1
|
Perpiñá-Clérigues C, Mellado S, Català-Senent JF, Ibáñez F, Costa P, Marcos M, Guerri C, García-García F, Pascual M. Lipidomic landscape of circulating extracellular vesicles isolated from adolescents exposed to ethanol intoxication: a sex difference study. Biol Sex Differ 2023; 14:22. [PMID: 37085905 PMCID: PMC10120207 DOI: 10.1186/s13293-023-00502-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Lipids represent essential components of extracellular vesicles (EVs), playing structural and regulatory functions during EV biogenesis, release, targeting, and cell uptake. Importantly, lipidic dysregulation has been linked to several disorders, including metabolic syndrome, inflammation, and neurological dysfunction. Our recent results demonstrated the involvement of plasma EV microRNAs as possible amplifiers and biomarkers of neuroinflammation and brain damage induced by ethanol intoxication during adolescence. Considering the possible role of plasma EV lipids as regulatory molecules and biomarkers, we evaluated how acute ethanol intoxication differentially affected the lipid composition of plasma EVs in male and female adolescents and explored the participation of the immune response. METHODS Plasma EVs were extracted from humans and wild-type (WT) and Toll-like receptor 4 deficient (TLR4-KO) mice. Preprocessing and exploratory analyses were conducted after the extraction of EV lipids and data acquisition by mass spectrometry. Comparisons between ethanol-intoxicated and control human female and male individuals and ethanol-treated and untreated WT and TLR4-KO female and male mice were used to analyze the differential abundance of lipids. Annotation of lipids into their corresponding classes and a lipid set enrichment analysis were carried out to evaluate biological functions. RESULTS We demonstrated, for the first time, that acute ethanol intoxication induced a higher enrichment of distinct plasma EV lipid species in human female adolescents than in males. We observed a higher content of the PA, LPC, unsaturated FA, and FAHFA lipid classes in females, whereas males showed enrichment in PI. These lipid classes participate in the formation, release, and uptake of EVs and the activation of the immune response. Moreover, we observed changes in EV lipid composition between ethanol-treated WT and TLR4-KO mice (e.g., enrichment of glycerophosphoinositols in ethanol-treated WT males), and the sex-based differences in lipid abundance are more notable in WT mice than in TLR4-KO mice. All data and results generated have been made openly available on a web-based platform ( http://bioinfo.cipf.es/sal ). CONCLUSIONS Our results suggest that binge ethanol drinking in human female adolescents leads to a higher content of plasma EV lipid species associated with EV biogenesis and the propagation of neuroinflammatory responses than in males. In addition, we discovered greater differences in lipid abundance between sexes in WT mice compared to TLR4-KO mice. Our findings also support the potential use of EV-enriched lipids as biomarkers of ethanol-induced neuroinflammation during adolescence.
Collapse
Affiliation(s)
- Carla Perpiñá-Clérigues
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - José F Català-Senent
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Francesc Ibáñez
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - Pilar Costa
- Emergency Department, University Hospital of Salamanca-IBSAL, University of Salamanca, 37007, Salamanca, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, 37007, Salamanca, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain
| | - Francisco García-García
- Bioinformatics and Biostatistics Unit, Príncipe Felipe Research Center, C/ Eduardo Primo Yúfera, 3, 46012, Valencia, Spain.
| | - María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Avda. Blasco Ibáñez, 15, 46010, Valencia, Spain.
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012, Valencia, Spain.
| |
Collapse
|
2
|
Boncompagni G, Varone A, Tatangelo V, Capitani N, Frezzato F, Visentin A, Trentin L, Corda D, Baldari CT, Patrussi L. Glycerophosphoinositol Promotes Apoptosis of Chronic Lymphocytic Leukemia Cells by Enhancing Bax Expression and Activation. Front Oncol 2022; 12:835290. [PMID: 35392232 PMCID: PMC8980805 DOI: 10.3389/fonc.2022.835290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
An imbalance in the expression of pro- and anti-apoptotic members of the Bcl-2 family of apoptosis-regulating proteins is one of the main biological features of CLL, highlighting these proteins as therapeutic targets for treatment of this malignancy. Indeed, the Bcl-2 inhibitor Venetoclax is currently used for both first-line treatment and treatment of relapsed or refractory CLL. An alternative avenue is the transcriptional modulation of Bcl-2 family members to tilt their balance towards apoptosis. Glycerophosphoinositol (GroPIns) is a biomolecule generated from membrane phosphoinositides by the enzymes phospholipase A2 and lysolipase that pleiotropically affects key cellular functions. Mass-spectrometry analysis of GroPIns interactors recently highlighted the ability of GroPIns to bind to the non-receptor tyrosine phosphatase SHP-1, a known promoter of Bax expression, suggesting that GroPIns might correct the Bax expression defect in CLL cells, thereby promoting their apoptotic demise. To test this hypothesis, we cultured CLL cells in the presence of GroPIns, alone or in combination with drugs commonly used for treatment of CLL. We found that GroPIns alone increases Bax expression and apoptosis in CLL cells and enhances the pro-apoptotic activity of drugs used for CLL treatment in a SHP-1 dependent manner. Interestingly, among GroPIns interactors we found Bax itself. Short-term treatments of CLL cells with GroPIns induce Bax activation and translocation to the mitochondria. Moreover, GroPIns enhances the pro-apoptotic activity of Venetoclax and Fludarabine in CLL cells. These data provide evidence that GroPIns exploits two different pathways converging on Bax to promote apoptosis of leukemic cells and pave the way to new studies aimed at testing GroPIns in combination therapies for the treatment of CLL.
