|
Welch BM, Keil AP, Bommarito PA, van T' Erve TJ, Deterding LJ, Williams JG, Lih FB, Cantonwine DE, McElrath TF, Ferguson KK. Longitudinal exposure to consumer product chemicals and changes in plasma oxylipins in pregnant women. ENVIRONMENT INTERNATIONAL 2021; 157:106787. [PMID: 34314981 PMCID: PMC8490329 DOI: 10.1016/j.envint.2021.106787] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Exposure to consumer product chemicals during pregnancy may increase susceptibility to pregnancy disorders by influencing maternal inflammation. However, effects on specific inflammatory pathways have not been well characterized. Oxylipins are a diverse class of lipids that act as important mediators and biomarkers of several biological pathways that regulate inflammation. Adverse pregnancy outcomes have been associated with circulating oxylipin levels in pregnancy. In this study, we aimed to determine the longitudinal associations between plasma oxylipins and urinary biomarkers of three classes of consumer product chemicals among pregnant women. METHODS Data come from a study of 90 pregnant women nested within the LIFECODES cohort. Maternal plasma and urine were collected at three prenatal visits. Plasma was analyzed for 61 oxylipins, which were grouped according to biosynthetic pathways that we defined by upstream: 1) fatty acid precursor, including linoleic, arachidonic, docosahexaenoic, or eicosapentaenoic acid; and 2) enzyme pathway, including cyclooxygenase (COX), lipoxygenase (LOX), or cytochrome P450 (CYP). Urine was analyzed for 12 phenol, 12 phthalate, and 9 organophosphate ester (OPE) biomarkers. Linear mixed effect models were used for single-pollutant analyses. We implemented a novel extension of quantile g-computation for longitudinal data to examine the joint effect of class-specific chemical mixtures on individual plasma oxylipin concentrations. RESULTS We found that urinary biomarkers of consumer product chemicals were positively associated with pro-inflammatory oxylipins from several biosynthetic pathways. Importantly, these associations depended upon the chemical class of exposure biomarker. We estimated positive associations between urinary phenol biomarkers and oxylipins produced from arachidonic acid by LOX enzymes, including several important pro-inflammatory hydroxyeicosatetraenoic acids (HETEs). On average, mean concentrations of oxylipin produced from the arachidonic acid/LOX pathway were 48%-71% higher per quartile increase in the phenol biomarker mixture. For example, a simultaneous quartile increase in all urinary phenols was associated with 53% higher (95% confidence interval [CI]: 11%, 111%) concentrations of 12-HETE. The positive associations among phenols were primarily driven by methyl paraben, 2,5-dichlorophenol, and triclosan. Additionally, we observed that phthalate and OPE metabolites were associated with higher concentrations of oxylipins produced from linoleic acid by CYP enzymes, including the pro-inflammatory dihydroxy-octadecenoic acids (DiHOMEs). Associations among DiHOME oxylipins were driven by metabolites of benzylbutyl and di-isodecyl phthalate, and by the metabolite of tris(1,3-dichloro-2-propyl) phosphate among OPEs. We also observed inverse associations between phthalate and OPE metabolites and oxylipins produced from other pathways; however, adjusting for a plasma indicator of dietary fatty acid intake attenuated those results. CONCLUSIONS Our findings support the hypothesis that consumer product chemicals may have diverse impacts on inflammation processes in pregnancy. Certain pro-inflammatory oxylipins were generally higher among participants with higher urinary chemical biomarker concentrations. Associations varied by class of chemical and by the biosynthetic pathway of oxylipin production, indicating potential specificity in the inflammatory effects of these environmental chemicals during pregnancy that warrant investigation in larger studies.
Collapse
Affiliation(s)
- Barrett M Welch
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States
| | - Alexander P Keil
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States; Department of Epidemiology, University of North Carolina, United States
| | - Paige A Bommarito
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States
| | | | | | - Jason G Williams
- Mass Spectrometry Research and Support Group, NIEHS, United States
| | - Fred B Lih
- Mass Spectrometry Research and Support Group, NIEHS, United States
| | - David E Cantonwine
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Thomas F McElrath
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Kelly K Ferguson
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), United States.
| |
Collapse
|
25
|
Lechner A, Bohnacker S, Esser-von Bieren J. Macrophage regulation & function in helminth infection. Semin Immunol 2021; 53:101526. [PMID: 34802871 DOI: 10.1016/j.smim.2021.101526] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 12/14/2022]
Abstract
Macrophages are innate immune cells with essential roles in host defense, inflammation, immune regulation and repair. During infection with multicellular helminth parasites, macrophages contribute to pathogen trapping and killing as well as to tissue repair and the resolution of type 2 inflammation. Macrophages produce a broad repertoire of effector molecules, including enzymes, cytokines, chemokines and growth factors that govern anti-helminth immunity and repair of parasite-induced tissue damage. Helminth infection and the associated type 2 immune response induces an alternatively activated macrophage (AAM) phenotype that - beyond driving host defense - prevents aberrant Th2 cell activation and type 2 immunopathology. The immune regulatory potential of macrophages is exploited by helminth parasites that induce the production of anti-inflammatory mediators such as interleukin 10 or prostaglandin E2 to evade host immunity. Here, we summarize current insights into the mechanisms of macrophage-mediated host defense and repair during helminth infection and highlight recent progress on the immune regulatory crosstalk between macrophages and helminth parasites. We also point out important remaining questions such as the translation of findings from murine models to human settings of helminth infection as well as long-term consequences of helminth-induced macrophage reprogramming for subsequent host immunity.
Collapse
Affiliation(s)
- Antonie Lechner
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Sina Bohnacker
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| | - Julia Esser-von Bieren
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany.
| |
Collapse
|
26
|
Distinct Metabolomic Signatures in Preclinical and Obstructive Hypertrophic Cardiomyopathy. Cells 2021; 10:cells10112950. [PMID: 34831173 PMCID: PMC8616419 DOI: 10.3390/cells10112950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
Hypertrophic Cardiomyopathy (HCM) is a common inherited heart disease with poor risk prediction due to incomplete penetrance and a lack of clear genotype–phenotype correlations. Advanced imaging techniques have shown altered myocardial energetics already in preclinical gene variant carriers. To determine whether disturbed myocardial energetics with the potential to serve as biomarkers are also reflected in the serum metabolome, we analyzed the serum metabolome of asymptomatic carriers in comparison to healthy controls and obstructive HCM patients (HOCM). We performed non-quantitative direct-infusion high-resolution mass spectrometry-based untargeted metabolomics on serum from fasted asymptomatic gene variant carriers, symptomatic HOCM patients and healthy controls (n = 31, 14 and 9, respectively). Biomarker panels that discriminated the groups were identified by performing multivariate modeling with gradient-boosting classifiers. For all three group-wise comparisons we identified a panel of 30 serum metabolites that best discriminated the groups. These metabolite panels performed equally well as advanced cardiac imaging modalities in distinguishing the groups. Seven metabolites were found to be predictive in two different comparisons and may play an important role in defining the disease stage. This study reveals unique metabolic signatures in serum of preclinical carriers and HOCM patients that may potentially be used for HCM risk stratification and precision therapeutics.
