1
|
Maurício T, Guerra IMS, Pinho M, Melo T, Bonciarelli S, Goracci L, Neves B, Domingues R, Domingues P. Phosphatidylethanolamine species with n-3 and n-6 fatty acids modulate macrophage lipidome and attenuate responses to LPS stimulation. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159614. [PMID: 40254048 DOI: 10.1016/j.bbalip.2025.159614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Phospholipids are increasingly recognized as key regulators of biological processes, including macrophage polarization and function. Among these, phosphatidylethanolamine (PE), a major constituent of cell membranes, is pivotal in maintaining cellular structure and function, yet the mechanisms through which native PE species influence macrophage immunometabolism remain largely unexplored. This study investigates the effects of two native PE species, PE 18:0/22:6 and PE 18:0/20:4, on the lipidome of resting and LPS-activated macrophages. Using C18 HPLC-MS/MS, we identified 337 lipid molecular species across 15 lipid subclasses, 332 of which varied significantly among conditions. Both PE 18:0/22:6 and PE 18:0/20:4 supplementation modulated the macrophage lipidome without inducing a pro-inflammatory phenotype under basal conditions. Notably, supplementation with PE 18:0/22:6 and PE 18:0/20:4 significantly increased lipid classes such as PE, PE O-, SM, CL, PG, LPE and PS, producing unique lipid profiles. Pre-treatment with PE 18:0/22:6 and PE 18:0/20:4 partially attenuated LPS-induced lipidomic changes, significantly reducing lipid classes like PC, including PC O- and PC P-, and Cer, which are typically linked to inflammation. While PE 18:0/20:4, from an individual lipid species perspective, may promote certain lipid profiles compatible with pro-inflammatory signaling pathways, particularly under inflammatory conditions, PE 18:0/22:6 seems to foster a lipid profile more supportive of inflammation resolution. This behaviour of PE 18:0/22:6 and PE 18:0/20:4 highlights the intricate complexity of lipid-mediated immunomodulation and emphasizes the critical role of cellular context in determining the functional outcomes of phospholipid supplementation in macrophage lipid metabolism and immune responses.
Collapse
Affiliation(s)
- Tatiana Maurício
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; Department of Medical Sciences and Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Inês M S Guerra
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Marisa Pinho
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Tânia Melo
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | | | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Bruno Neves
- Department of Medical Sciences and Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rosário Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal; CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, 3810-193 Aveiro, Portugal.
| |
Collapse
|
2
|
Rodríguez JP, Casas J, Balboa MA, Balsinde J. Bioactive lipid signaling and lipidomics in macrophage polarization: Impact on inflammation and immune regulation. Front Immunol 2025; 16:1550500. [PMID: 40028333 PMCID: PMC11867965 DOI: 10.3389/fimmu.2025.1550500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Macrophages, crucial innate immune cells, defend against pathogens and resolve inflammation, maintaining tissue balance. They perform phagocytosis, present antigens to T cells, and bond innate and adaptive immunity through various activation states. Classical activation is associated with Th1 responses and interferon γ production, while alternative activation, induced by interleukin 4, is characterized by increased endocytosis, reduced secretion of pro-inflammatory cytokines, and roles in immunoregulation and tissue remodeling. Although these represent opposite extremes observed in vitro, the remarkable plasticity of macrophages allows for a wide spectrum of activation phenotypes that are complex to characterize experimentally. While the application of omics techniques has resulted in significant advances in the characterization of macrophage polarization, lipidomic studies have received lesser attention. Beyond their role as structural components and energy sources, lipids function as signaling molecules that regulate macrophage activation and polarization, thereby shaping immune responses. This work reviews the interaction between lipid signaling and macrophage polarization, exploring how lipid metabolism influences macrophage phenotype and function. These insights offer potential therapeutic strategies for immune-mediated diseases and inflammation-related disorders, including inflammaging.
Collapse
Affiliation(s)
- Juan P. Rodríguez
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del Nordeste Argentino (IQUIBA-NEA), Universidad Nacional del Nordeste, Consejo Nacional de Investigaciones Científicas y Técnicas (UNNE-CONICET), Corrientes, Argentina
| | - Javier Casas
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Valladolid, Spain
| | - María A. Balboa
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Balsinde
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas Uva, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
3
|
Iswanto ABB, Vu MH, Shon JC, Kumar R, Wu S, Kang H, Kim DR, Son GH, Kim WY, Kwak YS, Liu KH, Kim SH, Kim JY. α1-COP modulates plasmodesmata function through sphingolipid enzyme regulation. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2024; 66:1639-1657. [PMID: 38888228 DOI: 10.1111/jipb.13711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/30/2024] [Indexed: 06/20/2024]
Abstract
Callose, a β-1,3-glucan plant cell wall polymer, regulates symplasmic channel size at plasmodesmata (PD) and plays a crucial role in a variety of plant processes. However, elucidating the molecular mechanism of PD callose homeostasis is limited. We screened and identified an Arabidopsis mutant plant with excessive callose deposition at PD and found that the mutated gene was α1-COP, a member of the coat protein I (COPI) coatomer complex. We report that loss of function of α1-COP elevates the callose accumulation at PD by affecting subcellular protein localization of callose degradation enzyme PdBG2. This process is linked to the functions of ERH1, an inositol phosphoryl ceramide synthase, and glucosylceramide synthase through physical interactions with the α1-COP protein. Additionally, the loss of function of α1-COP alters the subcellular localization of ERH1 and GCS proteins, resulting in a reduction of GlcCers and GlcHCers molecules, which are key sphingolipid (SL) species for lipid raft formation. Our findings suggest that α1-COP protein, together with SL modifiers controlling lipid raft compositions, regulates the subcellular localization of GPI-anchored PDBG2 proteins, and hence the callose turnover at PD and symplasmic movement of biomolecules. Our findings provide the first key clue to link the COPI-mediated intracellular trafficking pathway to the callose-mediated intercellular signaling pathway through PD.
Collapse
Affiliation(s)
- Arya Bagus Boedi Iswanto
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
| | - Minh Huy Vu
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
| | - Jong Cheol Shon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, Korea
| | - Ritesh Kumar
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
| | - Shuwei Wu
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
| | - Hobin Kang
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
| | - Da-Ran Kim
- Departement of Plant Medicine, Gyeongsang National University, Jinju, 52828, Korea
| | - Geon Hui Son
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
| | - Woe Yoen Kim
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
| | - Youn-Sig Kwak
- Departement of Plant Medicine, Gyeongsang National University, Jinju, 52828, Korea
| | - Kwang Hyeon Liu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 702-701, Korea
| | - Sang Hee Kim
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Korea
| | - Jae-Yean Kim
- Division of Applied Life Science (BK21 FOUR Program), Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Korea
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Korea
| |
Collapse
|
4
|
Ami D, Franco AR, Artusa V, Romerio A, Shaik MM, Italia A, Anguita J, Pasco S, Mereghetti P, Peri F, Natalello A. Vibrational spectroscopy coupled with machine learning sheds light on the cellular effects induced by rationally designed TLR4 agonists. Talanta 2024; 275:126104. [PMID: 38677166 DOI: 10.1016/j.talanta.2024.126104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
In this work, we present the potential of Fourier transform infrared (FTIR) microspectroscopy to compare on whole cells, in an unbiased and untargeted way, the capacity of bacterial lipopolysaccharide (LPS) and two rationally designed molecules (FP20 and FP20Rha) to activate molecular circuits of innate immunity. These compounds are important drug hits in the development of vaccine adjuvants and tumor immunotherapeutics. The biological assays indicated that FP20Rha was more potent than FP20 in inducing cytokine production in cells and in stimulating IgG antibody production post-vaccination in mice. Accordingly, the overall significant IR spectral changes induced by the treatment with LPS and FP20Rha were similar, lipids and glycans signals being the most diagnostic, while the effect of the less potent molecule FP20 on cells resulted to be closer to control untreated cells. We propose here the use of FTIR spectroscopy supported by artificial intelligence (AI) to achieve a more holistic understanding of the cell response to new drug candidates while screening them in cells.
Collapse
Affiliation(s)
- Diletta Ami
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Ana Rita Franco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Valentina Artusa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Alessio Romerio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Mohammed Monsoor Shaik
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Alice Italia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy
| | - Juan Anguita
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160, Derio, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbao, Bizkaia, Spain
| | - Samuel Pasco
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160, Derio, Bizkaia, Spain
| | | | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy.
| | - Antonino Natalello
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milano, Italy.
| |
Collapse
|
5
|
Dutta D, Kanca O, Byeon SK, Marcogliese PC, Zuo Z, Shridharan RV, Park JH, Lin G, Ge M, Heimer G, Kohler JN, Wheeler MT, Kaipparettu BA, Pandey A, Bellen HJ. A defect in mitochondrial fatty acid synthesis impairs iron metabolism and causes elevated ceramide levels. Nat Metab 2023; 5:1595-1614. [PMID: 37653044 PMCID: PMC11151872 DOI: 10.1038/s42255-023-00873-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 07/21/2023] [Indexed: 09/02/2023]
Abstract
In most eukaryotic cells, fatty acid synthesis (FAS) occurs in the cytoplasm and in mitochondria. However, the relative contribution of mitochondrial FAS (mtFAS) to the cellular lipidome is not well defined. Here we show that loss of function of Drosophila mitochondrial enoyl coenzyme A reductase (Mecr), which is the enzyme required for the last step of mtFAS, causes lethality, while neuronal loss of Mecr leads to progressive neurodegeneration. We observe a defect in Fe-S cluster biogenesis and increased iron levels in flies lacking mecr, leading to elevated ceramide levels. Reducing the levels of either iron or ceramide suppresses the neurodegenerative phenotypes, indicating an interplay between ceramide and iron metabolism. Mutations in human MECR cause pediatric-onset neurodegeneration, and we show that human-derived fibroblasts display similar elevated ceramide levels and impaired iron homeostasis. In summary, this study identifies a role of mecr/MECR in ceramide and iron metabolism, providing a mechanistic link between mtFAS and neurodegeneration.