Collapse
Affiliation(s)
| | - Alessia Varone
- Institute of Endocrinology and Experimental Oncology “G. Salvatore”, National Research Council, Naples, Italy
| | | | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Federica Frezzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Daniela Corda
- Department of Biomedical Sciences, National Research Council, Rome, Italy
| | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
3
|
Campos AM, Nuzzo G, Varone A, Italiani P, Boraschi D, Corda D, Fontana A. Direct LC-MS/MS Analysis of Extra- and Intracellular Glycerophosphoinositol in Model Cancer Cell Lines. Front Immunol 2021; 12:646681. [PMID: 33737939 PMCID: PMC7960645 DOI: 10.3389/fimmu.2021.646681] [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: 12/27/2020] [Accepted: 02/04/2021] [Indexed: 12/20/2022] Open
Abstract
Glycerophosphoinositols (GPIs) are water-soluble bioactive phospholipid derivatives of increasing interest as intracellular and paracrine mediators of eukaryotic cell functions. The most representative compound of the family is glycerophosphoinositol (GroPIns), an ubiquitous component of mammalian cells that participates in cell proliferation, cell survival and cell response to stimuli. Levels and activity of this compound vary among cell types and deciphering these functions requires accurate measurements in in vitro and in vivo models. The conventional approaches for the analysis of GroPIns pose several issues in terms of sensitivity and product resolution, especially when the product is in the extracellular milieu. Here we present an UPLC-MS study for the quantitative analysis of this lipid derivative in cells and, for the first time, culture supernatants. The method is based on a solid-phase extraction that allows for fast desalting and analyte concentration. The robustness of the procedure was tested on the simultaneous measurements of intra- and extracellular levels of GroPIns in a number of human cell lines where it has been shown that the non-transformed cells are characterized by high extracellular level of GroPIns, whereas the tumor cells tended to have higher intracellular levels.
Collapse
Affiliation(s)
- Ana Margarida Campos
- Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy.,Consiglio Nazionale delle Ricerche, Institute of Biochemistry and Cell Biology, Naples, Italy
| | - Genoveffa Nuzzo
- Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Alessia Varone
- Consiglio Nazionale delle Ricerche, Institute of Biochemistry and Cell Biology, Naples, Italy
| | - Paola Italiani
- Consiglio Nazionale delle Ricerche, Institute of Biochemistry and Cell Biology, Naples, Italy
| | - Diana Boraschi
- Consiglio Nazionale delle Ricerche, Institute of Biochemistry and Cell Biology, Naples, Italy
| | - Daniela Corda
- Consiglio Nazionale delle Ricerche, Institute of Biochemistry and Cell Biology, Naples, Italy.,Consiglio Nazionale delle Ricerche, Department of Biomedical Sciences Rome, Italy
| | - Angelo Fontana
- Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy.,Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
4
|
Watkins OC, Yong HEJ, Sharma N, Chan SY. A review of the role of inositols in conditions of insulin dysregulation and in uncomplicated and pathological pregnancy. Crit Rev Food Sci Nutr 2020; 62:1626-1673. [PMID: 33280430 DOI: 10.1080/10408398.2020.1845604] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Inositols, a group of 6-carbon polyols, are highly bioactive molecules derived from diet and endogenous synthesis. Inositols and their derivatives are involved in glucose and lipid metabolism and participate in insulin-signaling, with perturbations in inositol processing being associated with conditions involving insulin resistance, dysglycemia and dyslipidemia such as polycystic ovary syndrome and diabetes. Pregnancy is similarly characterized by substantial and complex changes in glycemic and lipidomic regulation as part of maternal adaptation and is also associated with physiological alterations in inositol processing. Disruptions in maternal adaptation are postulated to have a critical pathophysiological role in pregnancy complications such as gestational diabetes and pre-eclampsia. Inositol supplementation has shown promise as an intervention for the alleviation of symptoms in conditions of insulin resistance and for gestational diabetes prevention. However, the mechanisms behind these affects are not fully understood. In this review, we explore the role of inositols in conditions of insulin dysregulation and in pregnancy, and identify priority areas for research. We particularly examine the role and function of inositols within the maternal-placental-fetal axis in both uncomplicated and pathological pregnancies. We also discuss how inositols may mediate maternal-placental-fetal cross-talk, and regulate fetal growth and development, and suggest that inositols play a vital role in promoting healthy pregnancy.