Collapse
|
27
|
Morita Y, Kurano M, Sakai E, Sawabe M, Aoki J, Yatomi Y. Simultaneous analyses of urinary eicosanoids and related mediators identified tetranor-prostaglandin E metabolite as a novel biomarker of diabetic nephropathy. J Lipid Res 2021; 62:100120. [PMID: 34560080 PMCID: PMC8515300 DOI: 10.1016/j.jlr.2021.100120] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 09/01/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
Diabetic nephropathy is a major complication of diabetes mellitus, and thus novel biomarkers are desired to evaluate the presence and progression of diabetic nephropathy. In this study, we sought to identify possible metabolites related to diabetic nephropathy among urinary eicosanoids and related mediators. Using liquid chromatogram-tandem mass spectrometry, we optimized the lipid extraction from urine using the Monospin C18 as a solid-phase extraction cartridge and measured the urinary lipid mediators in 111 subjects with type 2 diabetes mellitus as well as 33 healthy subjects. We observed that 14 metabolites differed significantly among the clinical stages of nephropathy. Among them, levels of tetranor-prostaglandin E metabolite (tetranor-PGEM), an arachidonic acid metabolite, were significantly higher in subjects with stage 1 nephropathy than in healthy subjects and increased with the progression of nephropathy. We also observed that levels of maresin-1, a docosahexaenoic acid metabolite, and leukotriene B4-ethanolamide, an arachidonoyl ethanolamide metabolite, were significantly lower in subjects with stage 3–4 nephropathy than in healthy subjects and those with stage 1–2 nephropathy. Finally, using a comprehensive analysis of urinary eicosanoids and related mediators, we concluded that tetranor-PGEM was capable of discriminating clinical stages of nephropathy and thus useful as a novel biomarker for diabetic nephropathy.
Collapse
Affiliation(s)
- Yoshifumi Morita
- Department of Clinical Laboratory, University of Tokyo Hospital, Tokyo, Japan; Department of Molecular Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory, University of Tokyo Hospital, Tokyo, Japan; Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Eri Sakai
- Department of Clinical Laboratory, University of Tokyo Hospital, Tokyo, Japan
| | - Motoji Sawabe
- Department of Molecular Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory, University of Tokyo Hospital, Tokyo, Japan; Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Jakwerth CA, Chaker AM, Guerth F, Oelsner M, Pechtold L, Zur Bonsen LS, Ullmann JT, Krauss-Etschmann S, Erb A, Kau J, Plaschke M, Winkler M, Kurz A, Kloss A, Esser-von Bieren J, Schmidt-Weber CB, Zissler UM. Sputum microRNA-screening reveals Prostaglandin EP3 receptor as selective target in allergen-specific immunotherapy. Clin Exp Allergy 2021; 51:1577-1591. [PMID: 34514658 DOI: 10.1111/cea.14013] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/08/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Several microRNAs (miRs) have been described as potential biomarkers in liquid biopsies and in the context of allergic asthma, while therapeutic effects on the airway expression of miRs remain elusive. In this study, we investigated epigenetic miR-associated mechanisms in the sputum of grass pollen-allergic patients with and without allergen-specific immunotherapy (AIT). METHODS Induced sputum samples of healthy controls (HC), AIT-treated and -untreated grass pollen-allergic rhinitis patients with (AA) and without asthma (AR) were profiled using miR microarray and whole-transcriptome microarray analysis of the same samples. miR targets were predicted in silico and used to identify inverse regulation. Local PGE2 levels were measured using ELISA. RESULTS Two hundred and fifty nine miRs were upregulated in the sputum of AA patients compared with HC, while only one was downregulated. The inverse picture was observed in induced sputum of AIT-treated patients: while 21 miRs were downregulated, only 4 miRs were upregulated in asthmatics upon AIT. Of these 4 miRs, miR-3935 stood out, as its predicted target PTGER3, the prostaglandin EP3 receptor, was downregulated in treated AA patients compared with untreated. The levels of its ligand PGE2 in the sputum supernatants of these samples were increased in allergic patients, especially asthmatics, and downregulated after AIT. Finally, local PGE2 levels correlated with ILC2 frequencies, secreted sputum IL-13 levels, inflammatory cell load, sputum eosinophils and symptom burden. CONCLUSIONS While profiling the sputum of allergic patients for novel miR expression patterns, we uncovered an association between miR-3935 and its predicted target gene, the prostaglandin E3 receptor, which might mediate AIT effects through suppression of the PGE2 -PTGER3 axis.
Collapse
Affiliation(s)
- Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Adam M Chaker
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Ferdinand Guerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Madlen Oelsner
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Lisa Pechtold
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Lynn S Zur Bonsen
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Julia T Ullmann
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Susanne Krauss-Etschmann
- Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts University Kiel, Kiel, Germany
| | - Anna Erb
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Josephine Kau
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Mirjam Plaschke
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Marlene Winkler
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Alexandra Kurz
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Antonia Kloss
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Julia Esser-von Bieren
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| |
Collapse
|
29
|
Alvarez MDL, Lorenzetti F. Role of eicosanoids in liver repair, regeneration and cancer. Biochem Pharmacol 2021; 192:114732. [PMID: 34411565 DOI: 10.1016/j.bcp.2021.114732] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022]
Abstract
Eicosanoids are lipid signaling molecules derived from the oxidation of ω-6 fatty acids, usually arachidonic acid. There are three major pathways, including the cyclooxygenase (COX), lipoxygenase (LOX), and P450 cytochrome epoxygenase (CYP) pathway. Prostanoids, which include prostaglandins (PG) and thromboxanes (Tx), are formed via the COX pathway, leukotrienes (LT) and lipoxins (LX) by the action of 5-LOX, and hydroxyeicosatetraenoic acids (HETEs) and epoxyeicosatrienoic acids (EETs) by CYP. Although eicosanoids are usually associated with pro-inflammatory responses, non-classic eicosanoids, as LX, have anti-inflammatory and pro-resolving properties. Eicosanoids like PGE2, LTB4 and EETs have been involved in promoting liver regeneration after partial hepatectomy. PGE2 and LTB4 have also been reported to participate in the regenerative phase after ischemia and reperfusion (I/R), while cysteinyl leukotrienes (Cys-LT) contribute to the inflammatory process associated with I/R and are also involved in liver fibrosis and cirrhosis. However, LX, another product of 5-LOX, have the opposite effect, acting as pro-resolving mediators in these pathologies. In liver cancer, most studies show that eicosanoids, with the exception of LX, promote the proliferation of hepatocellular carcinoma cells and favor metastasis. This review summarizes the synthesis of different eicosanoids in the liver and discusses key findings from basic research linking eicosanoids to liver repair, regeneration and cancer and the impact of targeting eicosanoid cascade. In addition, studies in patients are presented that explore the potential use of eicosanoids as biomarkers and show correlations between eicosanoid production and the course and prognosis of liver disease.