Collapse
Affiliation(s)
- Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Paul C Marcogliese
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Rishi V Shridharan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Jun Hyoung Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Guang Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Ming Ge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Gali Heimer
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jennefer N Kohler
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew T Wheeler
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benny A Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Manipal Academy of Higher Education, Manipal, India
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
6
|
Jung JH, Yang DQ, Song H, Wang X, Wu X, Kim KP, Pandey A, Byeon SK. Characterization of Lipid Alterations by Oncogenic PIK3CA Mutations Using Untargeted Lipidomics in Breast Cancer. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:327-335. [PMID: 37463468 PMCID: PMC10366275 DOI: 10.1089/omi.2023.0076] [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] [Indexed: 07/20/2023]
Abstract
Lipids play crucial biological roles in health and disease, including in cancers. The phosphatidylinositol 3-kinase (PI3K) signaling pathway is a pivotal promoter of cell growth and proliferation in various types of cancer. The somatic mutations in PIK3CA, the gene coding for the catalytic subunit p110α of PI3K, are frequently present in cancer cells, including breast cancer. Although the most prominent mutants, represented by single amino acid substitutions in the helical domain in exon 9 (E545K) and the kinase domain in exon 20 (H1047R) are known to cause a gain of PI3K function, activate AKT signaling and induce oncogenic transformation, the effect of these mutations on cellular lipid profiles has not been studied. We carried out untargeted lipidomics using liquid chromatography-tandem mass spectrometry to detect the lipid alterations in mammary gland epithelial MCF10A cells with isogenic knockin of these mutations. A total of 536 species of lipids were analyzed. We found that the levels of monosialogangliosides, signaling molecules known to enhance cell motility through PI3K/AKT pathway, were significantly higher in both mutants. In addition, triglycerides and ceramides, lipid molecules known to be involved in promoting lipid droplet production, cancer cell migration and invasion, were increased, whereas lysophosphatidylcholines and phosphatidylcholines that are known to inhibit cancer cell motility were decreased in both mutants. Our results provide novel insights into a potential link between altered lipid profile and carcinogenesis caused by the PIK3CA hotspot mutations. In addition, we suggest untargeted lipidomics offers prospects for precision/personalized medicine by unpacking new molecular substrates of cancer biology.
Collapse
Affiliation(s)
- Jae Hun Jung
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Applied Chemistry, Kyung Hee University, Yongin, South Korea
| | - Da-Qing Yang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Hongming Song
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xiangyu Wang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xinyan Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Kyung Hee University, Yongin, South Korea
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Manipal Academy of Higher Education, Manipal, India
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Manabe Y, Takagi-Hayashi S, Mohri S, Sugawara T. Intestinal Absorption and Anti-Inflammatory Effects of Siphonein, a Siphonaxanthin Fatty Acid Ester from Green Algae. J Nutr Sci Vitaminol (Tokyo) 2023; 69:62-70. [PMID: 36858542 DOI: 10.3177/jnsv.69.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Siphonein is a C19 acylated siphonaxanthin found in some edible green algae (e.g., Codium fragile and Caulerpa lentillifera). Although the content of siphonein in these green algae is similar to or higher than that of siphonaxanthin, studies of health-related biological activity of siphonein are much less than those of siphonaxanthin. Given the difference in the position of the acyl chain, one cannot infer intestinal absorption of siphonein from other general carotenoid fatty acid esters. In this study, we first investigated the intestinal absorption of siphonein using mouse and cell culture models. A small amount of siphonein was detected in the plasma of treated mice, and its concentration was higher than that of siphonaxanthin (i.e., the hydrolyzed product of ingested siphonein) from 1 to 6 h after administration. Pharmacological inhibition tests with differentiated Caco-2 cells showed that Nieman-Pick C1-like 1-mediated facilitated diffusion was involved in the cellular uptake of siphonein. These results indicate that, unlike general carotenoid fatty acid esters, siphonein can be absorbed without hydrolysis. We also evaluated the anti-inflammatory effect of siphonein in differentiated Caco-2 cells. Siphonein pretreatment modulated lipopolysaccharide-induced cellular lipidome alterations and suppressed mRNA expression of proinflammatory chemokines, CXCL8 protein release, and activation of NF-κB. This study provides new insights into the absorption processes of carotenoids and shows the anti-inflammatory effect of siphonein for the first time.
Collapse
Affiliation(s)
- Yuki Manabe
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | | | - Shinsuke Mohri
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | - Tatsuya Sugawara
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| |
Collapse
|
8
|
Development of an Untargeted Metabolomics Strategy to Study the Metabolic Rewiring of Dendritic Cells upon Lipopolysaccharide Activation. Metabolites 2023; 13:metabo13030311. [PMID: 36984754 PMCID: PMC10058937 DOI: 10.3390/metabo13030311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023] Open
Abstract
Dendritic cells (DCs) are essential immune cells for defense against external pathogens. Upon activation, DCs undergo profound metabolic alterations whose precise nature remains poorly studied at a large scale and is thus far from being fully understood. The goal of the present work was to develop a reliable and accurate untargeted metabolomics workflow to get a deeper insight into the metabolism of DCs when exposed to an infectious agent (lipopolysaccharide, LPS, was used to mimic bacterial infection). As DCs transition rapidly from a non-adherent to an adherent state upon LPS exposure, one of the leading analytical challenges was to implement a single protocol suitable for getting comparable metabolomic snapshots of those two cellular states. Thus, a thoroughly optimized and robust sample preparation method consisting of a one-pot solvent-assisted method for the simultaneous cell lysis/metabolism quenching and metabolite extraction was first implemented to measure intracellular DC metabolites in an unbiased manner. We also placed special emphasis on metabolome coverage and annotation by using a combination of hydrophilic interaction liquid chromatography and reverse phase columns coupled to high-resolution mass spectrometry in conjunction with an in-house developed spectral database to identify metabolites at a high confidence level. Overall, we were able to characterize up to 171 unique meaningful metabolites in DCs. We then preliminarily compared the metabolic profiles of DCs derived from monocytes of 12 healthy donors upon in vitro LPS activation in a time-course experiment. Interestingly, the resulting data revealed differential and time-dependent activation of some particular metabolic pathways, the most impacted being nucleotides, nucleotide sugars, polyamines pathways, the TCA cycle, and to a lesser extent, the arginine pathway.
Collapse
|
9
|
Ami D, Franco AR, Artusa V, Mereghetti P, Peri F, Natalello A. A Global Picture of Molecular Changes Associated to LPS Treatment in THP-1 Derived Human Macrophages by Fourier Transform Infrared Microspectroscopy. Int J Mol Sci 2022; 23:13447. [PMID: 36362234 PMCID: PMC9656053 DOI: 10.3390/ijms232113447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 01/03/2025] Open
Abstract
Macrophages are among the first immune cells involved in the initiation of the inflammatory response to protect the host from pathogens. THP-1 derived macrophages (TDM) are used as a model to study the pro-inflammatory effects of lipopolysaccharide (LPS) exposure. Intact TDM cells were analysed by Fourier transform infrared (FTIR) microspectroscopy, supported by multivariate analysis, to obtain a snapshot of the molecular events sparked by LPS stimulation in macrophage-like cells. This spectroscopic analysis enabled the untargeted identification of the most significant spectral components affected by the treatment, ascribable mainly to lipid, protein, and sulfated sugar bands, thus stressing the fundamental role of these classes of molecules in inflammation and in immune response. Our study, therefore, shows that FTIR microspectroscopy enabled the identification of spectroscopic markers of LPS stimulation and has the potential to become a tool to assess those global biochemical changes related to inflammatory and anti-inflammatory stimuli of synthetic and natural immunomodulators different from LPS.
Collapse
Affiliation(s)
- Diletta Ami
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Ana Rita Franco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Valentina Artusa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | | | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Antonino Natalello
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
10
|
Muylle E, Jiang H, Johnsen C, Byeon SK, Ranatunga W, Garapati K, Zenka RM, Preston G, Pandey A, Kozicz T, Fang F, Morava E. TRIT1 defect leads to a recognizable phenotype of myoclonic epilepsy, speech delay, strabismus, progressive spasticity, and normal lactate levels. J Inherit Metab Dis 2022; 45:1039-1047. [PMID: 36047296 PMCID: PMC9826177 DOI: 10.1002/jimd.12550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 01/11/2023]
Abstract
TRIT1 defect is a rare, autosomal-recessive disorder of transcription, initially described as a condition with developmental delay, myoclonic seizures, and abnormal mitochondrial function. Currently, only 13 patients have been reported. We reviewed the genetic, clinical, and metabolic aspects of the disease in all known patients, including two novel, unrelated TRIT1 cases with abnormalities in oxidative phosphorylation complexes I and IV in fibroblasts. Taken together the features of all 15 patients, TRIT1 defect could be identified as a potentially recognizable syndrome including myoclonic epilepsy, speech delay, strabismus, progressive spasticity, and variable microcephaly, with normal lactate levels. Half of the patients had oxidative phosphorylation complex measurements and had multiple complex abnormalities.