Collapse
Affiliation(s)
- Oliver C Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hannah E J Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
5
|
Tsutsumi T, Matsuda R, Morito K, Kawabata K, Yokota M, Nikawadori M, Inoue-Fujiwara M, Kawashima S, Hidaka M, Yamamoto T, Yamazaki N, Tanaka T, Shinohara Y, Nishi H, Tokumura A. Identification of human glycerophosphodiesterase 3 as an ecto phospholipase C that converts the G protein-coupled receptor 55 agonist lysophosphatidylinositol to bioactive monoacylglycerols in cultured mammalian cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158761. [PMID: 32629025 DOI: 10.1016/j.bbalip.2020.158761] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/30/2022]
Abstract
A family of glycerol-based lysolipid mediators comprises lysophosphatidic acid as a representative phospholipidic member but also a monoacylglycerol as a non-phosphorus-containing member. These critical lysolipid mediators are known to be produced from different lysophospholipids by actions of lysophospholipases C and D in mammals. Some members of the glycerophosphodiesterase (GDE) family have attracted recent attention due to their phospholipid-metabolizing activity. In this study, we found selective depletion of lysophosphatidylinositol among lysophospholipids in the culture medium of COS-7 cells transfected with a vector containing glycerophosphodiester phosphodiesterase 2 (GDPD2, GDE3). Thin-layer chromatography and liquid chromatography-tandem mass spectrometry of lipids extracted from GDE3-transfected COS-7 cells exposed to fluorescent analogs of phosphatidylinositol (PI) revealed that GDE3 acted as an ecto-type lysophospholipase C preferring endogenous lysophosphatidylinositol and PI having a long-chain acyl and a short-chain acyl group rather than endogenous PI and its fluorescent analog having two long chain acyl groups. In MC3T3-E1 cells cultured with an osteogenic or mitogenic medium, mRNA expression of GDE3 was increased by culturing in 10% fetal bovine serum for several days, concomitant with increased activity of ecto-lysophospholipase C, converting arachidonoyl-lysophosphatidylinositol, a physiological agonist of G protein-coupled receptor 55, to arachidonoylglycerol, a physiological agonist of cannabinoid receptors 1 and 2. We suggest that GDE3 acts as an ecto-lysophospholipase C, by switching signaling from lysophosphatidylinositol to that from arachidonoylglycerol in an opposite direction in mouse bone remodeling.
Collapse
Affiliation(s)
- Toshihiko Tsutsumi
- Graduate School of Clinical Pharmacy, Kyushu University of Health and Welfare, 1714-1 Yoshinomachi, Nobeoka 882-8508, Japan
| | - Risa Matsuda
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Katsuya Morito
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Kohei Kawabata
- Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan
| | - Miho Yokota
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Miki Nikawadori
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Manami Inoue-Fujiwara
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Satoshi Kawashima
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan; Institute of Advanced Medical Sciences, Tokushima University, Kuramotocho 3, Tokushima 770-8503, Japan
| | - Mayumi Hidaka
- Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan
| | - Takenori Yamamoto
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan; Institute of Advanced Medical Sciences, Tokushima University, Kuramotocho 3, Tokushima 770-8503, Japan
| | - Naoshi Yamazaki
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | - Tamotsu Tanaka
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan; Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan
| | - Yasuo Shinohara
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan; Institute of Advanced Medical Sciences, Tokushima University, Kuramotocho 3, Tokushima 770-8503, Japan
| | - Hiroyuki Nishi
- Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan
| | - Akira Tokumura
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan; Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan.
| |
Collapse
|
6
|
Patel D, Ahmad F, Kambach DM, Sun Q, Halim AS, Kramp T, Camphausen KA, Stommel JM. LXRβ controls glioblastoma cell growth, lipid balance, and immune modulation independently of ABCA1. Sci Rep 2019; 9:15458. [PMID: 31664073 PMCID: PMC6820787 DOI: 10.1038/s41598-019-51865-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 10/04/2019] [Indexed: 01/07/2023] Open
Abstract
Cholesterol is a critical component of membranes and a precursor for hormones and other signaling molecules. Previously, we showed that unlike astrocytes, glioblastoma cells do not downregulate cholesterol synthesis when plated at high density. In this report, we show that high cell density induces ABCA1 expression in glioblastoma cells, enabling them to get rid of excess cholesterol generated by an activated cholesterol biosynthesis pathway. Because oxysterols are agonists for Liver X Receptors (LXRs), we investigated whether increased cholesterol activates LXRs to maintain cholesterol homeostasis in highly-dense glioblastoma cells. We observed that dense cells had increased oxysterols, which activated LXRβ to upregulate ABCA1. Cells with CRISPR-mediated knockdown of LXRβ, but not ABCA1, had decreased cell cycle progression and cell survival, and decreased feedback repression of the mevalonate pathway in densely-plated glioma cells. LXRβ gene expression poorly correlates with ABCA1 in glioblastoma patients, and expression of each gene correlates with poor patient prognosis in different prognostic subtypes. Finally, gene expression and lipidomics analyses cells revealed that LXRβ regulates the expression of immune response gene sets and lipids known to be involved in immune modulation. Thus, therapeutic targeting of LXRβ in glioblastoma might be effective through diverse mechanisms.