Collapse
Affiliation(s)
- María de Luján Alvarez
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, UNR, Suipacha 570 (S2002LRL), Rosario, Argentina; Área Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Suipacha 570 (S2002LRL), Rosario, Argentina; Centro de Altos Estudios en Ciencias Humanas y de la Salud (CAECIHS) Sede Regional Rosario, Universidad Abierta Interamericana, Av. Pellegrini 1618 (S2000BUG), Rosario, Argentina.
| | - Florencia Lorenzetti
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, UNR, Suipacha 570 (S2002LRL), Rosario, Argentina
| |
Collapse
|
30
|
Djuricic I, Calder PC. Beneficial Outcomes of Omega-6 and Omega-3 Polyunsaturated Fatty Acids on Human Health: An Update for 2021. Nutrients 2021; 13:nu13072421. [PMID: 34371930 PMCID: PMC8308533 DOI: 10.3390/nu13072421] [Citation(s) in RCA: 372] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/03/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress and inflammation have been recognized as important contributors to the risk of chronic non-communicable diseases. Polyunsaturated fatty acids (PUFAs) may regulate the antioxidant signaling pathway and modulate inflammatory processes. They also influence hepatic lipid metabolism and physiological responses of other organs, including the heart. Longitudinal prospective cohort studies demonstrate that there is an association between moderate intake of the omega-6 PUFA linoleic acid and lower risk of cardiovascular diseases (CVDs), most likely as a result of lower blood cholesterol concentration. Current evidence suggests that increasing intake of arachidonic acid (up to 1500 mg/day) has no adverse effect on platelet aggregation and blood clotting, immune function and markers of inflammation, but may benefit muscle and cognitive performance. Many studies show that higher intakes of omega-3 PUFAs, especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are associated with a lower incidence of chronic diseases characterized by elevated inflammation, including CVDs. This is because of the multiple molecular and cellular actions of EPA and DHA. Intervention trials using EPA + DHA indicate benefit on CVD mortality and a significant inverse linear dose-response relationship has been found between EPA + DHA intake and CVD outcomes. In addition to their antioxidant and anti-inflammatory roles, omega-3 fatty acids are considered to regulate platelet homeostasis and lower risk of thrombosis, which together indicate their potential use in COVID-19 therapy.
Collapse
Affiliation(s)
- Ivana Djuricic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia;
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
- Correspondence:
| |
Collapse
|
31
|
Torres W, Chávez-Castillo M, Peréz-Vicuña JL, Carrasquero R, Díaz MP, Gomez Y, Ramírez P, Cano C, Rojas-Quintero J, Chacín M, Velasco M, de Sanctis JB, Bermudez V. Potential role of bioactive lipids in rheumatoid arthritis. Curr Pharm Des 2021; 27:4434-4451. [PMID: 34036919 DOI: 10.2174/1381612827666210525164734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/08/2021] [Indexed: 11/22/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease, which involves a pathological inflammatory response against articular cartilage in multiple joints throughout the body. It is a complex disorder associated with comorbidities such as depression, lymphoma, osteoporosis and cardiovascular disease (CVD), which significantly deteriorate patients' quality of life and prognosis. This has ignited a large initiative to elucidate the physiopathology of RA, aiming to identify new therapeutic targets and approaches in its multidisciplinary management. Recently, various lipid bioactive products have been proposed to have an essential role in this process; including eicosanoids, specialized pro-resolving mediators, phospholipids/sphingolipids, and endocannabinoids. Dietary interventions using omega-3 polyunsaturated fatty acids or treatment with synthetic endocannabinoids agonists have been shown to significantly ameliorate RA symptoms. Indeed, the modulation of lipid metabolism may be crucial in the pathophysiology and treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Wheeler Torres
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - Mervin Chávez-Castillo
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - José L Peréz-Vicuña
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - Rubén Carrasquero
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - María P Díaz
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - Yosselin Gomez
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - Paola Ramírez
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center. School of Medicine. University of Zulia. Maracaibo. Venezuela
| | - Joselyn Rojas-Quintero
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston. 0
| | - Maricarmen Chacín
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla. Colombia
| | - Manuel Velasco
- Universidad Central de Venezuela, Escuela de Medicina José María Vargas, Caracas. Venezuela
| | - Juan Bautista de Sanctis
- Institute of Molecular and Translational Medicine. Faculty of Medicine and Dentistry. Palacky University. Czech Republic
| | - Valmore Bermudez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla. Colombia
| |
Collapse
|
32
|
Laiakis EC, Shuryak I, Deziel A, Wang YW, Barnette BL, Yu Y, Ullrich RL, Fornace AJ, Emmett MR. Effects of Low Dose Space Radiation Exposures on the Splenic Metabolome. Int J Mol Sci 2021; 22:3070. [PMID: 33802822 PMCID: PMC8002539 DOI: 10.3390/ijms22063070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Future space missions will include a return to the Moon and long duration deep space roundtrip missions to Mars. Leaving the protection that Low Earth Orbit provides will unavoidably expose astronauts to higher cumulative doses of space radiation, in addition to other stressors, e.g., microgravity. Immune regulation is known to be impacted by both radiation and spaceflight and it remains to be seen whether prolonged effects that will be encountered in deep space can have an adverse impact on health. In this study, we investigated the effects in the overall metabolism of three different low dose radiation exposures (γ-rays, 16O, and 56Fe) in spleens from male C57BL/6 mice at 1, 2, and 4 months after exposure. Forty metabolites were identified with significant enrichment in purine metabolism, tricarboxylic acid cycle, fatty acids, acylcarnitines, and amino acids. Early perturbations were more prominent in the γ irradiated samples, while later responses shifted towards more prominent responses in groups with high energy particle irradiations. Regression analysis showed a positive correlation of the abundance of identified fatty acids with time and a negative association with γ-rays, while the degradation pathway of purines was positively associated with time. Taken together, there is a strong suggestion of mitochondrial implication and the possibility of long-term effects on DNA repair and nucleotide pools following radiation exposure.
Collapse
Affiliation(s)
- Evagelia C. Laiakis
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University, New York, NY 10032, USA;
| | - Annabella Deziel
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Yi-Wen Wang
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Brooke L. Barnette
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | - Yongjia Yu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | | | - Albert J. Fornace
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Mark R. Emmett
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
33
|
Mosaad E, Peiris HN, Holland O, Morean Garcia I, Mitchell MD. The Role(s) of Eicosanoids and Exosomes in Human Parturition. Front Physiol 2020; 11:594313. [PMID: 33424622 PMCID: PMC7786405 DOI: 10.3389/fphys.2020.594313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
The roles that eicosanoids play during pregnancy and parturition are crucial to a successful outcome. A better understanding of the regulation of eicosanoid production and the roles played by the various end products during pregnancy and parturition has led to our view that accurate measurements of a panel of those end products has exciting potential as diagnostics and prognostics of preterm labor and delivery. Exosomes and their contents represent an exciting new area for research of movement of key biological factors circulating between tissues and organs akin to a parallel endocrine system but involving key intracellular mediators. Eicosanoids and enzymes regulating their biosynthesis and metabolism as well as regulatory microRNAs have been identified within exosomes. In this review, the regulation of eicosanoid production, abundance and actions during pregnancy will be explored. Additionally, the functional significance of placental exosomes will be discussed.