Collapse
Affiliation(s)
- Ewout Muylle
- Department of Clinical GenomicsMayo ClinicRochesterMinnesotaUSA
| | - Huafang Jiang
- Department of NeurologyBeijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | | | - Seul Kee Byeon
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | | | - Kishore Garapati
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Institute of Bioinformatics, International Technology ParkBangaloreKarnatakaIndia
- Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Roman M. Zenka
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Graeme Preston
- Department of Clinical GenomicsMayo ClinicRochesterMinnesotaUSA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Tamas Kozicz
- Department of Clinical GenomicsMayo ClinicRochesterMinnesotaUSA
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Fang Fang
- Department of NeurologyBeijing Children's Hospital, Capital Medical University, National Center for Children's HealthBeijingChina
| | - Eva Morava
- Department of Clinical GenomicsMayo ClinicRochesterMinnesotaUSA
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Department of Medical GeneticsUniversity of Pecs Medical SchoolPecsHungary
- Department of BiophysicsUniversity of Pecs Medical SchoolPecsHungary
| |
Collapse
|
11
|
Lee HG, Joo M, Park JM, Kim MA, Mok J, Cho SH, Sohn YC, Lee H. Lipid Profiling of Pacific Abalone ( Haliotis discus hannai) at Different Developmental Stages Using Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectrometry. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2022; 2022:5822562. [PMID: 36299711 PMCID: PMC9592233 DOI: 10.1155/2022/5822562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/12/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
Pacific abalone (Haliotis discus hannai) is a commercially important mollusk; therefore, improvement of its growth performance and quality has been emphasized. During embryonic development, abalones undergo a series of distinct larval stages, including swimming veliger larvae, juveniles, and mature individuals, and their biomolecular composition varies depending on the developmental stage. Therefore, in the present study, we performed untargeted lipid profiling of abalone tissues at different developmental stages as well as the hemolymph of mature female and male abalones using ultrahigh-performance liquid chromatography-tandem mass spectrometry. These profiles can provide meaningful information to understand compositional changes in lipids through abalone metamorphosis and development. A total of 132 lipids belonging to 15 classes were identified from abalone tissues at different developmental stages. Moreover, 21 lipids belonging to 8 classes were identified from the hemolymph of mature abalones. All data were processed following strict criteria to provide accurate information. Triglycerides and phosphatidylcholines were the major lipid components identified in both tissues and hemolymph, accounting for, respectively, 27% and 15% of all lipids in tissues and, respectively, 24% and 38% of all lipids in the hemolymph. Of note, lysophosphatidylcholine was only detected in the tissues of mature abalones, paving the way for further analyses of abalone lipids based on developmental stages. The present findings offer novel insights into the lipidome of abalone tissues and hemolymph at different developmental stages, building a foundation for improving the efficiency and quality of abalone aquaculture.
Collapse
Affiliation(s)
- Hey Gene Lee
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - MinJoong Joo
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | | | - Mi Ae Kim
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
- East Coast Life Sciences Institute, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - JeongHun Mok
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Seong-Hyeon Cho
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Young Chang Sohn
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Hookeun Lee
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
12
|
Yoon JH, Seo Y, Jo YS, Lee S, Cho E, Cazenave-Gassiot A, Shin YS, Moon MH, An HJ, Wenk MR, Suh PG. Brain lipidomics: From functional landscape to clinical significance. SCIENCE ADVANCES 2022; 8:eadc9317. [PMID: 36112688 PMCID: PMC9481132 DOI: 10.1126/sciadv.adc9317] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/01/2022] [Indexed: 05/23/2023]
Abstract
Lipids are crucial components of cellular function owing to their role in membrane formation, intercellular signaling, energy storage, and homeostasis maintenance. In the brain, lipid dysregulations have been associated with the etiology and progression of neurodegeneration and other neurological pathologies. Hence, brain lipids are emerging as important potential targets for the early diagnosis and prognosis of neurological diseases. This review aims to highlight the significance and usefulness of lipidomics in diagnosing and treating brain diseases. We explored lipid alterations associated with brain diseases, paying attention to organ-specific characteristics and the functions of brain lipids. As the recent advances in brain lipidomics would have been impossible without advances in analytical techniques, we provide up-to-date information on mass spectrometric approaches and integrative analysis with other omic approaches. Last, we present the potential applications of lipidomics combined with artificial intelligence techniques and interdisciplinary collaborative research for treating brain diseases with clinical heterogeneities.
Collapse
Affiliation(s)
- Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Youngsuk Seo
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Yeon Suk Jo
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Seulah Lee
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Eunji Cho
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Yong-Seung Shin
- Laboratory Solutions Sales, Agilent Technologies Korea Ltd., Seoul, 06621, Republic of Korea
| | - Myeong Hee Moon
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyun Joo An
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Markus R. Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu 41062, Republic of Korea
| |
Collapse
|
13
|
Comparative lipid profiling of murine and human atherosclerotic plaques using high-resolution MALDI MSI. Pflugers Arch 2021; 474:231-242. [PMID: 34797426 PMCID: PMC8766400 DOI: 10.1007/s00424-021-02643-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 10/19/2021] [Accepted: 11/06/2021] [Indexed: 11/20/2022]
Abstract
The distribution of atherosclerotic lesions in the aorta and its branches of ApoE knockout (ApoE−/−) mice is like that of patients with atherosclerosis. By using high-resolution MALDI mass spectrometry imaging (MSI), we aimed at characterizing universally applicable physiological biomarkers by comparing the murine lipid marker profile with that of human atherosclerotic arteries. Therefore, the aorta or carotid artery of male ApoE−/− mice at different ages, human arteries with documented atherosclerotic changes originated from amputated limbs, and corresponding controls were analysed. Obtained data were subjected to multivariate statistical analysis to identify potential biomarkers. Thirty-one m/z values corresponding to individual lipid species of cholesterol esters, lysophosphatidylcholines, lysophosphatidylethanolamines, and cholesterol derivatives were found to be specific in aortic atherosclerotic plaques of old ApoE−/− mice. The lipid composition at related vessel positions of young ApoE−/− mice was more comparable with wild-type mice. Twenty-six m/z values of the murine lipid markers were found in human atherosclerotic peripheral arteries but also control vessels and showed a more patient-dependent diverse distribution. Extensive data analysis without marker preselection based on mouse data revealed lysophosphatidylcholine and glucosylated cholesterol species, the latter not being detected in the murine atherosclerotic tissue, as specific potential novel human atherosclerotic vessel markers. Despite the heterogeneous lipid profile of atherosclerotic peripheral arteries derived from human patients, we identified lipids specifically colocalized to atherosclerotic human tissue and plaques in ApoE−/− mice. These data highlight species-dependent differences in lipid profiles between peripheral artery disease and aortic atherosclerosis.
Collapse
|
14
|
Byeon SK, Madugundu AK, Pandey A. Automated data-driven mass spectrometry for improved analysis of lipids with dual dissociation techniques. J Mass Spectrom Adv Clin Lab 2021; 22:43-49. [PMID: 34939054 PMCID: PMC8662330 DOI: 10.1016/j.jmsacl.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 11/15/2022] Open
Abstract
Lipidomics is an important component of most multi-Omics systems biology studies and is largely driven by mass spectrometry (MS). Because lipids are tight regulators of multiple cellular functions, including energy homeostasis, membrane structures and cell signaling, lipidomics can provide a deeper understanding of variations underlying disease states and can become an even more powerful platform when combined with other omics, including genomics or proteomics. However, data analysis, especially in lipid annotation, poses challenges due to the heterogeneity of functional head groups and fatty acyl chains of varying hydrocarbon lengths and degrees of unsaturation. As there are various MS/MS fragmentation sites in lipids that are class-dependent, obtaining MS/MS data that includes as many fragment ions as possible is critical for structural characterization of lipids in lipidomics workflow. Here, we report an improved lipidomics methodology that resulted in increased coverage of lipidome using: 1) An automated data-driven MS/MS acquisition scheme in which inclusion and exclusion lists were automatically generated from the full scan MS of sample injections, followed by creation of updated lists over iterative analyses; and, 2) Incorporation of dual dissociation techniques of higher-energy collision dissociation and collision-induced dissociation for more accurate characterization of phosphatidylcholine species. Inclusion lists were created automatically based on full scan MS signals from samples and through iterative analyses, ions in the inclusion list that were fragmented were automatically moved to the exclusion list in subsequent runs. We confirmed that analytes with low MS response that did not undergo MS/MS events in conventional data-dependent analysis were successfully fragmented using this approach. Overall, this automated data-driven data acquisition approach resulted in a higher coverage of lipidome and the use of dual dissociation techniques provided additional information that was critical in characterizing the side chains of phosphatidylcholine species.