Collapse
Affiliation(s)
- Deven Patel
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Fahim Ahmad
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Diane M Kambach
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qian Sun
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alan S Halim
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tamalee Kramp
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kevin A Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jayne M Stommel
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
7
|
Bocca C, Kane MS, Veyrat-Durebex C, Nzoughet JK, Chao de la Barca JM, Chupin S, Alban J, Procaccio V, Bonneau D, Simard G, Lenaers G, Reynier P, Chevrollier A. Lipidomics Reveals Triacylglycerol Accumulation Due to Impaired Fatty Acid Flux in Opa1-Disrupted Fibroblasts. J Proteome Res 2019; 18:2779-2790. [PMID: 31199663 DOI: 10.1021/acs.jproteome.9b00081] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OPA1 is a dynamin GTPase implicated in mitochondrial membrane fusion. Despite its involvement in lipid remodeling, the function of OPA1 has never been analyzed by whole-cell lipidomics. We used a nontargeted, reversed-phase lipidomics approach, validated for cell cultures, to investigate OPA1-inactivated mouse embryonic fibroblasts ( Opa1 -/- MEFs). This led to the identification of a wide range of 14 different lipid subclasses comprising 212 accurately detected lipids. Multivariate and univariate statistical analyses were then carried out to assess the differences between the Opa1 -/- and Opa1 +/+ genotypes. Of the 212 lipids identified, 69 were found to discriminate between Opa1 -/- MEFs and Opa1 +/+ MEFs. Among these lipids, 34 were triglycerides, all of which were at higher levels in Opa1 -/- MEFs with fold changes ranging from 3.60 to 17.93. Cell imaging with labeled fatty acids revealed a sharp alteration of the fatty acid flux with a reduced mitochondrial uptake. The other 35 discriminating lipids included phosphatidylcholines, lysophosphatidylcholines, phosphatidylethanolamine, and sphingomyelins, mainly involved in membrane remodeling, and ceramides, gangliosides, and phosphatidylinositols, mainly involved in apoptotic cell signaling. Our results show that the inactivation of OPA1 severely affects the mitochondrial uptake of fatty acids and lipids through membrane remodeling and apoptotic cell signaling.
Collapse
Affiliation(s)
- Cinzia Bocca
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France
| | - Mariame Selma Kane
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France
| | - Charlotte Veyrat-Durebex
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France.,Département de Biochimie et Génétique , Centre Hospitalier Universitaire , 49933 Angers , France
| | - Judith Kouassi Nzoughet
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France
| | - Juan Manuel Chao de la Barca
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France.,Département de Biochimie et Génétique , Centre Hospitalier Universitaire , 49933 Angers , France
| | - Stephanie Chupin
- Département de Biochimie et Génétique , Centre Hospitalier Universitaire , 49933 Angers , France
| | - Jennifer Alban
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France
| | - Vincent Procaccio
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France.,Département de Biochimie et Génétique , Centre Hospitalier Universitaire , 49933 Angers , France
| | - Dominique Bonneau
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France.,Département de Biochimie et Génétique , Centre Hospitalier Universitaire , 49933 Angers , France
| | - Gilles Simard
- Département de Biochimie et Génétique , Centre Hospitalier Universitaire , 49933 Angers , France.,INSERM U1063 , Université d'Angers , 49933 Angers , France
| | - Guy Lenaers
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France
| | - Pascal Reynier
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France.,Département de Biochimie et Génétique , Centre Hospitalier Universitaire , 49933 Angers , France
| | - Arnaud Chevrollier
- Equipe Mitolab, Institut MITOVASC, CNRS 6015, INSERM U1083 , Université d'Angers , 49933 Angers , France
| |
Collapse
|
8
|
Varone A, Mariggiò S, Patheja M, Maione V, Varriale A, Vessichelli M, Spano D, Formiggini F, Lo Monte M, Brancati N, Frucci M, Del Vecchio P, D'Auria S, Flagiello A, Iannuzzi C, Luini A, Pucci P, Banci L, Valente C, Corda D. A signalling cascade involving receptor-activated phospholipase A 2, glycerophosphoinositol 4-phosphate, Shp1 and Src in the activation of cell motility. Cell Commun Signal 2019; 17:20. [PMID: 30823936 PMCID: PMC6396489 DOI: 10.1186/s12964-019-0329-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/13/2019] [Indexed: 12/28/2022] Open
