Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38514392

ABSTRACT

OBJECTIVE: Chronic inflammatory diseases, like Systemic Lupus Erythematosus (SLE), carry an increased risk for atherosclerosis and cardiovascular events, accompanied by impairment of atheroprotective properties of high-density lipoprotein (HDL). In SLE, serum BAFF (B cell-activating factor), a cytokine implicated in disease progression, has been correlated with subclinical atherosclerosis. We investigated the impact of treatment with belimumab -an anti-BAFF monoclonal antibody- on HDL atheroprotective properties and composition in SLE patients. METHODS: Serum samples were collected from 35 SLE patients with active disease despite conventional therapy, before and after 6-month add-on treatment with belimumab, and 26 matched healthy individuals. We measured cholesterol efflux and antioxidant capacities, paraoxonase-1 activity, serum amyloid A1, myeloperoxidase and lipid peroxidation product levels of HDL. LC-MS/MS was performed to analyze the HDL lipidome. RESULTS: Following treatment with belimumab, cholesterol efflux and antioxidant capacities of HDL were significantly increased in SLE patients and restored to levels of controls. HDL-associated paraoxonase-1 activity was also increased, whereas lipid peroxidation products were decreased following treatment. HDL cholesterol efflux and antioxidant capacities correlated negatively with the disease activity. Changes were noted in the HDL lipidome of SLE patients following belimumab treatment, as well as between SLE patients and healthy individuals, and specific changes in lipid species correlated with functional parameters of HDL. CONCLUSIONS: HDL of SLE patients with active disease displays impaired atheroprotective properties accompanied by distinct lipidomic signature compared with controls. Belimumab treatment may improve the HDL atheroprotective properties and modify the HDL lipidomic signature in SLE patients, thus potentially mitigating atherosclerosis development.

2.
Sci Rep ; 13(1): 21778, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38066223

ABSTRACT

The lipidome of equine BALF cells has not been described. The objectives of this prospective repeated-measures study were to explore the BALF cells' lipidome in horses and to identify lipids associated with progression or resolution of airway inflammation. BALF cells from 22 horses exposed to two bedding materials (Peat 1-Wood shavings [WS]-Peat 2) were studied by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The effects of bedding on lipid class and species compositions were tested with rmANOVA. Correlations between lipids and cell counts were examined. The BALF cells' lipidome showed bedding-related differences for molar percentage (mol%) of 60 species. Whole phosphatidylcholine (PC) class and its species PC 32:0 (main molecular species 16:0_16:0) had higher mol% after Peat 2 compared with WS. Phosphatidylinositol 38:4 (main molecular species 18:0_20:4) was higher after WS compared with both peat periods. BALF cell count correlated positively with mol% of the lipid classes phosphatidylserine, sphingomyelin, ceramide, hexosylceramide, and triacylglycerol but negatively with PC. BALF cell count correlated positively with phosphatidylinositol 38:4 mol%. In conclusion, equine BALF cells' lipid profiles explored with MS-based lipidomics indicated subclinical inflammatory changes after WS. Inflammatory reactions in the cellular lipid species composition were detected although cytological responses indicating inflammation were weak.


Subject(s)
Inflammation , Tandem Mass Spectrometry , Animals , Horses , Bronchoalveolar Lavage Fluid/chemistry , Chromatography, Liquid , Prospective Studies , Inflammation/veterinary , Soil , Phosphatidylinositols , Bedding and Linens , Bronchoalveolar Lavage
3.
Proc Natl Acad Sci U S A ; 120(30): e2210599120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37463214

ABSTRACT

Cardiolipin (CL) is an essential phospholipid for mitochondrial structure and function. Here, we present a small mitochondrial protein, NERCLIN, as a negative regulator of CL homeostasis and mitochondrial ultrastructure. Primate-specific NERCLIN is expressed ubiquitously from the GRPEL2 locus on a tightly regulated low level. NERCLIN overexpression severely disrupts mitochondrial cristae structure and induces mitochondrial fragmentation. Proximity labeling and immunoprecipitation analysis suggested interactions of NERCLIN with CL synthesis and prohibitin complexes on the matrix side of the inner mitochondrial membrane. Lipid analysis indicated that NERCLIN regulates mitochondrial CL content. Furthermore, NERCLIN is responsive to heat stress ensuring OPA1 processing and cell survival. Thus, we propose that NERCLIN contributes to the stress-induced adaptation of mitochondrial dynamics. Our findings add NERCLIN to the group of recently identified small mitochondrial proteins with important regulatory functions.