Collapse
Affiliation(s)
- Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN 55905 USA
| | - Anil K. Madugundu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN 55905 USA
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Institute of Bioinformatics, International Technology Park, Bangalore 560006, India
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560029, Karnataka, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN 55905 USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
15
|
Byeon SK, Khanam R, Rahman S, Hasan T, Rizvi SJR, Madugundu AK, Ramarajan MG, Jung JH, Chowdhury NH, Ahmed S, Raqib R, Kim KP, Piazza AL, Rinaldo P, Pandey A, Baqui AH, Amanhi Bio-Banking Study Group. Maternal serum lipidomics identifies lysophosphatidic acid as a predictor of small for gestational age neonates. Mol Omics 2021; 17:956-966. [PMID: 34519752 DOI: 10.1039/d1mo00131k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To discover lipidomic alterations during pregnancy in mothers who subsequently delivered small for gestational age (SGA) neonates and identify predictive lipid markers that can help recognize and manage these mothers, we carried out untargeted lipidomics on maternal serum samples collected between 24-28 weeks of gestation. We used a nested case-control study design and serum from mothers who delivered SGA and appropriate for gestational age babies. We applied untargeted lipidomics using mass spectrometry to characterize lipids and discover changes associated with SGA births during pregnancy. Multivariate pattern recognition software Collaborative Laboratory Integrated Reports (CLIR) was used for the post-analytical recognition of range differences in lipid ratios that could differentiate between SGA and control mothers and their integration for complete separation between the two groups. Here, we report changes in lipids from serum collected during pregnancy in mothers who delivered SGA neonates. In contrast to normal pregnancies where lysophosphatidic acid increased over the course of the pregnancy owing to increased activity of lysophospholipase D, we observed a decrease (32%; P = 0.05) of 20:4-lysophosphatidic acid in SGA mothers, which could potentially compromise fetal growth and development. Integration of lipid ratios in an interpretive tool (CLIR) could completely separate SGA mothers from controls demonstrating the power of untargeted lipidomic analyses for identifying novel predictive biomarkers. Additional studies are required for further assessment of the lipid biomarkers identified in this report.
Collapse
Affiliation(s)
- Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA. .,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rasheda Khanam
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | | | - Tarik Hasan
- Projahnmo Research Foundation, Dhaka, Bangladesh
| | | | - Anil K Madugundu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA. .,Institute of Bioinformatics, International Technology Park, Bangalore 560006, India.,Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.,Center for Molecular Medicine, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore, 560029, Karnataka, India
| | - Madan Gopal Ramarajan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA. .,Institute of Bioinformatics, International Technology Park, Bangalore 560006, India.,Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jae Hun Jung
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA. .,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Chemistry, Kyung Hee University, Yongin 17104, South Korea
| | | | | | - Rubhana Raqib
- Division of Infectious Diseases, International Centre for Diarrhoeal Disease Research, Bangladesh
| | - Kwang Pyo Kim
- Department of Chemistry, Kyung Hee University, Yongin 17104, South Korea
| | - Amy L Piazza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Piero Rinaldo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA. .,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Abdullah H Baqui
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | | |
Collapse
|
16
|
Che LH, Liu JW, Huo JP, Luo R, Xu RM, He C, Li YQ, Zhou AJ, Huang P, Chen YY, Ni W, Zhou YX, Liu YY, Li HY, Zhou R, Mo H, Li JM. A single-cell atlas of liver metastases of colorectal cancer reveals reprogramming of the tumor microenvironment in response to preoperative chemotherapy. Cell Discov 2021; 7:80. [PMID: 34489408 PMCID: PMC8421363 DOI: 10.1038/s41421-021-00312-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 07/18/2021] [Indexed: 02/08/2023] Open
Abstract
Metastasis is the primary cause of cancer-related mortality in colorectal cancer (CRC) patients. How to improve therapeutic options for patients with metastatic CRC is the core question for CRC treatment. However, the complexity and diversity of stromal context of the tumor microenvironment (TME) in liver metastases of CRC have not been fully understood, and the influence of stromal cells on response to chemotherapy is unclear. Here we performed an in-depth analysis of the transcriptional landscape of primary CRC, matched liver metastases and blood at single-cell resolution, and a systematic examination of transcriptional changes and phenotypic alterations of the TME in response to preoperative chemotherapy (PC). Based on 111,292 single-cell transcriptomes, our study reveals that TME of treatment-naïve tumors is characterized by the higher abundance of less-activated B cells and higher heterogeneity of tumor-associated macrophages (TAMs). By contrast, in tumors treated with PC, we found activation of B cells, lower diversity of TAMs with immature and less activated phenotype, lower abundance of both dysfunctional T cells and ECM-remodeling cancer-associated fibroblasts, and an accumulation of myofibroblasts. Our study provides a foundation for future investigation of the cellular mechanisms underlying liver metastasis of CRC and its response to PC, and opens up new possibilities for the development of therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Li-Heng Che
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing-Wen Liu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian-Ping Huo
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rong Luo
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rui-Ming Xu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Cai He
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yu-Qing Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ai-Jun Zhou
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Piao Huang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yong-Yu Chen
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wen Ni
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yun-Xia Zhou
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuan-Yuan Liu
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hui-Yan Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rong Zhou
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hui Mo
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
17
|
Byeon SK, Ramarajan MG, Madugundu AK, Oglesbee D, Vernon HJ, Pandey A. High-resolution mass spectrometric analysis of cardiolipin profiles in Barth syndrome. Mitochondrion 2021; 60:27-32. [PMID: 34273557 DOI: 10.1016/j.mito.2021.07.003] [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: 04/23/2021] [Revised: 06/16/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022]
Abstract
Barth syndrome is an X-linked recessive disorder caused by pathogenic variants in TAZ, which leads to a reduction in cardiolipin with a concomitant elevation of monolysocardiolipins. There is a paucity of studies characterizing changes in individual species of monolysocardiolipins, dilysocardiolipins and cardiolipin in Barth syndrome using high resolution untargeted lipidomics that can accurately annotate and quantify diverse lipids. We confirmed the structural diversity monolysocardiolipins, dilysocardiolipins and cardiolipin and identified individual species that showed previously unreported alterations in BTHS. Development of mass spectrometry-based targeted assays for these lipid biomarkers should provide an important tool for clinical diagnosis of Barth syndrome.
Collapse
Affiliation(s)
- Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Madan Gopal Ramarajan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States; Institute of Bioinformatics, International Technology Park, Bangalore, India; Manipal Academy of Higher Education, Manipal, India
| | - Anil K Madugundu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States; Institute of Bioinformatics, International Technology Park, Bangalore, India; Manipal Academy of Higher Education, Manipal, India; Center for Molecular Medicine, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560029, Karnataka, India
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States; Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
18
|
Li J, Chen YH, Li LZ, Wang F, Song W, Alolga RN, Zhou W, Yu H, Huang FQ, Yin X. Omics and Transgenic Analyses Reveal that Salvianolic Acid B Exhibits its Anti-Inflammatory Effects through Inhibiting the Mincle-Syk-Related Pathway in Macrophages. J Proteome Res 2021; 20:3734-3748. [PMID: 34080425 DOI: 10.1021/acs.jproteome.1c00325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Salvianolic acid B (Sal B), the main water-soluble compound in Salvia miltiorrhiza, is known to exhibit anti-inflammatory activity, however, the underlying mechanism(s) is not completely uncovered. In this study, Sal B inhibited lipopolysaccharide (LPS)-induced M1 activation and promoted the transformation of macrophages from M1- to M2-type polarization. The altered lipid profiles of LPS-induced RAW 264.7 macrophages were partly restored by Sal B treatment. At the proteomic level, a total of 5612 proteins were identified and 432 were significantly changed in macrophages under LPS treatment. The differential proteins were classified into four clusters according to their expression level in blank, LPS, and Sal B groups. LPS-induced proteins in Cluster IV including Kif14, Mincle, and Sec62 were significantly recovered to almost normal levels by Sal B treatment. Use of knockdown Mincle or picetannol (inhibitor of Syk) led to significant reductions in the gene expressions of IL-1β, iNOS, and IL-12 and the release of NO. The converse was, however, observed for overexpressed Mincle. In addition, LPS- or trehalose-6,6-dibehenate-induced phosphorylation of Syk and PKCδ was decreased by Sal B treatment. These results suggest that Sal B inhibition of LPS-induced inflammation might be through inhibition of the Mincle-Syk-PKCδ signaling pathway.
Collapse
Affiliation(s)
- Jia Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Ya-Hui Chen
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Lan-Zhu Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
- Beijing, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feizuo Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Wei Song
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Raphael N Alolga
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Wei Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Heming Yu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Feng-Qing Huang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Xiaojian Yin
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
19
|
Byeon SK, Madugundu AK, Jain AP, Bhat FA, Jung JH, Renuse S, Darrow J, Bakker A, Albert M, Moghekar A, Pandey A. Cerebrospinal fluid lipidomics for biomarkers of Alzheimer's disease. Mol Omics 2021; 17:454-463. [PMID: 34125126 PMCID: PMC8210464 DOI: 10.1039/d0mo00186d] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and is associated with serious neurologic sequelae resulting from neurodegenerative changes. Identification of markers of early-stage AD could be important for designing strategies to arrest the progression of the disease. The brain is rich in lipids because they are crucial for signal transduction and anchoring of membrane proteins. Cerebrospinal fluid (CSF) is an excellent specimen for studying the metabolism of lipids in AD because it can reflect changes occurring in the brain. We aimed to identify CSF lipidomic alterations associated with AD, using untargeted lipidomics, carried out in positive and negative ion modes. We found CSF lipids that were significantly altered in AD cases. In addition, comparison of CSF lipid profiles between persons with mild cognitive impairment (MCI) and AD showed a strong positive correlation between the lipidomes of the MCI and AD groups. The novel lipid biomarkers identified in this study are excellent candidates for validation in a larger set of patient samples and as predictive biomarkers of AD through future longitudinal studies. Once validated, the lipid biomarkers could lead to early detection, disease monitoring and the ability to measure the efficacy of potential therapeutic interventions in AD.