Abstract
Background Shp1, a tyrosine-phosphatase-1 containing the Src-homology 2 (SH2) domain, is involved in inflammatory and immune reactions, where it regulates diverse signalling pathways, usually by limiting cell responses through dephosphorylation of target molecules. Moreover, Shp1 regulates actin dynamics. One Shp1 target is Src, which controls many cellular functions including actin dynamics. Src has been previously shown to be activated by a signalling cascade initiated by the cytosolic-phospholipase A2 (cPLA2) metabolite glycerophosphoinositol 4-phosphate (GroPIns4P), which enhances actin polymerisation and motility. While the signalling cascade downstream Src has been fully defined, the mechanism by which GroPIns4P activates Src remains unknown. Methods Affinity chromatography, mass spectrometry and co-immunoprecipitation studies were employed to identify the GroPIns4P-interactors; among these Shp1 was selected for further analysis. The specific Shp1 residues interacting with GroPIns4P were revealed by NMR and validated by site-directed mutagenesis and biophysical methods such as circular dichroism, isothermal calorimetry, fluorescence spectroscopy, surface plasmon resonance and computational modelling. Morphological and motility assays were performed in NIH3T3 fibroblasts. Results We find that Shp1 is the direct cellular target of GroPIns4P. GroPIns4P directly binds to the Shp1-SH2 domain region (with the crucial residues being Ser 118, Arg 138 and Ser 140) and thereby promotes the association between Shp1 and Src, and the dephosphorylation of the Src-inhibitory phosphotyrosine in position 530, resulting in Src activation. As a consequence, fibroblast cells exposed to GroPIns4P show significantly enhanced wound healing capability, indicating that GroPIns4P has a stimulatory role to activate fibroblast migration. GroPIns4P is produced by cPLA2 upon stimulation by diverse receptors, including the EGF receptor. Indeed, endogenously-produced GroPIns4P was shown to mediate the EGF-induced cell motility. Conclusions This study identifies a so-far undescribed mechanism of Shp1/Src modulation that promotes cell motility and that is dependent on the cPLA2 metabolite GroPIns4P. We show that GroPIns4P is required for EGF-induced fibroblast migration and that it is part of a cPLA2/GroPIns4P/Shp1/Src cascade that might have broad implications for studies of immune-inflammatory response and cancer. ![]()
Electronic supplementary material The online version of this article (10.1186/s12964-019-0329-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alessia Varone
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.
| | - Stefania Mariggiò
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Manpreet Patheja
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Vincenzo Maione
- Magnetic Resonance Centre (CERM), University of Florence, 50019, Sesto Fiorentino, Italy
| | - Antonio Varriale
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.,Institute of Food Science, National Research Council, Via Roma 64, 83100, Avellino, Italy
| | - Mariangela Vessichelli
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Daniela Spano
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Fabio Formiggini
- Italian Institute of Technology, Centre for Advanced Biomaterials for Health Care at CRIB, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
| | - Matteo Lo Monte
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Nadia Brancati
- Institute of High Performance Computing and Networking, National Research Council, Via P. Castellino 111, 80131, Naples, Italy
| | - Maria Frucci
- Institute of High Performance Computing and Networking, National Research Council, Via P. Castellino 111, 80131, Naples, Italy
| | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy
| | - Sabato D'Auria
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.,Institute of Food Science, National Research Council, Via Roma 64, 83100, Avellino, Italy
| | - Angela Flagiello
- CEINGE Advanced Biotechnology, Via G. Salvatore 486, 80145, Naples, Italy
| | - Clara Iannuzzi
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.,Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. de Crecchio 7, 80138, Naples, Italy
| | - Alberto Luini
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Piero Pucci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126, Naples, Italy.,CEINGE Advanced Biotechnology, Via G. Salvatore 486, 80145, Naples, Italy
| | - Lucia Banci
- Magnetic Resonance Centre (CERM), University of Florence, 50019, Sesto Fiorentino, Italy
| | - Carmen Valente
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Daniela Corda
- Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.