Collapse
Affiliation(s)
- Eman Mosaad
- School of Biomedical Science, Institute of Health and Biomedical Innovation – Centre for Children’s Health Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Hassendrini N. Peiris
- School of Biomedical Science, Institute of Health and Biomedical Innovation – Centre for Children’s Health Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Olivia Holland
- School of Biomedical Science, Institute of Health and Biomedical Innovation – Centre for Children’s Health Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Isabella Morean Garcia
- School of Biomedical Science, Institute of Health and Biomedical Innovation – Centre for Children’s Health Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Murray D. Mitchell
- School of Biomedical Science, Institute of Health and Biomedical Innovation – Centre for Children’s Health Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Wei L, Zheng YY, Sun J, Wang P, Tao T, Li Y, Chen X, Sang Y, Chong D, Zhao W, Zhou Y, Wang Y, Jiang Z, Qiu T, Li CJ, Zhu MS, Zhang X. GGPP depletion initiates metaflammation through disequilibrating CYB5R3-dependent eicosanoid metabolism. J Biol Chem 2020; 295:15988-16001. [PMID: 32913122 DOI: 10.1074/jbc.ra120.015020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/20/2020] [Indexed: 12/30/2022] Open
Abstract
Metaflammation is a primary inflammatory complication of metabolic disorders characterized by altered production of many inflammatory cytokines, adipokines, and lipid mediators. Whereas multiple inflammation networks have been identified, the mechanisms by which metaflammation is initiated have long been controversial. As the mevalonate pathway (MVA) produces abundant bioactive isoprenoids and abnormal MVA has a phenotypic association with inflammation/immunity, we speculate that isoprenoids from the MVA may provide a causal link between metaflammation and metabolic disorders. Using a line with the MVA isoprenoid producer geranylgeranyl diphosphate synthase (GGPPS) deleted, we find that geranylgeranyl pyrophosphate (GGPP) depletion causes an apparent metaflammation as evidenced by abnormal accumulation of fatty acids, eicosanoid intermediates, and proinflammatory cytokines. We also find that GGPP prenylate cytochrome b 5 reductase 3 (CYB5R3) and the prenylated CYB5R3 then translocate from the mitochondrial to the endoplasmic reticulum (ER) pool. As CYB5R3 is a critical NADH-dependent reductase necessary for eicosanoid metabolism in ER, we thus suggest that GGPP-mediated CYB5R3 prenylation is necessary for metabolism. In addition, we observe that pharmacological inhibition of the MVA pathway by simvastatin is sufficient to inhibit CYB5R3 translocation and induces smooth muscle death. Therefore, we conclude that the dysregulation of MVA intermediates is an essential mechanism for metaflammation initiation, in which the imbalanced production of eicosanoid intermediates in the ER serve as an important pathogenic factor. Moreover, the interplay of MVA and eicosanoid metabolism as we reported here illustrates a model for the coordinating regulation among metabolite pathways.
Collapse
Affiliation(s)
- Lisha Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yan-Yan Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Jie Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Pei Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Tao Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yeqiong Li
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xin Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yongjuan Sang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Danyang Chong
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Wei Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yuwei Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Ye Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zhihui Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Tiantian Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Chao-Jun Li
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China.
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China.
| | - Xuena Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center and Medical School of Nanjing University and Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
35
|
Johnson AM, Kleczko EK, Nemenoff RA. Eicosanoids in Cancer: New Roles in Immunoregulation. Front Pharmacol 2020; 11:595498. [PMID: 33364964 PMCID: PMC7751756 DOI: 10.3389/fphar.2020.595498] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022] Open
Abstract
Eicosanoids represent a family of active biolipids derived from arachidonic acid primarily through the action of cytosolic phospholipase A2-α. Three major downstream pathways have been defined: the cyclooxygenase (COX) pathway which produces prostaglandins and thromboxanes; the 5-lipoxygenase pathway (5-LO), which produces leukotrienes, lipoxins and hydroxyeicosatetraenoic acids, and the cytochrome P450 pathway which produces epoxygenated fatty acids. In general, these lipid mediators are released and act in an autocrine or paracrine fashion through binding to cell surface receptors. The pattern of eicosanoid production is cell specific, and is determined by cell-specific expression of downstream synthases. Increased eicosanoid production is associated with inflammation and a panel of specific inhibitors have been developed designated non-steroidal anti-inflammatory drugs. In cancer, eicosanoids are produced both by tumor cells as well as cells of the tumor microenvironment. Earlier studies demonstrated that prostaglandin E2, produced through the action of COX-2, promoted cancer cell proliferation and metastasis in multiple cancers. This resulted in the development of COX-2 inhibitors as potential therapeutic agents. However, cardiac toxicities associated with these agents limited their use as therapeutic agents. The advent of immunotherapy, especially the use of immune checkpoint inhibitors has revolutionized cancer treatment in multiple malignancies. However, the majority of patients do not respond to these agents as monotherapy, leading to intense investigation of other pathways mediating immunosuppression in order to develop rational combination therapies. Recent data have indicated that PGE2 has immunosuppressive activity, leading to renewed interest in targeting this pathway. However, little is known regarding the role of other eicosanoids in modulating the tumor microenvironment, and regulating anti-tumor immunity. This article reviews the role of eicosanoids in cancer, with a focus on their role in modulating the tumor microenvironment. While the role of PGE2 will be discussed, data implicating other eicosanoids, especially products produced through the lipoxygenase and cytochrome P450 pathway will be examined. The existence of small molecular inhibitors and activators of eicosanoid pathways such as specific receptor blockers make them attractive candidates for therapeutic trials, especially in combination with novel immunotherapies such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | - Raphael A. Nemenoff
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
36
|
Yasukawa K, Okuno T, Yokomizo T. Eicosanoids in Skin Wound Healing. Int J Mol Sci 2020; 21:ijms21228435. [PMID: 33182690 PMCID: PMC7698125 DOI: 10.3390/ijms21228435] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/17/2022] Open
Abstract
Wound healing is an important process in the human body to protect against external threats. A dysregulation at any stage of the wound healing process may result in the development of various intractable ulcers or excessive scar formation. Numerous factors such as growth factors, cytokines, and chemokines are involved in this process and play vital roles in tissue repair. Moreover, recent studies have demonstrated that lipid mediators derived from membrane fatty acids are also involved in the process of wound healing. Among these lipid mediators, we focus on eicosanoids such as prostaglandins, thromboxane, leukotrienes, and specialized pro-resolving mediators, which are produced during wound healing processes and play versatile roles in the process. This review article highlights the roles of eicosanoids on skin wound healing, especially focusing on the biosynthetic pathways and biological functions, i.e., inflammation, proliferation, migration, angiogenesis, remodeling, and scarring.
Collapse
Affiliation(s)
- Ken Yasukawa
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (K.Y.); (T.Y.)