Collapse
Affiliation(s)
- Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55902, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Claus RA, Graeler MH. Sphingolipidomics in Translational Sepsis Research-Biomedical Considerations and Perspectives. Front Med (Lausanne) 2021; 7:616578. [PMID: 33553212 PMCID: PMC7854573 DOI: 10.3389/fmed.2020.616578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Scientific Background: Sphingolipids are a highly diverse group of lipids with respect to physicochemical properties controlling either structure, distribution, or function, all of them regulating cellular response in health and disease. Mass spectrometry, on the other hand, is an analytical technique characterizing ionized molecules or fragments thereof by mass-to-charge ratios, which has been prosperingly developed for rapid and reliable qualitative and quantitative identification of lipid species. Parallel to best performance of in-depth chromatographical separation of lipid classes, preconditions of precise quantitation of unique molecular species by preprocessing of biological samples have to be fulfilled. As a consequence, “lipid profiles” across model systems and human individuals, esp. complex (clinical) samples, have become eminent over the last couple of years due to sensitivity, specificity, and discriminatory capability. Therefore, it is significance to consider the entire experimental strategy from sample collection and preparation, data acquisition, analysis, and interpretation. Areas Covered: In this review, we outline considerations with clinical (i.e., human) samples with special emphasis on sample handling, specific physicochemical properties, target measurements, and resulting profiling of sphingolipids in biomedicine and translational research to maximize sensitivity and specificity as well as to provide robust and reproducible results. A brief commentary is also provided regarding new insights of “clinical sphingolipidomics” in translational sepsis research. Expert Opinion: The role of mass spectrometry of sphingolipids and related species (“sphingolipidomics”) to investigate cellular and compartment-specific response to stress, e.g., in generalized infection and sepsis, is on the rise and the ability to integrate multiple datasets from diverse classes of biomolecules by mass spectrometry measurements and metabolomics will be crucial to fostering our understanding of human health as well as response to disease and treatment.
Collapse
Affiliation(s)
- Ralf A Claus
- Department for Anesthesiology and Intensive Care Medicine, Sepsis Research, Jena University Hospital, Jena, Germany
| | - Markus H Graeler
- Department for Anesthesiology and Intensive Care Medicine, Sepsis Research, Jena University Hospital, Jena, Germany.,Center for Sepsis Care & Control, Jena University Hospital, Jena, Germany.,Center for Molecular Biomedicine (CMB), Jena University Hospital, Jena, Germany
| |
Collapse
|
21
|
Radenkovic S, Fitzpatrick-Schmidt T, Byeon SK, Madugundu AK, Saraswat M, Lichty A, Wong SYW, McGee S, Kubiak K, Ligezka A, Ranatunga W, Zhang Y, Wood T, Friez MJ, Clarkson K, Pandey A, Jones JR, Morava E. Expanding the clinical and metabolic phenotype of DPM2 deficient congenital disorders of glycosylation. Mol Genet Metab 2021; 132:27-37. [PMID: 33129689 PMCID: PMC7855207 DOI: 10.1016/j.ymgme.2020.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/05/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022]
Abstract
Pathogenic alterations in the DPM2 gene have been previously described in patients with hypotonia, progressive muscle weakness, absent psychomotor development, intractable seizures, and early death. We identified biallelic DPM2 variants in a 23-year-old male with truncal hypotonia, hypertonicity, congenital heart defects, intellectual disability, and generalized muscle wasting. His clinical presentation was much less severe than that of the three previously described patients. This is the second report on this ultra-rare disorder. Here we review the characteristics of previously reported individuals with a defect in the DPM complex while expanding the clinical phenotype of DPM2-Congenital Disorders of Glycosylation. In addition, we offer further insights into the pathomechanism of DPM2-CDG disorder by introducing glycomics and lipidomics analysis.
Collapse
Affiliation(s)
- Silvia Radenkovic
- Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA; Metabolomics Expertise Center, CCB, KU Leuven-VIB, Leuven, Belgium; Laboratory of Hepatology, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | | | - Seul Kee Byeon
- Mayo Clinic, Department of Laboratory of Medical Pathology, Rochester, MN, USA
| | - Anil K Madugundu
- Mayo Clinic, Department of Laboratory of Medical Pathology, Rochester, MN, USA; Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mayank Saraswat
- Mayo Clinic, Department of Laboratory of Medical Pathology, Rochester, MN, USA; Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | - Sunnie Y W Wong
- Tulane University Medical School, New Orleans, LA, USA; Stanford University, CA, USA
| | | | | | - Anna Ligezka
- Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA
| | | | - Yuebo Zhang
- Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA
| | - Tim Wood
- Greenwood Genetic Center, Greenwood, SC, USA
| | | | | | - Akhilesh Pandey
- Mayo Clinic, Department of Laboratory of Medical Pathology, Rochester, MN, USA; Mayo Clinic, Center for Individualized Medicine, Rochester, MN, USA
| | | | - Eva Morava
- Mayo Clinic, Department of Clinical Genomics, Rochester, MN, USA; Mayo Clinic, Department of Laboratory of Medical Pathology, Rochester, MN, USA
| |
Collapse
|
22
|
Lipidomic changes in mouse oocytes vitrified in PEG 8000-supplemented vitrification solutions. Cryobiology 2020; 99:140-148. [PMID: 33242477 DOI: 10.1016/j.cryobiol.2020.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 01/24/2023]
Abstract
Cryopreserved oocytes are inevitably exposed to cold stress, which negatively affects the cellular aspects of the oocytes. Lipidomic analysis of the oocytes reveals quantitative changes in lipid classes under conditions of cold stress, leading to potential freezing-vulnerability. We had previously shown that specific phospholipids are significantly downregulated in vitrified-warmed mouse oocytes compared to those in fresh oocytes. In this study, we examined whether supplementation of polyethylene glycol 8000 (PEG 8000) during vitrification influences the lipidome of the oocytes. We used liquid chromatography with tandem mass spectrometry (LC-MS/MS) to study the alteration in the lipidome in three groups of mouse oocytes: fresh, vitrified-warmed, and vitrified with PEG 8000-warmed during vitrification. In these groups, we targeted to analyze 21 lipid classes. We profiled 132 lipid species in the oocytes and statistical analyses revealed lipid classes that were up- or downregulated in these groups. Overall, our data revealed that several classes of lipids were affected during vitrification, and that oocytes vitrified with PEG 8000 to some extent alleviated the levels of changes in phospholipid and sphingolipid contents during vitrification. These results suggest that phospholipids and sphingolipids are influenced by PEG 8000 during vitrification and that PEG 8000 can be considered as a potential candidate for preserving membrane integrity during oocyte cryopreservation.
Collapse
|
23
|
Kwon YJ, Jang SN, Liu KH, Jung DH. Effect of Korean Red Ginseng on Cholesterol Metabolites in Postmenopausal Women with Hypercholesterolemia: A Pilot Randomized Controlled Trial. Nutrients 2020; 12:nu12113423. [PMID: 33171597 PMCID: PMC7695162 DOI: 10.3390/nu12113423] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/27/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
Korean red ginseng (KRG) is known to exert beneficial effects on cardiovascular health. Meanwhile, reduced estrogen at menopause has been shown to have various adverse impacts on cardiovascular risk factors, including blood lipids. The aim of this pilot study was to investigate the effect of KRG on cholesterol metabolites, which are surrogate markers of cholesterol absorption and biosynthesis, in postmenopausal women with hypercholesterolemia. The present study is an exploratory study which used data from a 4-week, double-blinded, placebo-controlled clinical pilot study in 68 postmenopausal women with hypercholesterolemia. Patients received KRG (2 g) or placebo (2 g) once daily. The primary endpoints were changes in the levels of nine sterols. Serum sterols were analyzed using liquid chromatography-mass spectrometry (LC-MS)/MS analysis. Among the sterols, reduction in cholesterol level were significantly larger in the KRG group than in the placebo group (the changes: −148.3 ± 261.1 nmol/mL in the ginseng group vs. −23.0 ± 220.5 nmol/mL in the placebo group, p = 0.039). Additionally, changes in 7-hydroxycholesterol (7-OHC) were significantly larger in the KRG group than in the placebo group (the changes: −0.05 ± 0.09 nmol/mL in the ginseng group vs. −0.002 ± 0.1 nmol/mL in the placebo group, p = 0.047). Oxysterols, cholesterol derivates, have been known to play a role in chronic inflammation diseases such as cardiovascular diseases. KRG improves sterol metabolism by decreasing cholesterol and 7-OHC levels in postmenopausal women with hypercholesterolemia.