Subject(s)
Cardiolipins , Mitochondrial Proteins , Animals , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Cardiolipins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Homeostasis
4.
Sci Rep ; 13(1): 9821, 2023 06 17.
Article in English | MEDLINE | ID: mdl-37330591

ABSTRACT

Equine asthma (EA) is an inflammatory disease of the lower airways driven by mediators released from cells. Extracellular vesicles (EVs) are vehicles for lipid mediators, which possess either pro-inflammatory or dual anti-inflammatory and pro-resolving functions. In this study, we investigated how the respiratory fatty acid (FA) profile reflects airway inflammatory status. The FA composition of bronchoalveolar lavage fluid (BALF), BALF supernatant, and bronchoalveolar EVs of healthy horses (n = 15) and horses with mild/moderate EA (n = 10) or severe EA (SEA, n = 5) was determined with gas chromatography and mass spectrometry. The FA profiles distinguished samples with different diagnoses in all sample types, yet they were insufficient to predict the health status of uncategorized samples. Different individual FAs were responsible for the discrimination of the diagnoses in different sample types. Particularly, in the EVs of SEA horses the proportions of palmitic acid (16:0) decreased and those of eicosapentaenoic acid (20:5n-3) increased, and all sample types of asthmatic horses had elevated dihomo-γ-linolenic acid (20:3n-6) proportions. The results suggest simultaneous pro-inflammatory and resolving actions of FAs and a potential role for EVs as vehicles for lipid mediators in asthma pathogenesis. EV lipid manifestations of EA can offer translational targets to study asthma pathophysiology and treatment options.


Subject(s)
Asthma , Extracellular Vesicles , Horse Diseases , Animals , Horses , Bronchoalveolar Lavage Fluid/chemistry , Fatty Acids , Gas Chromatography-Mass Spectrometry , Asthma/diagnosis , Asthma/veterinary , Horse Diseases/diagnosis , Bronchoalveolar Lavage
6.
J Clin Lipidol ; 15(5): 743-751, 2021.
Article in English | MEDLINE | ID: mdl-34548243

ABSTRACT

BACKGROUND: There is little knowledge on the effects of alpha-linolenic acid (ALA) and n-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA) on the LDL lipidome and aggregation of LDL particles. OBJECTIVE: We examined if consumption of Camelina sativa oil (CSO) as a source of ALA, fatty fish (FF) as a source of n-3 LCPUFA and lean fish (LF) as a source of fish protein affect the lipidome of LDL as compared to a control diet. METHODS: Participants with impaired glucose tolerance (39 women and 40 men) were randomized to 4 study groups (CSO providing 10 g/d ALA, FF and LF [both 4 fish meals/wk] and control limiting their fish and ALA intake) in a 12-week, parallel trial. Diets were instructed and dietary fats were provided to the participants. The lipidome of LDL particles isolated from samples collected at baseline and after intervention was analyzed with electrospray ionization-tandem mass spectrometry. RESULTS: In the CSO group, the relative concentrations of saturated and monounsaturated cholesteryl ester species in LDL decreased and the species with ALA increased. In the FF group, LDL phosphatidylcholine (PC) species containing n-3 LCPUFA increased. There was a significant positive correlation between the change in total sphingomyelin and change in LDL aggregation, while total PC and triunsaturated PC species were inversely associated with LDL aggregation when all the study participants were included in the analysis. CONCLUSION: Dietary intake of CSO and FF modifies the LDL lipidome to contain more polyunsaturated and less saturated lipid species. The LDL surface lipids are associated with LDL aggregation.


Subject(s)
Camellia/chemistry , Dietary Fats, Unsaturated/administration & dosage , Eating/physiology , Fatty Acids, Omega-3/administration & dosage , Fish Oils/administration & dosage , Fishes , Glucose Intolerance/metabolism , Lipoproteins, LDL/metabolism , Plant Oils/administration & dosage , alpha-Linolenic Acid/administration & dosage , Aged , Animals , Female , Glucose Intolerance/blood , Humans , Lipoproteins, LDL/blood , Male , Middle Aged , Protein Aggregates , Spectrometry, Mass, Electrospray Ionization
7.
Am J Physiol Gastrointest Liver Physiol ; 321(3): G298-G307, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34259586