- Drug Discovery Research Department, Sato Pharmaceutical Co., Ltd., Tokyo 140-0011, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (K.Y.); (T.Y.)
- Correspondence: ; Tel.: +81-3-5802-1031
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (K.Y.); (T.Y.)
| |
Collapse
|
37
|
McCabe JT, Tucker LB. Sex as a Biological Variable in Preclinical Modeling of Blast-Related Traumatic Brain Injury. Front Neurol 2020; 11:541050. [PMID: 33101170 PMCID: PMC7554632 DOI: 10.3389/fneur.2020.541050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022] Open
Abstract
Approaches to furthering our understanding of the bioeffects, behavioral changes, and treatment options following exposure to blast are a worldwide priority. Of particular need is a more concerted effort to employ animal models to determine possible sex differences, which have been reported in the clinical literature. In this review, clinical and preclinical reports concerning blast injury effects are summarized in relation to sex as a biological variable (SABV). The review outlines approaches that explore the pertinent role of sex chromosomes and gonadal steroids for delineating sex as a biological independent variable. Next, underlying biological factors that need exploration for blast effects in light of SABV are outlined, including pituitary, autonomic, vascular, and inflammation factors that all have evidence as having important SABV relevance. A major second consideration for the study of SABV and preclinical blast effects is the notable lack of consistent model design—a wide range of devices have been employed with questionable relevance to real-life scenarios—as well as poor standardization for reporting of blast parameters. Hence, the review also provides current views regarding optimal design of shock tubes for approaching the problem of primary blast effects and sex differences and outlines a plan for the regularization of reporting. Standardization and clear description of blast parameters will provide greater comparability across models, as well as unify consensus for important sex difference bioeffects.
Collapse
Affiliation(s)
- Joseph T McCabe
- Pre-clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Bethesda, IL, United States.,Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Laura B Tucker
- Pre-clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Bethesda, IL, United States.,Department of Anatomy, Physiology & Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
38
|
Yang L, Han X, Zhang L, Li N, Zhao Z, Bai J. Changes in expression of prostaglandin synthase in ovine liver during early pregnancy. CANADIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1139/cjas-2019-0171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver can function as part of the innate and adaptive immune systems. We hypothesize that prostaglandins participate in the regulation of hepatic immune function during early pregnancy in sheep. The objective of this study was to elucidate expression of prostaglandin synthase in ovine liver during early pregnancy. Ovine livers were sampled on day 16 of the estrous cycle, and days 13, 16, and 25 of pregnancy, and the expression of prostaglandin synthases, including prostaglandin-endoperoxide synthase 1 (PTGS1), PTGS2, prostaglandin E synthase (PTGES), and aldo-keto reductase family 1, member B1, a prostaglandin F synthase (PGFS), were detected by quantitative real-time polymerase chain reaction, Western blot, and immunohistochemistry analysis. There were increases in the expression of mRNA and the proteins of PTGS2, PTGES, and PGFS in the livers during early pregnancy, but PTGS1 was decreased in the pregnant ewes. The PGFS protein was limited to the hepatocytes and the endothelial cells of the proper hepatic arteries and hepatic portal veins. In summary, the upregulation of PTGS2, PTGES, and PGFS and downregulation of PTGS1 may be involved in the maternal hepatic immune adjustment during early pregnancy in sheep.
Collapse
Affiliation(s)
- Ling Yang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Xu Han
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Leying Zhang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Ning Li
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Zimo Zhao
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Jiachen Bai
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| |
Collapse
|
39
|
Catala A, Youssef LA, Reisz JA, Dzieciatkowska M, Powers NE, Marchetti C, Karafin M, Zimring JC, Hudson KE, Hansen KC, Spitalnik SL, D'Alessandro A. Metabolic Reprogramming of Mouse Bone Marrow Derived Macrophages Following Erythrophagocytosis. Front Physiol 2020; 11:396. [PMID: 32425810 PMCID: PMC7204509 DOI: 10.3389/fphys.2020.00396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/02/2020] [Indexed: 01/24/2023] Open
Abstract
Reticuloendothelial macrophages engulf ∼0.2 trillion senescent erythrocytes daily in a process called erythrophagocytosis (EP). This critical mechanism preserves systemic heme-iron homeostasis by regulating red blood cell (RBC) catabolism and iron recycling. Although extensive work has demonstrated the various effects on macrophage metabolic reprogramming by stimulation with proinflammatory cytokines, little is known about the impact of EP on the macrophage metabolome and proteome. Thus, we performed mass spectrometry-based metabolomics and proteomics analyses of mouse bone marrow-derived macrophages (BMDMs) before and after EP of IgG-coated RBCs. Further, metabolomics was performed on BMDMs incubated with free IgG to ensure that changes to macrophage metabolism were due to opsonized RBCs and not to free IgG binding. Uniformly labeled tracing experiments were conducted on BMDMs in the presence and absence of IgG-coated RBCs to assess the flux of glucose through the pentose phosphate pathway (PPP). In this study, we demonstrate that EP significantly alters amino acid and fatty acid metabolism, the Krebs cycle, OXPHOS, and arachidonate-linoleate metabolism. Increases in levels of amino acids, lipids and oxylipins, heme products, and RBC-derived proteins are noted in BMDMs following EP. Tracing experiments with U-13C6 glucose indicated a slower flux through glycolysis and enhanced PPP activation. Notably, we show that it is fueled by glucose derived from the macrophages themselves or from the extracellular media prior to EP, but not from opsonized RBCs. The PPP-derived NADPH can then fuel the oxidative burst, leading to the generation of reactive oxygen species necessary to promote digestion of phagocytosed RBC proteins via radical attack. Results were confirmed by redox proteomics experiments, demonstrating the oxidation of Cys152 and Cys94 of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and hemoglobin-β, respectively. Significant increases in early Krebs cycle and C5-branched dibasic acid metabolites (α-ketoglutarate and 2-hydroxyglutarate, respectively) indicate that EP promotes the dysregulation of mitochondrial metabolism. Lastly, EP stimulated aminolevulinic acid (ALA) synthase and arginase activity as indicated by significant accumulations of ALA and ornithine after IgG-mediated RBC ingestion. Importantly, EP-mediated metabolic reprogramming of BMDMs does not occur following exposure to IgG alone. In conclusion, we show that EP reprograms macrophage metabolism and modifies macrophage polarization.