Collapse
Affiliation(s)
- Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Korea;
| | - Su-Nyeong Jang
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Kwang-Hyeon Liu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Dong-Hyuk Jung
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Korea;
- Correspondence: ; Tel.: +82-10-4204-8998
| |
Collapse
|
24
|
Gil-de-Gómez L, Balgoma D, Montero O. Lipidomic-Based Advances in Diagnosis and Modulation of Immune Response to Cancer. Metabolites 2020; 10:metabo10080332. [PMID: 32824009 PMCID: PMC7465074 DOI: 10.3390/metabo10080332] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
While immunotherapies for diverse types of cancer are effective in many cases, relapse is still a lingering problem. Like tumor cells, activated immune cells have an anabolic metabolic profile, relying on glycolysis and the increased uptake and synthesis of fatty acids. In contrast, immature antigen-presenting cells, as well as anergic and exhausted T-cells have a catabolic metabolic profile that uses oxidative phosphorylation to provide energy for cellular processes. One goal for enhancing current immunotherapies is to identify metabolic pathways supporting the immune response to tumor antigens. A robust cell expansion and an active modulation via immune checkpoints and cytokine release are required for effective immunity. Lipids, as one of the main components of the cell membrane, are the key regulators of cell signaling and proliferation. Therefore, lipid metabolism reprogramming may impact proliferation and generate dysfunctional immune cells promoting tumor growth. Based on lipid-driven signatures, the discrimination between responsiveness and tolerance to tumor cells will support the development of accurate biomarkers and the identification of potential therapeutic targets. These findings may improve existing immunotherapies and ultimately prevent immune escape in patients for whom existing treatments have failed.
Collapse
Affiliation(s)
- Luis Gil-de-Gómez
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Colket Translational Research Center, 3501 Civic Center Blvd, PA 19104, USA
- Correspondence:
| | - David Balgoma
- Analytical Pharmaceutical Chemistry, Department of Medicinal Chemistry, Uppsala University, Husarg. 3, 75123 Uppsala, Sweden;
| | - Olimpio Montero
- Spanish National Research Council (CSIC), Boecillo’s Technological Park Bureau, Av. Francisco Vallés 8, 47151 Boecillo, Spain;
| |
Collapse
|
25
|
Conde TA, Mendes L, Gaspar VM, Mano JF, Melo T, Domingues MR, Duarte IF. Differential Modulation of the Phospholipidome of Proinflammatory Human Macrophages by the Flavonoids Quercetin, Naringin and Naringenin. Molecules 2020; 25:molecules25153460. [PMID: 32751373 PMCID: PMC7436172 DOI: 10.3390/molecules25153460] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/03/2023] Open
Abstract
The immunomodulatory activity of flavonoids is increasingly appreciated. Macrophage phospholipids (PLs) play crucial roles in cell-mediated inflammatory responses. However, little is known on how these PLs are affected upon flavonoid treatment. In this work, we have used mass-spectrometry-based lipidomics to characterize the changes in the phospholipidome of proinflammatory human-macrophage-like cells (THP-1-derived and LPS+IFN-γ-stimulated) incubated with non-cytotoxic concentrations of three flavonoids: quercetin, naringin and naringenin. One hundred forty-seven PL species belonging to various classes were identified, and their relative abundances were determined. Each flavonoid displayed its own unique signature of induced effects. Quercetin produced the strongest impact, acting both on constitutive PLs (phosphatidylcholines, phosphatidylethanolamines and sphingomyelins) and on minor signaling lipids, such as phosphatidylinositol (PI) and phosphatidylserine (PS) species. Conversely, naringin hardly affected structural PLs, producing changes in signaling molecules that were opposite to those seen in quercetin-treated macrophages. In turn, albeit sharing some effects with quercetin, naringenin did not change PI and PS levels and interfered with a set of phosphatidylcholines distinct from those modulated by quercetin. These results demonstrate that flavonoids bioactivity involves profound and specific remodeling of macrophage phospholipidome, paving the way to future studies on the role of cellular phospholipids in flavonoid-mediated immunomodulatory effects.
Collapse
Affiliation(s)
- Tiago A. Conde
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (T.A.C.); (L.M.); (V.M.G.); (J.F.M.)
- LAQV-REQUIMTE, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Luís Mendes
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (T.A.C.); (L.M.); (V.M.G.); (J.F.M.)
| | - Vítor M. Gaspar
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (T.A.C.); (L.M.); (V.M.G.); (J.F.M.)
| | - João F. Mano
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (T.A.C.); (L.M.); (V.M.G.); (J.F.M.)
| | - Tânia Melo
- LAQV-REQUIMTE, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- CESAM, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - M. Rosário Domingues
- LAQV-REQUIMTE, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- CESAM, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence: (M.R.D.); (I.F.D.)
| | - Iola F. Duarte
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (T.A.C.); (L.M.); (V.M.G.); (J.F.M.)
- Correspondence: (M.R.D.); (I.F.D.)
| |
Collapse
|
26
|
Seim GL, John SV, Fan J. Metabolomic and Lipidomic Analysis of Bone Marrow Derived Macrophages. Bio Protoc 2020; 10:e3693. [PMID: 33659361 DOI: 10.21769/bioprotoc.3693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/27/2020] [Accepted: 05/10/2020] [Indexed: 12/20/2022] Open
Abstract
Macrophages are highly plastic immune cells that are capable of adopting a wide array of functional phenotypes in response to environmental stimuli. The changes in macrophage function are often supported and regulated by changes in cellular metabolism. Capturing a comprehensive picture of metabolism is vital for understanding the role of metabolic rewiring in the immune response. Here we present a method for systematically quantifying the abundance of metabolites and lipids in primary murine bone marrow derived macrophages (BMDMs). This method simultaneously extracts polar metabolites and lipids from BMDMs using a rapid two-phase extraction procedure. The polar metabolite fraction and lipid fraction are subsequently analyzed by separate liquid chromatography-mass spectrometry (LC-MS) methods for optimized coverage and quantification. This allows for a comprehensive characterization of cellular metabolism that can be used to understand the impact of a variety of environmental stimuli on macrophage metabolism and function.
Collapse
Affiliation(s)
- Gretchen L Seim
- Morgridge Institute for Research, Madison, WI, USA.,Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Jing Fan
- Morgridge Institute for Research, Madison, WI, USA.,Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
27
|
Advances in lipidomics. Clin Chim Acta 2020; 510:123-141. [PMID: 32622966 DOI: 10.1016/j.cca.2020.06.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/24/2023]
Abstract
The present article examines recently published literature on lipids, mainly focusing on research involving glycero-, glycerophospho- and sphingo-lipids. The primary aim is identification of distinct profiles in biologic lipidomic systems by ultra-high-performance liquid chromatography (UHPLC) coupled with mass spectrometry (MS, tandem MS) with multivariate data analysis. This review specifically targets lipid biomarkers and disease pathway mechanisms in humans and artificial targets. Different specimen matrices such as primary blood derivatives (plasma, serum, erythrocytes, and blood platelets), faecal matter, urine, as well as biologic tissues (liver, lung and kidney) are highlighted.
Collapse
|
28
|
Luo L, Zhou J, Zhao H, Fan M, Gao W. The anti-inflammatory effects of formononetin and ononin on lipopolysaccharide-induced zebrafish models based on lipidomics and targeted transcriptomics. Metabolomics 2019; 15:153. [PMID: 31768751 DOI: 10.1007/s11306-019-1614-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Formononetin (MBHS) and its glycosylated derivative ononin (MBHG), as the major isoflavones, have exhibited the anti-inflammatory impacts on the lipopolysaccharide (LPS)-induced inflammation. Although various researches have focused on interpreting the pharmaceutical activities of MBHG and MBHS, the molecular mechanisms in zebrafish models are still unclear. OBJECTIVE The purpose of the present work is to investigate the molecular mechanisms of the anti-inflammatory effects of MGHG and MBHS based on lipidomics and targeted transcriptomics. METHODS UHPLC-MS was applied for the lipid analyses and RT-PCR was adopted for the mRNA analyses, and the results of different groups were compared for exploring the significantly changed lipids and mRNAs. RESULTS The results of lipidomics revealed that phosphatidylcholines (PCs) were drastically down-regulated in the MBHG or MBHS treated LPS-induced inflammatory zebrafish models. Besides, MBHS can also decrease the levels of triacylglycerols (TAGs). For the targeted transcriptomics analyses, 4 cytokines (TNF-α, IL-1β, IL-6 and IFN-γ) and 3 mRNA (JNK1, ERK1 and p38a) involved in the MAPK pathway were down-regulated and IL-10 was up-regulated under the treatment of MBHG or MBHS. CONCLUSION Combining the results of lipidomics and targeted transcriptomics, we indicated that MBHG and MBHS exerted potent anti-inflammatory effects on the LPS-induced zebrafish models through the MyD88 or TRIF MAPK/ERK and MAPK/JNK pathways and the glycerophospholipid, glycosylphosphatidylinositol (GPI)-anchor biosynthesis and glycerolipid metabolisms. Our results provided new insights into the anti-inflammatory mechanisms of MBHG or MBHS and supplied an effective method to interpret the pharmacological mechanisms of drugs.
Collapse
Affiliation(s)
- Liyu Luo
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Junyi Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Miaoxuan Fan
- Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute of Drug Control, Beijing, 102206, China.