| |
Collapse
|
9
|
Elich M, Sauer K. Regulation of Hematopoietic Cell Development and Function Through Phosphoinositides. Front Immunol 2018; 9:931. [PMID: 29780388 PMCID: PMC5945867 DOI: 10.3389/fimmu.2018.00931] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/16/2018] [Indexed: 01/01/2023] Open
Abstract
One of the most paramount receptor-induced signal transduction mechanisms in hematopoietic cells is production of the lipid second messenger phosphatidylinositol(3,4,5)trisphosphate (PIP3) by class I phosphoinositide 3 kinases (PI3K). Defective PIP3 signaling impairs almost every aspect of hematopoiesis, including T cell development and function. Limiting PIP3 signaling is particularly important, because excessive PIP3 function in lymphocytes can transform them and cause blood cancers. Here, we review the key functions of PIP3 and related phosphoinositides in hematopoietic cells, with a special focus on those mechanisms dampening PIP3 production, turnover, or function. Recent studies have shown that beyond “canonical” turnover by the PIP3 phosphatases and tumor suppressors phosphatase and tensin homolog (PTEN) and SH2 domain-containing inositol-5-phosphatase-1 (SHIP-1/2), PIP3 function in hematopoietic cells can also be dampened through antagonism with the soluble PIP3 analogs inositol(1,3,4,5)tetrakisphosphate (IP4) and inositol-heptakisphosphate (IP7). Other evidence suggests that IP4 can promote PIP3 function in thymocytes. Moreover, IP4 or the kinases producing it limit store-operated Ca2+ entry through Orai channels in B cells, T cells, and neutrophils to control cell survival and function. We discuss current models for how soluble inositol phosphates can have such diverse functions and can govern as distinct processes as hematopoietic stem cell homeostasis, neutrophil macrophage and NK cell function, and development and function of B cells and T cells. Finally, we will review the pathological consequences of dysregulated IP4 activity in immune cells and highlight contributions of impaired inositol phosphate functions in disorders such as Kawasaki disease, common variable immunodeficiency, or blood cancer.
Collapse
Affiliation(s)
- Mila Elich
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA, United States
| | - Karsten Sauer
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States.,Oncology R&D, Pfizer Worldwide R&D, San Diego, CA, United States
| |
Collapse
|
10
|
Vessichelli M, Mariggiò S, Varone A, Zizza P, Di Santo A, Amore C, Dell'Elba G, Cutignano A, Fontana A, Cacciapuoti C, Di Costanzo G, Zannini M, de Cristofaro T, Evangelista V, Corda D. The natural phosphoinositide derivative glycerophosphoinositol inhibits the lipopolysaccharide-induced inflammatory and thrombotic responses. J Biol Chem 2017; 292:12828-12841. [PMID: 28600357 PMCID: PMC5546025 DOI: 10.1074/jbc.m116.773861] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/22/2017] [Indexed: 12/25/2022] Open
Abstract
Inflammatory responses are elicited through lipid products of phospholipase A2 activity that acts on the membrane phospholipids, including the phosphoinositides, to form the proinflammatory arachidonic acid and, in parallel, the glycerophosphoinositols. Here, we investigate the role of the glycerophosphoinositol in the inflammatory response. We show that it is part of a negative feedback loop that limits proinflammatory and prothrombotic responses in human monocytes stimulated with lipopolysaccharide. This inhibition is exerted both on the signaling cascade initiated by the lipopolysaccharide with the glycerophosphoinositol-dependent decrease in IκB kinase α/β, p38, JNK, and Erk1/2 kinase phosphorylation and at the nuclear level with decreased NF-κB translocation and binding to inflammatory gene promoters. In a model of endotoxemia in the mouse, treatment with glycerophosphoinositol reduced TNF-α synthesis, which supports the concept that glycerophosphoinositol inhibits the de novo synthesis of proinflammatory and prothrombotic compounds and might thus have a role as an endogenous mediator in the resolution of inflammation. As indicated, this effect of glycerophosphoinositol can also be exploited in the treatment of manifestations of severe inflammation by exogenous administration of the compound.
Collapse
Affiliation(s)
- Mariangela Vessichelli
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, 80131 Naples, Italy
| | - Stefania Mariggiò
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, 80131 Naples, Italy
| | - Alessia Varone
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, 80131 Naples, Italy
| | - Pasquale Zizza
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, 80131 Naples, Italy
| | - Angelomaria Di Santo
- Laboratory of Vascular Biology and Pharmacology, Consorzio and Fondazione Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti, Italy
| | - Concetta Amore
- Laboratory of Vascular Biology and Pharmacology, Consorzio and Fondazione Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti, Italy
| | - Giuseppe Dell'Elba
- Laboratory of Vascular Biology and Pharmacology, Consorzio and Fondazione Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti, Italy
| | - Adele Cutignano
- Institute of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Angelo Fontana
- Institute of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Carmela Cacciapuoti
- Transfusion Service, Department of Hematology-Oncology and Stem Cell Transplantation Unit, National Cancer Institute G. Pascale Foundation, Istituto di Ricovero e Cura a Carattere Scientifico, Via M. Semmola 52, 80131 Naples, Italy
| | - Gaetano Di Costanzo
- Transfusion Service, Department of Hematology-Oncology and Stem Cell Transplantation Unit, National Cancer Institute G. Pascale Foundation, Istituto di Ricovero e Cura a Carattere Scientifico, Via M. Semmola 52, 80131 Naples, Italy
| | - Mariastella Zannini
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via S. Pansini 5, 80131 Naples, Italy
| | - Tiziana de Cristofaro
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via S. Pansini 5, 80131 Naples, Italy
| | - Virgilio Evangelista
- Laboratory of Vascular Biology and Pharmacology, Consorzio and Fondazione Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti, Italy.