Collapse
Affiliation(s)
- Alexis Catala
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States.,Program in Structural Biology and Biochemistry, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States
| | - Lyla A Youssef
- Department of Microbiology and Immunology, Columbia University, New York, NY, United States
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States
| | - Nicholas E Powers
- Department of Medicine - Division of Infectious Diseases, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States
| | - Carlo Marchetti
- Department of Medicine - Division of Infectious Diseases, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States
| | - Matthew Karafin
- Medical Sciences Institute, Blood Center of Wisconsin (Versiti), Milwaukee, WI, United States
| | - James C Zimring
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Krystalyn E Hudson
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States
| | - Steven L Spitalnik
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States.,Department of Medicine - Division of Hematology, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
40
|
Mackel JJ, Garth JM, Blackburn JP, Jones M, Steele C. 12/15-Lipoxygenase Deficiency Impairs Neutrophil Granulopoiesis and Lung Proinflammatory Responses to Aspergillus fumigatus. THE JOURNAL OF IMMUNOLOGY 2020; 204:1849-1858. [PMID: 32102903 PMCID: PMC8771824 DOI: 10.4049/jimmunol.1900808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/28/2020] [Indexed: 12/21/2022]
Abstract
Development of invasive aspergillosis correlates with impairments in innate immunity. We and others have recently shown that arachidonic acid metabolism pathways, specifically the cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX) pathways, participate in the induction of protective innate immune responses during invasive aspergillosis. Based on the high degree of cooperation and interconnection within the eicosanoid network, we hypothesized that 12/15-LOX is also active during invasive aspergillosis. We report in this study that mice deficient in the gene encoding 12/15-LOX (Alox15) are profoundly susceptible to invasive aspergillosis. Decreased survival correlated with increased fungal burden and evidence of increased lung damage. These defects were associated with very early (6 and 12 h) 12/15-LOX-dependent inflammatory cytokine (IL-1α, IL-1β, and TNF-α) and chemokine (CCL3 and CCL4) production. Neutrophil levels in the lung were blunted in the absence of 12/15-LOX, although neutrophil antifungal activity was intact. However, lower neutrophil levels in the lungs of Alox15−/− mice were not a result of impaired recruitment or survival; rather, Alox15−/− mice demonstrated impaired neutrophil granulopoiesis in the bone marrow intrinsically and after fungal exposure. Employing a lower inoculum to allow for better survival allowed the identification of 12/15-LOX-dependent induction of IL-17A and IL-22. Impaired IL-17A and IL-22 production correlated with reduced invariant NKT cell numbers as well as lower IL-23 levels. Together, these data indicate that 12/15-LOX is a critical player in induction of the earliest aspects of the innate immune response to Aspergillus fumigatus.
Collapse
Affiliation(s)
- Joseph J Mackel
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Jaleesa M Garth
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Jonathan P Blackburn
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - MaryJane Jones
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112
| | - Chad Steele
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA 70112
| |
Collapse
|
41
|
Yang C, Wang C, Rong Z, Xu Z, Deng K, Zhao W, Cao L, Lu Y, Adnan H, Li K, Hou Y. Mediation Analysis Reveals Potential Biological Mechanism of Ascites Influencing Recurrence in Patients with Epithelial Ovarian Cancer. Cancer Manag Res 2020; 12:793-799. [PMID: 32099475 PMCID: PMC7007789 DOI: 10.2147/cmar.s232357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/28/2019] [Indexed: 12/21/2022] Open
Abstract
Objective Ascites, an accumulation of peritoneal fluid, is associated with poor prognosis of certain cancers. The potential mechanism that ascites worsens prognosis has not been well understood. Lipids have been reported to correlate with the prognosis of patients with epithelial ovarian cancer (EOC). Therefore, we aimed here to investigate whether lipids mediate the effect of ascites on the recurrence of EOC. Methods We collected the demographic and pathological data of 437 previously untreated patients with EOC to investigate the influence of ascites on recurrence. To identify the mechanism that mediates the potential influence of ascites on recurrence, we used ultra-performance liquid chromatography coupled with mass spectrometry (UPLC-MS) to determine the plasma lipid profiles of 53 patients with EOC. We used mediation analysis to evaluate if lipids mediated the effects of ascites on the recurrence of EOC. Results Patients with ascites had a poorer prognosis, which was associated with higher levels of carbohydrate antigen-CA125 (CA125) and FIGO stage. We identified six different lipid metabolites that were associated with ascites and recurrence. Mediation analysis revealed that the lipids LysoPC(P-15:0), PC(P-34:4), and PC(38:6) may mediate the effects of ascites on recurrence. Conclusion Our findings suggest that LysoPC(P-15:0), PC(P-34:4), and PC(38:6) mediate the effect of ascites on the prognosis of patients with EOC. We believe therefore that it is reasonable to consider metabolic interventions targeting the metabolism of LysoPC(P-15:0), PC(P-34:4), and PC(38:6) as a palliative treatment for patients with EOC with ascites. Further studies of more patients will be required to validate our findings.
Collapse
Affiliation(s)
- Chunyan Yang
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Ce Wang
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Zhiwei Rong
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Zhenyi Xu
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Kui Deng
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Weiwei Zhao
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Lei Cao
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Yaxin Lu
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Humara Adnan
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Kang Li
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| | - Yan Hou
- Department of Epidemiology and Biostatistics, Public Health School, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
42
|
Peixoto MJ, Ferraz R, Magnoni LJ, Pereira R, Gonçalves JF, Calduch-Giner J, Pérez-Sánchez J, Ozório ROA. Protective effects of seaweed supplemented diet on antioxidant and immune responses in European seabass (Dicentrarchus labrax) subjected to bacterial infection. Sci Rep 2019; 9:16134. [PMID: 31695116 PMCID: PMC6834676 DOI: 10.1038/s41598-019-52693-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 10/02/2019] [Indexed: 12/16/2022] Open
Abstract
European seabass (Dicentrarchus labrax) production is often hampered by bacterial infections such as photobacteriosis caused by Photobacterium damselae subsp. piscicida (Phdp). Since diet can impact fish immunity, this work investigated the effect of dietary supplementation of 5% Gracilaria sp. aqueous extract (GRA) on seabass antioxidant capacity and resistance against Phdp. After infection, mortality was delayed in fish fed GRA, which also revealed increased lysozyme activity levels, as well as decreased lipid peroxidation, suggesting higher antioxidant capacity than in fish fed a control diet. Dietary GRA induced a down-regulation of hepatic stress-responsive heat shock proteins (grp-78, grp-170, grp-94, grp-75), while bacterial infection caused a down-regulation in antioxidant genes (prdx4 and mn-sod). Diet and infection interaction down-regulated the transcription levels of genes associated with oxidative stress response (prdx5 and gpx4) in liver. In head-kidney, GRA led to an up-regulation of genes associated with inflammation (il34, ccr9, cd33) and a down-regulation of genes related to cytokine signalling (mif, il1b, defb, a2m, myd88). Additionally, bacterial infection up-regulated immunoglobulins production (IgMs) and down-regulated the transcription of the antimicrobial peptide leap2 in head kidney. Overall, we found that GRA supplementation modulated seabass resistance to Phdp infection.