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
| |
Collapse
|
29
|
Yu Q, Tian X, Shao L, Li X, Dai R. Targeted metabolomics to reveal muscle-specific energy metabolism between bovine longissimus lumborum and psoas major during early postmortem periods. Meat Sci 2019; 156:166-173. [DOI: 10.1016/j.meatsci.2019.05.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/28/2019] [Accepted: 05/28/2019] [Indexed: 11/16/2022]
|
30
|
Noh SA, Kim SM, Park SH, Kim DJ, Lee JW, Kim YG, Moon JY, Lim SJ, Lee SH, Kim KP. Alterations in Lipid Profile of the Aging Kidney Identified by MALDI Imaging Mass Spectrometry. J Proteome Res 2019; 18:2803-2812. [PMID: 31244212 DOI: 10.1021/acs.jproteome.9b00108] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During aging, the kidney undergoes functional and physiological changes that are closely affiliated with chronic kidney disease (CKD). There is increasing evidence supporting the role of lipid or lipid-derived mediators in the pathogenesis of CKD and other aging-related diseases. To understand the role of lipids in various metabolic processes during kidney aging, we conducted matrix-assisted laser desorption/ionization-imaging mass spectrometry (MALDI-IMS) analysis in kidneys harvested from young (2 months old, n = 3) and old mice (24 months old, n = 3). MALDI-IMS analysis showed an increase in ceramide level and a decrease in sphingomyelin (SM) and phosphatidylcholine (PC) levels in kidneys of old mice. The increased expression of cPLA2 and SMPD1 protein in aged kidney was confirmed by immunohistochemistry and Western blot analysis. Our MALDI-IMS data showed the altered distribution of lipids in aged kidney as indicative of aging-related functional changes of the kidney. Combined analysis of MALDI-IMS and IHC confirmed lipidomic changes and expression levels of responsible enzymes as well as morphological changes.
Collapse
Affiliation(s)
- Sue Ah Noh
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials , Kyung Hee University , Yongin , Republic of Korea
| | - Su-Mi Kim
- Department of Nephrology, School of Medicine , Kyung Hee University , Seoul , Republic of Korea
| | - Seon Hwa Park
- Department of Nephrology, School of Medicine , Kyung Hee University , Seoul , Republic of Korea
| | - Dong-Jin Kim
- Department of Nephrology, School of Medicine , Kyung Hee University , Seoul , Republic of Korea
| | - Joon Won Lee
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials , Kyung Hee University , Yongin , Republic of Korea
| | - Yang Gyun Kim
- Department of Nephrology, School of Medicine , Kyung Hee University , Seoul , Republic of Korea
| | - Ju-Young Moon
- Department of Nephrology, School of Medicine , Kyung Hee University , Seoul , Republic of Korea
| | - Sung-Jig Lim
- Department of Pathology , Kyung Hee University Hospital at Gangdong , Seoul , Republic of Korea
| | - Sang-Ho Lee
- Department of Nephrology, School of Medicine , Kyung Hee University , Seoul , Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials , Kyung Hee University , Yongin , Republic of Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute , Kyung Hee University , Seoul , Republic of Korea
| |
Collapse
|
31
|
Khan MM, Ernst O, Manes NP, Oyler BL, Fraser IDC, Goodlett DR, Nita-Lazar A. Multi-Omics Strategies Uncover Host-Pathogen Interactions. ACS Infect Dis 2019; 5:493-505. [PMID: 30857388 DOI: 10.1021/acsinfecdis.9b00080] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With the success of the Human Genome Project, large-scale systemic projects became a reality that enabled rapid development of the systems biology field. Systems biology approaches to host-pathogen interactions have been instrumental in the discovery of some specifics of Gram-negative bacterial recognition, host signal transduction, and immune tolerance. However, further research, particularly using multi-omics approaches, is essential to untangle the genetic, immunologic, (post)transcriptional, (post)translational, and metabolic mechanisms underlying progression from infection to clearance of microbes. The key to understanding host-pathogen interactions lies in acquiring, analyzing, and modeling multimodal data obtained through integrative multi-omics experiments. In this article, we will discuss how multi-omics analyses are adding to our understanding of the molecular basis of host-pathogen interactions and systemic maladaptive immune response of the host to microbes and microbial products.
Collapse
Affiliation(s)
- Mohd M. Khan
- Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, Maryland 20814, United States
- University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, Maryland 21201, United States
| | - Orna Ernst
- Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, Maryland 20814, United States
| | - Nathan P. Manes
- Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, Maryland 20814, United States
| | - Benjamin L. Oyler
- University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, Maryland 21201, United States
| | - Iain D. C. Fraser
- Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, Maryland 20814, United States
| | - David R. Goodlett
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 North Pine Street, Baltimore, Maryland 21201, United States
| | - Aleksandra Nita-Lazar
- Laboratory of Immune System Biology (LISB), National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bethesda, Maryland 20814, United States
| |
Collapse
|
32
|
Wang S, Liu R, Yu Q, Dong L, Bi Y, Liu G. Metabolic reprogramming of macrophages during infections and cancer. Cancer Lett 2019; 452:14-22. [PMID: 30905817 DOI: 10.1016/j.canlet.2019.03.015] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/20/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022]
Abstract
In response to different microenvironmental stimuli, macrophages are polarized into two populations, M1 macrophages which are classically activated by interferon (IFN)-γ with lipopolysaccharides (LPSs) and M2 macrophages which are alternatively activated by interleukin-4 (IL-4), to perform specific roles in innate immune responses. Accordingly, macrophages occupy distinct metabolic profiles, regulated by orchestrated factors and signaling pathways, including the PI3K-AKT, HIF, c-Myc, AMPK, and PPARs pathways. These factors and pathways play pivotal roles not only in metabolic regulation but also in macrophage polarization. After activation, classically activated M1 macrophages and alternatively activated M2 macrophages display distinct patterns in glucose, lipid, amino acid and iron metabolism. Here, we summarized recently discovered metabolism-related inflammatory signaling factors, along with reprogrammed metabolism, after the activation of macrophages under conditions related to immunity and cancer. Additionally, macrophage regulatory roles in infectious diseases, cancer progression and anti-cancer immunotherapy are discussed in terms of metabolic profiles, providing insight into the prevention and treatment of immune-associated diseases.
Collapse
Affiliation(s)
- Shiyao Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Ruichen Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Qing Yu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Lin Dong
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China.
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
33
|
Im SS, Park HY, Shon JC, Chung IS, Cho HC, Liu KH, Song DK. Plasma sphingomyelins increase in pre-diabetic Korean men with abdominal obesity. PLoS One 2019; 14:e0213285. [PMID: 30835753 PMCID: PMC6400388 DOI: 10.1371/journal.pone.0213285] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/18/2019] [Indexed: 12/29/2022] Open
Abstract
Abdominal or visceral obesity is a well-known risk factor for metabolic diseases. However, whether abdominal obesity significantly affects plasma lipid profile during the development of type 2 diabetes has not been fully elucidated. We investigated the differences in plasma lipid concentrations in 63 participants categorized into six groups (middle-aged Korean men); Normal, Pre-diabetes (pre-DM), and Diabetes mellitus (DM) with or without abdominal obesity (AO or lean). The lipidomic profiles were determined by using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Sphingomyelin (SM) levels in plasma were significantly higher in the pre-DM with AO than in pre-DM with lean (p = 0.021). SM concentrations correlated positively with waist-to-hip ratio (WHR) (r = 0.256, p = 0.044), cholesteryl ester (CE) (r = 0.483, p < 0.0001), ceramide (r = 0.489, p < 0.0001) and plasmanyl phosphatidylcholine (PC) (r = 0.446, p < 0.0001). The present study found that pre-diabetic patients with AO were characterized by increased plasma concentrations of SM. Plasma SM levels in individuals with AO may be an early prognostic biomarker to better predict the progression toward type 2 diabetes and metabolic syndrome.
Collapse
Affiliation(s)
- Seung-Soon Im
- Department of Physiology and Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, Korea
| | - Hyeon Young Park
- Department of Physiology and Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, Korea
| | - Jong Cheol Shon
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Korea
| | - In-Sung Chung
- Department of Occupational & Environmental Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Ho Chan Cho
- Department of Clinical Endocrinology, Keimyung University School of Medicine, Daegu, Korea
| | - Kwang-Hyeon Liu
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Korea
| | - Dae-Kyu Song
- Department of Physiology and Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
34
|
Beyer S, Koch M, Lee YH, Jung F, Blocki A. An In Vitro Model of Angiogenesis during Wound Healing Provides Insights into the Complex Role of Cells and Factors in the Inflammatory and Proliferation Phase. Int J Mol Sci 2018; 19:ijms19102913. [PMID: 30257508 PMCID: PMC6213254 DOI: 10.3390/ijms19102913] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/04/2018] [Accepted: 09/16/2018] [Indexed: 12/21/2022] Open
Abstract
Successful vascularization is essential in wound healing, the histo-integration of biomaterials, and other aspects of regenerative medicine. We developed a functional in vitro assay to dissect the complex processes directing angiogenesis during wound healing, whereby vascular cell spheroids were induced to sprout in the presence of classically (M1) or alternatively (M2) activated macrophages. This simulated a microenvironment, in which sprouting cells were exposed to the inflammatory or proliferation phases of wound healing, respectively. We showed that M1 macrophages induced single-cell migration of endothelial cells and pericytes. In contrast, M2 macrophages augmented endothelial sprouting, suggesting that vascular cells infiltrate the wound bed during the inflammatory phase and extensive angiogenesis is initiated upon a switch to a predominance of M2. Interestingly, M1 and M2 shared a pro-angiogenic secretome, whereas pro-inflammatory cytokines were solely secreted by M1. These results suggested that acute inflammatory factors act as key inducers of vascular cell infiltration and as key negative regulators of angiogenesis, whereas pro-angiogenic factors are present throughout early wound healing. This points to inflammatory factors as key targets to modulate angiogenesis. The here-established wound healing assay represents a useful tool to investigate the effect of biomaterials and factors on angiogenesis during wound healing.