| | - Daniela Corda
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, 80131 Naples, Italy.
| |
Collapse
|
11
|
Georgii E, Jin M, Zhao J, Kanawati B, Schmitt-Kopplin P, Albert A, Winkler JB, Schäffner AR. Relationships between drought, heat and air humidity responses revealed by transcriptome-metabolome co-analysis. BMC PLANT BIOLOGY 2017; 17:120. [PMID: 28693422 PMCID: PMC5504741 DOI: 10.1186/s12870-017-1062-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 06/25/2017] [Indexed: 05/17/2023]
Abstract
BACKGROUND Elevated temperature and reduced water availability are frequently linked abiotic stresses that may provoke distinct as well as interacting molecular responses. Based on non-targeted metabolomic and transcriptomic measurements from Arabidopsis rosettes, this study aims at a systematic elucidation of relevant components in different drought and heat scenarios as well as relationships between molecular players of stress response. RESULTS In combined drought-heat stress, the majority of single stress responses are maintained. However, interaction effects between drought and heat can be discovered as well; these relate to protein folding, flavonoid biosynthesis and growth inhibition, which are enhanced, reduced or specifically induced in combined stress, respectively. Heat stress experiments with and without supplementation of air humidity for maintenance of vapor pressure deficit suggest that decreased relative air humidity due to elevated temperature is an important component of heat stress, specifically being responsible for hormone-related responses to water deprivation. Remarkably, this "dry air effect" is the primary trigger of the metabolomic response to heat. In contrast, the transcriptomic response has a substantial temperature component exceeding the dry air component and including up-regulation of many transcription factors and protein folding-related genes. Data level integration independent of prior knowledge on pathways and condition labels reveals shared drought and heat responses between transcriptome and metabolome, biomarker candidates and co-regulation between genes and metabolic compounds, suggesting novel players in abiotic stress response pathways. CONCLUSIONS Drought and heat stress interact both at transcript and at metabolite response level. A comprehensive, non-targeted view of this interaction as well as non-interacting processes is important to be taken into account when improving tolerance to abiotic stresses in breeding programs. Transcriptome and metabolome may respond with different extent to individual stress components. Their contrasting behavior in response to temperature stress highlights that the protein folding machinery effectively shields the metabolism from stress. Disentangling the complex relationships between transcriptome and metabolome in response to stress is an enormous challenge. As demonstrated by case studies with supporting evidence from additional data, the large dataset provided in this study may assist in determining linked genetic and metabolic features as candidates for future mechanistic analyses.
Collapse
Affiliation(s)
- Elisabeth Georgii
- Helmholtz Zentrum München, Department of Environmental Sciences, Institute of Biochemical Plant Pathology, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Ming Jin
- Helmholtz Zentrum München, Department of Environmental Sciences, Institute of Biochemical Plant Pathology, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Jin Zhao
- Helmholtz Zentrum München, Department of Environmental Sciences, Institute of Biochemical Plant Pathology, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Basem Kanawati
- Helmholtz Zentrum München, Department of Environmental Sciences, Research Unit Analytical Biogeochemistry, Ingolstädter Landstr, 1, 85764, Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- Helmholtz Zentrum München, Department of Environmental Sciences, Research Unit Analytical Biogeochemistry, Ingolstädter Landstr, 1, 85764, Neuherberg, Germany
| | - Andreas Albert
- Helmholtz Zentrum München, Department of Environmental Sciences, Research Unit Environmental Simulation, Ingolstädter Landstr, 1, 85764, Neuherberg, Germany
| | - J Barbro Winkler
- Helmholtz Zentrum München, Department of Environmental Sciences, Research Unit Environmental Simulation, Ingolstädter Landstr, 1, 85764, Neuherberg, Germany
| | - Anton R Schäffner
- Helmholtz Zentrum München, Department of Environmental Sciences, Institute of Biochemical Plant Pathology, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
| |
Collapse
|
12
|
Xie T, Zhou X, Wang S, Lu Y, Zhu H, Kang A, Deng H, Xu J, Shen C, Di L, Shan J. Development and application of a comprehensive lipidomic analysis to investigate Tripterygium wilfordii-induced liver injury. Anal Bioanal Chem 2016; 408:4341-55. [PMID: 27086014 DOI: 10.1007/s00216-016-9533-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/17/2016] [Accepted: 03/30/2016] [Indexed: 12/16/2022]
Abstract
Lipid metabolic pathways play pivotal roles in liver function, and disturbances of these pathways are associated with various diseases. Thus, comprehensive characterization and measurement of lipid metabolites are essential to deciphering the contributions of lipid network metabolism to diseases or its responses to drug intervention. Here, we report an integrated lipidomic analysis for the comprehensive detection of lipid metabolites. To facilitate the characterization of untargeted lipids through fragmentation analysis, nine formulas were proposed to identify the fatty acid composition of lipids from complex MS (n) spectrum information. By these formulas, the co-eluted isomeric compounds could be distinguished. In total, 250 lipids were detected and characterized, including diacylglycerols, triacylglycerols, glycerophosphoethanolamines, glycerophosphocholines, glycerophosphoserines, glycerophosphoglycerols, glycerophosphoinositols, cardiolipins, ceramides, and sphingomyelins. Integrated with the targeted lipidomics, a total of 27 inflammatory oxylipins were also measured. To evaluate the aberrant lipid metabolism involved in liver injury induced by Tripterygium wilfordii, lipid network metabolism was further investigated. Results indicated that energy lipid modification, membrane remodeling, potential signaling lipid alterations, and abnormal inflammation response were associated with injury. Because of the important roles of lipids in liver metabolism, this new method is expected to be useful in analyzing other lipid metabolism diseases.