Collapse
Affiliation(s)
- Maria J Peixoto
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Av. General Norton de Matos s/n, 4450-208, Matosinhos, Portugal, Portugal.,ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Renato Ferraz
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Av. General Norton de Matos s/n, 4450-208, Matosinhos, Portugal, Portugal.,ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Leonardo J Magnoni
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Av. General Norton de Matos s/n, 4450-208, Matosinhos, Portugal, Portugal.,IIB-INTECH - Instituto de Investigaciones Biotecnológicas - Instituto Tecnológico de Chascomús (CONICET), Chascomús, Argentina
| | - Rui Pereira
- ALGAPLUS, Lda - Travessa Alexandre da Conceição S/N, 3830-196, Ílhavo, Portugal
| | - José F Gonçalves
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Josep Calduch-Giner
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595, Ribera de Cabanes, Castellón, Spain
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595, Ribera de Cabanes, Castellón, Spain
| | - Rodrigo O A Ozório
- CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Av. General Norton de Matos s/n, 4450-208, Matosinhos, Portugal, Portugal. .,ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
43
|
Giles T, Sakkas P, Belkhiri A, Barrow P, Kyriazakis I, Foster N. Differential immune response to Eimeria maxima infection in fast- and slow-growing broiler genotypes. Parasite Immunol 2019; 41:e12660. [PMID: 31230360 DOI: 10.1111/pim.12660] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/28/2022]
Abstract
Very little has been reported comparing resistance to coccidiosis in fast or slow growing broilers, the latter of which are becoming more prevalent in the broiler industry. We examined mRNA expression in the intestines of fast and slow growing broilers following Eimeria infection. We show that by day 13 post-infection (d pi) with 2500 or 7000 oocysts of Eimeria maxima, slower-growing (Ranger Classic) broilers significantly (P < 0.01) upregulated expression of proinflammatory cyclooxygenase genes (LTB4DH, PTSG1 and PTSG2) above that detected in fast growing (Ross 308) broilers. Expression of CD8α mRNA was downregulated in Ross 308 at day 6d pi with either 2500 or 7000 oocysts of E maxima (P < 0.05), compared to uninfected controls, but was not differentially expressed in Ranger Classic. CD4 genes were not differentially expressed in either chicken line infected with either infectious oocyst dose at d6 pi, compared to uninfected controls. However, at d13 pi, CD4 expression was significantly upregulated in both chicken lines infected with either infectious oocyst dose, compared to uninfected controls (P < 0.05) but this was significantly greater in Ranger Classic broilers compared to Ross 308 (P < 0.05). At d13 pi, expression of CD3 chains (required for T lymphocyte activation) was significantly increased in Ranger Classic compared to Ross 308, infected with either oocyst dose (P < 0.05-0.01). Expression of IL-2 and IL-15 mRNA, required for T lymphocyte proliferation was also significantly upregulated, or maintained longer, in Ranger Classic broilers compared to Ross 308. These differences in immune response to E maxima corresponded with a reduction in E maxima genome detected in the intestines of Ranger Classic compared to Ross 308.
Collapse
Affiliation(s)
- Tim Giles
- University of Nottingham, Sutton Bonington, UK
| | | | | | - Paul Barrow
- University of Nottingham, Sutton Bonington, UK
| | | | - Neil Foster
- University of Nottingham, Sutton Bonington, UK
| |
Collapse
|
44
|
Motta AC, Strassburg K, Oranje P, Vreeken RJ, Jacobs DM. Oxylipin profiling in endothelial cells in vitro - Effects of DHA and hydrocortisone upon an inflammatory challenge. Prostaglandins Other Lipid Mediat 2019; 144:106352. [PMID: 31260749 DOI: 10.1016/j.prostaglandins.2019.106352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/17/2019] [Accepted: 06/27/2019] [Indexed: 11/27/2022]
Abstract
Omega-3 poly-unsaturated fatty acids have been shown to have beneficial effects on several inflammatory-driven endpoints such as cardiovascular diseases. The anti-inflammatory effects of docosahexaenoic acid (DHA) are largely mediated through various oxylipins. Yet, mechanistic insights are limited. Here, we measured 53 oxylipins using LC-MS/MS in an in vitro model of endothelial cell inflammation, and compared the changes induced by DHA to hydrocortisone, a well-established anti-inflammatory drug. DHA modified several oxylipins derived from different precursors such as DHA, AA, LA and EPA. In response to a TNFα and IL-1-β challenge, DHA clearly reduced many COX-derived pro-inflammatory oxylipins, yet to a minor extent when compared to hydrocortisone. DHA also upregulated metabolites from the CYP and LOX pathways as opposed to hydrocortisone. Thus, DHA reduced pro-inflammation and enhanced pro-resolution, while hydrocortisone blunted both the pro- and anti-inflammatory pathways. Our results may fuel further research on the mitigation of corticosteroids adverse side-effects.
Collapse
Affiliation(s)
- A C Motta
- Unilever R&D, Vlaardingen, The Netherlands.
| | - K Strassburg
- Netherlands Metabolomics Centre, LACDR, Leiden University, Leiden, the Netherlands; Analytical Biosciences, LACDR, Leiden University, Leiden, the Netherlands
| | - P Oranje
- Unilever R&D, Vlaardingen, The Netherlands
| | - R J Vreeken
- Netherlands Metabolomics Centre, LACDR, Leiden University, Leiden, the Netherlands; Analytical Biosciences, LACDR, Leiden University, Leiden, the Netherlands
| | - D M Jacobs
- Unilever R&D, Vlaardingen, The Netherlands
| |
Collapse
|
45
|
Alarcón-Vila C, Pizzuto M, Pelegrín P. Purinergic receptors and the inflammatory response mediated by lipids. Curr Opin Pharmacol 2019; 47:90-96. [PMID: 30952060 DOI: 10.1016/j.coph.2019.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/15/2019] [Accepted: 02/18/2019] [Indexed: 02/07/2023]
Abstract
The inflammatory response is regulated by the production of different extracellular mediators, including lipids and extracellular nucleotides. In the extracellular environment, intermediate lipids activate specific G-protein-coupled receptors (GPCRs) in target cells and promote cell recruitment and activation. Extracellular nucleotides activate two types of receptors, the ionotropic purinergic P2X and the metabotropic purinergic P2Y receptors, inducing the release of cytokines and promoting cell recruitment. Several P2X receptors are associated with an increase in the production of immunoactive lipids mediators, which in turn are able to interfere with the activation of different P2Y receptors, establishing a tight signalling link between purinergic receptors and lipid mediators. In this review, we summarise recent studies indicating signalling crosstalk between purinergic P2X and P2Y receptor activation and lipid mediators with a focus on inflammatory diseases. Novel concepts arising from this crosstalk would result in the development of combinatorial therapies targeting lipid synthesis together with individual P2 receptors for the management of inflammatory diseases.
Collapse
Affiliation(s)
- Cristina Alarcón-Vila
- Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Malvina Pizzuto
- Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Pablo Pelegrín
- Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain.
| |
Collapse
|
46
|
Esser-von Bieren J. Eicosanoids in tissue repair. Immunol Cell Biol 2019; 97:279-288. [PMID: 30680784 DOI: 10.1111/imcb.12226] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/29/2022]
Abstract
Trauma or infection can result in tissue damage, which needs to be repaired in a well-orchestrated manner to restore tissue function and homeostasis. Lipid mediators derived from arachidonic acid (termed eicosanoids) play central and versatile roles in the regulation of tissue repair. Here, I summarize the current state-of the-art regarding the functional activities of eicosanoids in tissue repair responses during homeostasis and disease. I also describe how eicosanoids are produced during tissue damage and repair in a time-, cell- and tissue-dependent fashion. In particular, recent insights into the roles of eicosanoids in epithelial barrier repair are reviewed. Furthermore, the distinct roles of different eicosanoids in settings of pathological tissue repair such as chronic wounds, scarring or fibrosis are discussed. Finally, an outlook is provided on how eicosanoids may be targeted by future therapeutic strategies to achieve physiological tissue repair and prevent scarring and loss of tissue function in various disease contexts.