Collapse
Affiliation(s)
- Sebastian Beyer
- Institute for Tissue Engineering and Regenerative Medicine, Chinese University of Hong Kong, New Territories, Hong Kong, China.
- BioSyM Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 229899, Singapore.
| | - Maria Koch
- BioSyM Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 229899, Singapore.
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia.
| | - Yie Hou Lee
- BioSyM Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 229899, Singapore.
- Translational 'Omics and Biomarkers core, KK Research Centre, KK Women's and Children's Hospital, Singapore 169857, Singapore.
- Obstetrics & Gynaecology-Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore.
| | - Friedrich Jung
- Institute for Clinical Hemostasiology and Transfusion Medicine, University Saarland, 66421 Homburg/Saar, Germany.
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, Chinese University of Hong Kong, New Territories, Hong Kong, China.
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, New Territories, Hong Kong, China.
| |
Collapse
|
35
|
Rebollo-Ramirez S, Krokowski S, Lobato-Márquez D, Thomson M, Pennisi I, Mostowy S, Larrouy-Maumus G. Intact Cell Lipidomics Reveal Changes to the Ratio of Cardiolipins to Phosphatidylinositols in Response to Kanamycin in HeLa and Primary Cells. Chem Res Toxicol 2018; 31:688-696. [PMID: 29947513 PMCID: PMC6103485 DOI: 10.1021/acs.chemrestox.8b00038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Indexed: 01/03/2023]
Abstract
Antimicrobial resistance is a major threat the world is currently facing. Development of new antibiotics and the assessment of their toxicity represent important challenges. Current methods for addressing antibiotic toxicity rely on measuring mitochondrial damage using ATP and/or membrane potential as a readout. In this study, we propose an alternative readout looking at changes in the lipidome on intact and unprocessed cells by matrix-assisted laser desorption ionization mass spectrometry. As a proof of principle, we evaluated the impact of known antibiotics (levofloxacin, ethambutol, and kanamycin) on the lipidome of HeLa cells and mouse bone marrow-derived macrophages. Our methodology revealed that clinically relevant concentrations of kanamycin alter the ratio of cardiolipins to phosphatidylinositols. Unexpectedly, only kanamycin had this effect even though all antibiotics used in this study led to a decrease in the maximal mitochondrial respiratory capacity. Altogether, we report that intact cell-targeted lipidomics can be used as a qualitative method to rapidly assess the toxicity of aminoglycosides in HeLa and primary cells. Moreover, these results demonstrate there is no direct correlation between the ratio of cardiolipins to phosphatidylinositols and the maximal mitochondrial respiratory capacity.
Collapse
Affiliation(s)
- Sonia Rebollo-Ramirez
- MRC
Centre for Molecular Bacteriology and Infection, Department of Life
Sciences, Faculty of Natural Sciences, Imperial
College London, London SW7 2AZ, U.K.
| | - Sina Krokowski
- MRC
Centre for Molecular Bacteriology and Infection, Department of Medicine,
Section of Microbiology, Imperial College
London, London W12 0NN, U.K.
- Department
of Immunology and Infection, London School
of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Damian Lobato-Márquez
- MRC
Centre for Molecular Bacteriology and Infection, Department of Medicine,
Section of Microbiology, Imperial College
London, London W12 0NN, U.K.
- Department
of Immunology and Infection, London School
of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Michael Thomson
- MRC
Centre for Molecular Bacteriology and Infection, Department of Life
Sciences, Faculty of Natural Sciences, Imperial
College London, London SW7 2AZ, U.K.
| | - Ivana Pennisi
- MRC
Centre for Molecular Bacteriology and Infection, Department of Life
Sciences, Faculty of Natural Sciences, Imperial
College London, London SW7 2AZ, U.K.
| | - Serge Mostowy
- MRC
Centre for Molecular Bacteriology and Infection, Department of Medicine,
Section of Microbiology, Imperial College
London, London W12 0NN, U.K.
- Department
of Immunology and Infection, London School
of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Gerald Larrouy-Maumus
- MRC
Centre for Molecular Bacteriology and Infection, Department of Life
Sciences, Faculty of Natural Sciences, Imperial
College London, London SW7 2AZ, U.K.
| |
Collapse
|
36
|
Saha S, Shalova IN, Biswas SK. Metabolic regulation of macrophage phenotype and function. Immunol Rev 2018; 280:102-111. [PMID: 29027220 DOI: 10.1111/imr.12603] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Studies in the last 20 years have given us a remarkable insight into the functional and phenotypic diversity of macrophages which reflects their integral role in host defence, homeostasis and pathogenesis. Mouse genetics, transcriptomic and epigenetic studies have provided an ontogenic and molecular perspective to the phenotypic diversity of these cells. Recently, metabolic studies have revealed the crucial role of metabolism and metabolites in shaping the phenotype and function of macrophages. Evidence pertaining to this aspect will be reviewed here.
Collapse
Affiliation(s)
- Shilpi Saha
- Singapore Immunology Network (SIgN), Agency for Science, Technology & Research (A*STAR), Singapore
| | - Irina N Shalova
- Singapore Immunology Network (SIgN), Agency for Science, Technology & Research (A*STAR), Singapore
| | - Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science, Technology & Research (A*STAR), Singapore
| |
Collapse
|
37
|
Gong L, Xu H, Yuan H, Wang L, Yin X, Fan M, Cheng L, Ma X, Liang R, Yang H. Identification of absorbed constituents and evaluation of the pharmacokinetics of main compounds after oral administration of yindanxinnaotong by UPLC-Q-TOF-MS and UPLC-QqQ-MS. RSC Adv 2018; 8:15725-15739. [PMID: 35539456 PMCID: PMC9080097 DOI: 10.1039/c7ra12659j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/14/2018] [Indexed: 11/21/2022] Open
Abstract
Yindanxinnaotong capsule (YDXNT), a traditional Chinese formula, has been used to treat cardio-cerebrovascular diseases for several decades. Previous research has focused on evaluating the pharmacological properties and main compounds of YDXNT in vitro and in vivo. However, the multiple bioactive compounds in vivo remain poorly understood. In the present research, an integrative strategy using UPLC-Q-TOF-MS combined with UPLC-QqQ-MS was employed to detect the absorbed constituents and investigate the pharmacokinetics of main compounds in the plasma after oral administration of YDXNT. UPLC-Q-TOF-MS was developed to detect the absorbed constituents and their metabolites in the plasma after oral administration in rats. A total of 52 constituents, including 44 prototype compounds and 8 metabolites, were identified or tentatively characterized. Then, nine main compounds (quercetin, isorhamnetin, kaempferol, ginkgolide A, ginkgolide B, ginkgolide C, bilobalide, tanshinone IIA, and salvianolic acid B) were chosen to further investigate the pharmacokinetic behavior of YDXNT using UPLC-QqQ-MS. The concentration of nine main constituents were in the range of 27.85–76.54 ng mL−1. This research provides a systematic approach for rapid qualitative analysis of absorbed constituents and for evaluating the pharmacokinetics of the main ingredients of YDXNT following its oral administration. More importantly, this work provides key information on the identification of bioactive compounds and the clarification of their action mechanisms, as well as on the pharmacological actions of YDXNT. Yindanxinnaotong capsule (YDXNT), a traditional Chinese formula, has been used to treat cardio-cerebrovascular diseases for several decades.![]()
Collapse
Affiliation(s)
- Leilei Gong
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Science
- Beijing
- P. R. China
| | - Haiyu Xu
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Science
- Beijing
- P. R. China
| | - Huijun Yuan
- School of Traditional Chinese Medicine
- Capital Medical University
- Beijing
- P. R. China
| | - Lan Wang
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Science
- Beijing
- P. R. China
| | - Xiaojie Yin
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Science
- Beijing
- P. R. China
| | - Moqi Fan
- The first clinical medical college of Beijing University of Chinese Medicine
- Beijing
- P. R. China
| | - Long Cheng
- Key Laboratory of Bioactive Substance and Resource Utilisation of the Chinese Herbal Medicine Ministry of Education
- Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing
| | - Xiaojing Ma
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Science
- Beijing
- P. R. China
| | - Rixin Liang
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Science
- Beijing
- P. R. China
| | - Hongjun Yang
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Science
- Beijing
- P. R. China
| |
Collapse
|
38
|
Abstract
There is a growing appreciation that metabolic processes and individual metabolites can shape the function of immune cells and thereby play important roles in the outcome of immune responses. In this respect, the use of MS- and NMR spectroscopy-based platforms to characterize and quantify metabolites in biological samples has recently yielded important novel insights into how our immune system functions and has contributed to the identification of biomarkers for immune-mediated diseases. Here, these recent immunological studies in which metabolomics has been used and made significant contributions to these fields will be discussed. In particular the role of metabolomics to the rapidly advancing field of cellular immunometabolism will be highlighted as well as the future prospects of such metabolomic tools in immunology.
Collapse
Affiliation(s)
- Bart Everts
- Department of Parasitology, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| |
Collapse
|