Collapse
Affiliation(s)
- Tong Xie
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China.
| | - Xueping Zhou
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Shouchuan Wang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yan Lu
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Huaxu Zhu
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - An Kang
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Haishan Deng
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Jianya Xu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Cunsi Shen
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Liuqing Di
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
13
|
Lipidomic profiling of tryptophan hydroxylase 2 knockout mice reveals novel lipid biomarkers associated with serotonin deficiency. Anal Bioanal Chem 2016; 408:2963-73. [DOI: 10.1007/s00216-015-9256-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/21/2015] [Accepted: 12/07/2015] [Indexed: 12/22/2022]
|
14
|
Sauer K, Okkenhaug K. Editorial: Lipid Signaling in T Cell Development and Function. Front Immunol 2015; 6:410. [PMID: 26322043 PMCID: PMC4530596 DOI: 10.3389/fimmu.2015.00410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/27/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Karsten Sauer
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| |
Collapse
|
15
|
An improved UPLC-MS/MS platform for quantitative analysis of glycerophosphoinositol in mammalian cells. PLoS One 2015; 10:e0123198. [PMID: 25860666 PMCID: PMC4393254 DOI: 10.1371/journal.pone.0123198] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/19/2015] [Indexed: 02/02/2023] Open
Abstract
The glycerophosphoinositols constitute a class of biologically active lipid-derived mediators whose intracellular levels are modulated during physiological and pathological cell processes. Comprehensive assessment of the role of these compounds expands beyond the cellular biology of lipids and includes rapid and unambiguous measurement in cells and tissues. Here we describe a sensitive and simple liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for quantitative analysis of the most abundant among these phosphoinositide derivatives in mammalian cells, the glycerophosphoinositol (GroPIns). The method has been developed in mouse Raw 264.7 macrophages with limits of quantitation at 3 ng/ml. Validation on the same cell line showed excellent response in terms of linear dynamic range (from 3 to 3,000 ng/ml), intra-day and inter-day precision (coefficient of variation ≤ 7.10%) and accuracy (between 98.1 and 109.0%) in the range 10-320 ng/ml. As proof of concept, a simplified analytical platform based on this method and external calibration was also tested on four stimulated and unstimulated cell lines, including Raw 264.7 macrophages, Jurkat T-cells, A375MM melanoma cells and rat basophilic leukemia RBL-2H3 cells. The results indicate a wide variation in GroPIns levels among different cell lines and stimulation conditions, although the measurements were always in line with the literature. No significant matrix effects were observed thus indicating that the here proposed method can be of general use for similar determinations in cells of different origin.
Collapse
|
16
|
De Luca AC, Reader-Harris P, Mazilu M, Mariggiò S, Corda D, Di Falco A. Reproducible surface-enhanced Raman quantification of biomarkers in multicomponent mixtures. ACS NANO 2014; 8:2575-2583. [PMID: 24524333 DOI: 10.1021/nn406200y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Direct and quantitative detection of unlabeled glycerophosphoinositol (GroPIns), an abundant cytosolic phosphoinositide derivative, would allow rapid evaluation of several malignant cell transformations. Here we report label-free analysis of GroPIns via surface-enhanced Raman spectroscopy (SERS) with a sensitivity of 200 nM, well below its apparent concentration in cells. Crucially, our SERS substrates, based on lithographically defined gold nanofeatures, can be used to predict accurately the GroPIns concentration even in multicomponent mixtures, avoiding the preliminary separation of individual compounds. Our results represent a critical step toward the creation of SERS-based biosensor for rapid, label-free, and reproducible detection of specific molecules, overcoming limits of current experimental methods.
Collapse
Affiliation(s)
- Anna Chiara De Luca
- Institute of Protein Biochemistry, National Research Council , Via P. Castellino 111, 80131 Naples, Italy
| | | | | | | | | | | |
Collapse
|