Collapse
Affiliation(s)
- Julia Esser-von Bieren
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, 80802, Munich, Germany
| |
Collapse
|
47
|
Chang J, Frandsen S, D’Annibale-Tolhurst M, Palumbo N, Gadsby J. Prostaglandin (PTG) E and F receptors in the porcine corpus luteum; effect of tumor necrosis factor-α. Anim Reprod Sci 2018; 195:139-148. [DOI: 10.1016/j.anireprosci.2018.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 01/22/2023]
|
48
|
Hu DY, Luo Y, Li CB, Zhou CY, Li XH, Peng A, Liu JY. Oxylipin profiling of human plasma reflects the renal dysfunction in uremic patients. Metabolomics 2018; 14:104. [PMID: 30830362 DOI: 10.1007/s11306-018-1402-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/26/2018] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Nearly all the enzymes that mediate the metabolism of polyunsaturated fatty acids (PUFAs) are present in the kidney. However, the correlation of renal dysfunction with PUFAs metabolism in uremic patients remains unknown. OBJECTIVES To test whether the alterations in the metabolism of PUFAs reflect the renal dysfunction in uremic patients. METHODS LC-MS/MS-based oxylipin profiling was conducted for the plasma samples from the uremic patients and controls. The data were analyzed by principal component analysis (PCA) and orthogonal partial least squares-discriminant analysis (OPLS-DA). The receiver operating characteristic (ROC) curves and the correlation of the estimated glomerular filtration rate (eGFR) with the key markers were evaluated. Furthermore, qPCR analysis of the whole blood cells was conducted to investigate the possible mechanisms. In addition, a 2nd cohort was used to validate the findings from the 1st cohort. RESULTS The plasma oxylipin profile distinguished the uremic patients from the controls successfully by using both PCA and OPLS-DA models. 5,6-Dihydroxyeicosatrienoic acid (5,6-DHET), 5-hydroxyeicosatetraenoic acid (5-HETE), 9(10)-epoxyoctadecamonoenoic acid [9(10)-EpOME] and 12(13)-EpOME were identified as the key markers to discriminate the patients from controls. The excellent predictive performance of these four markers was validated by ROC analysis. The eGFR significantly correlated with plasma levels of 5,6-DHET and 5-HETE positively but with plasma 9(10)-EpOME and 12(13)-EpOME negatively. The changes of these markers may account for the inactivation of cytochrome P450 2C18, 2C19, microsome epoxide hydrolase (EPHX1), and 5-lipoxygenase in the patients. CONCLUSION The alterations in plasma metabolic profile reflect the renal dysfunction in the uremic patients.
Collapse
Affiliation(s)
- Da-Yong Hu
- Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Ying Luo
- Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chang-Bin Li
- Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chun-Yu Zhou
- Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xin-Hua Li
- Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Ai Peng
- Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jun-Yan Liu
- Division of Nephrology and Rheumatology, Shanghai Tenth People's Hospital, Center for Nephrology and Metabolomics, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
49
|
Plasma lipidomic profile signature of rheumatoid arthritis versus Lyme arthritis patients. Arch Biochem Biophys 2018; 654:105-114. [PMID: 30059653 DOI: 10.1016/j.abb.2018.07.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/09/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Distinguishing of rheumatoid arthritis (RA) and Lyme arthritis (LA) is difficult, because of similar symptoms. This presents a significant clinical problem since treatments are quite different in both diseases. We investigated the plasma phospholipid profiles of RA and LA patients versus healthy subjects to find metabolic changes responsible for differentiation of both diseases. METHODS Plasma was collected from 9 RA, 9 LA, and 9 healthy subjects. Extracted lipids were analyzed using LC- MS/MS to characterize phospholipid profiles of RA, LA and healthy subjects. Principal components analysis (PCA), partial least squares-discriminate analysis (PLS-DA) and variable importance in projection (VIP) scores were used to estimate the importance of each phospholipid variable. RESULTS We identified 114 phospholipids in plasma. Phospholipid profiles were significantly different in RA and LA patients than in healthy subjects. Principal discriminant phospholipids between RA and LA groups were LPE (14:0), LPC(14:0) PI(18:0/20:4), PI(18:2/18:0), PI(16:1/18:2), PI(18:1/18:0), and PI(18:0/20:3). CONCLUSIONS Our study provides insights into the alteration of the plasma phospholipid profile of LA patients, resulting from Borrelia burgdorferi infection, that may lead to improved LA diagnosis and differentiation of this disease from RA. Furthermore, LPE (14:0) was found to have a high potential to be a possible biomarker of LA.
Collapse
|
50
|
Li Y, Lin N, Xu J, Lu Y, Chen S, Pan C, Wang C, Xu M, Zhou B, Xu R, Xu YJ. Measurement of Serum and Hepatic Eicosanoids by Liquid Chromatography Tandem-Mass Spectrometry (LC-MS/MS) in a Mouse Model of Hepatocellular Carcinoma (HCC) with Delivery of c-Met and Activated β-Catenin by Hepatocyte Hydrodynamic Injection. Med Sci Monit 2018; 24:1670-1679. [PMID: 29560932 PMCID: PMC5877205 DOI: 10.12659/msm.908931] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/05/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Most forms of cancer, including hepatocellular carcinoma (HCC), are associated with varying degrees of chronic inflammation. The association between the expression of eicosanoids, which are bioactive lipid mediators of inflammation, and HCC remains unknown. The aim of this study was to measure serum and hepatic eicosanoids in a mouse model of HCC with the delivery of c-Met and activated b-catenin by hepatocyte hydrodynamic injection. MATERIAL AND METHODS The HCC mouse model, and normal control mice, were used in this study with co-delivery of human c-Met combined with activated β-catenin into hepatocytes through hydrodynamic injection. Liquid chromatography tandem-mass spectrometry (LC-MS/MS) analysis was used to measure serum and hepatic eicosanoid levels. RESULTS The combined activation of c-Met and β-catenin was induced in the HCC mouse model. LC-MS/MS showed that a total of 13 eicosanoids in serum and 12 eicosanoids in liver tissue were significantly increased in the HCC mice, when compared with control mice. CONCLUSIONS In a mouse model of HCC, co-activation of the c-Met and β-catenin signaling pathway resulted in increased levels of serum and hepatic eicosanoids.
Collapse
Affiliation(s)
- Yanjie Li
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Jianliang Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Yi Lu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Shuxian Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Chuzhi Pan
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Chusi Wang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Mingxing Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Boxuan Zhou
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ruiyun Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Yong-Jiang Xu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P.R. China
| |
Collapse
|