ABSTRACT

Drug-induced liver injury is an emerging form of acute and chronic liver disease that may manifest as fatty liver. Amiodarone (AMD), a widely used antiarrhythmic drug, can cause hepatic injury and steatosis by a variety of mechanisms, not all completely understood. We hypothesized that repetitive AMD administration may induce hepatic lipotoxicity not only via effects on the liver but also via effects on adipose tissue. Indeed, repetitive AMD administration induced endoplasmic reticulum (ER) stress in both liver and adipose tissue. In adipose tissue, AMD reduced lipogenesis and increased lipolysis. Moreover, AMD treatment induced ER stress and ER stress-dependent lipolysis in 3T3L1 adipocytes in vitro. In the liver, AMD caused increased expression of genes encoding proteins involved in fatty acid (FA) uptake and transfer (Cd36, Fabp1, and Fabp4), and resulted in increased hepatic accumulation of free FAs, but not of triacylglycerols. In line with this, there was increased expression of hepatic de novo FA synthesis genes. However, AMD significantly reduced the expression of the desaturase Scd1 and elongase Elovl6, detected at mRNA and protein levels. Accordingly, the FA profile of hepatic total lipids revealed increased accumulation of palmitate, an SCD1 and ELOVL6 substrate, and reduced levels of palmitoleate and cis-vaccenate, products of the enzymes. In addition, AMD-treated mice displayed increased hepatic apoptosis. The studies show that repetitive AMD induces ER stress and aggravates lipolysis in adipose tissue while inducing a lipotoxic hepatic lipid environment, suggesting that AMD-induced liver damage is due to compound insult to liver and adipose tissue.NEW & NOTEWORTHY AMD chronic administration induces hepatic lipid accumulation by several mechanisms, including induction of hepatic ER stress, impairment of ß-oxidation, and inhibition of triacylglycerol secretion. Our study shows that repetitive AMD treatment induces not only hepatic ER stress but also adipose tissue ER stress and lipolysis and hepatic accumulation of free fatty acids and enrichment of palmitate in the total lipids. Understanding the toxicity mechanisms of AMD would help devise ways to limit liver damage.


Subject(s)
Amiodarone/pharmacology , Endoplasmic Reticulum Stress/drug effects , Fatty Acids, Nonesterified/metabolism , Fatty Acids/metabolism , Lipolysis/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Amiodarone/metabolism , Animals , Fatty Acid-Binding Proteins/metabolism , Fatty Liver/metabolism , Lipid Metabolism/drug effects , Lipogenesis/physiology , Liver/drug effects , Liver/metabolism , Liver Diseases/metabolism , Mice , Triglycerides/metabolism
8.
Cells ; 9(9)2020 09 22.
Article in English | MEDLINE | ID: mdl-32972000

ABSTRACT

Human mesenchymal stromal/stem cells (hMSCs) show great promise in cell therapy due to their immunomodulatory properties. The overall immunomodulatory response of hMSCs resembles the resolution of inflammation, in which lipid mediators and regulatory macrophages (Mregs) play key roles. We investigated the effect of hMSC cell-cell contact and secretome on macrophages polarized and activated toward Mreg phenotype. Moreover, we studied the effect of supplemented polyunsaturated fatty acids (PUFAs): docosahexaenoic acid (DHA) and arachidonic acid, the precursors of lipid mediators, on hMSC immunomodulation. Our results show that unlike hMSC cell-cell contact, the hMSC secretome markedly increased the CD206 expression in both Mreg-polarized and Mreg-activated macrophages. Moreover, the secretome enhanced the expression of programmed death-ligand 1 on Mreg-polarized macrophages and Mer receptor tyrosine kinase on Mreg-activated macrophages. Remarkably, these changes were translated into improved Candida albicans phagocytosis activity of macrophages. Taken together, these results demonstrate that the hMSC secretome promotes the immunoregulatory and proresolving phenotype of Mregs. Intriguingly, DHA supplementation to hMSCs resulted in a more potentiated immunomodulation with increased CD163 expression and decreased gene expression of matrix metalloproteinase 2 in Mreg-polarized macrophages. These findings highlight the potential of PUFA supplementations as an easy and safe method to improve the hMSC therapeutic potential.


Subject(s)
Arachidonic Acid/pharmacology , Cell Communication/immunology , Docosahexaenoic Acids/pharmacology , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Phagocytosis/drug effects , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Candida albicans/growth & development , Candida albicans/immunology , Cell Communication/drug effects , Cell Polarity/drug effects , Gene Expression Regulation/drug effects , Humans , Immunomodulation/drug effects , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-23/genetics , Interleukin-23/immunology , Macrophage Activation/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/microbiology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mesenchymal Stem Cells/cytology , Phenotype , Primary Cell Culture , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/immunology
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(10): 1350-1362, 2019 10.
Article in English | MEDLINE | ID: mdl-31207356

ABSTRACT

Human mesenchymal stromal/stem cells (hMSCs) are used in experimental cell therapy to treat various immunological disorders, and the extracellular vesicles (hMSC-EVs) they produce have emerged as an option for cell-free therapeutics. The immunomodulatory function of hMSCs resembles the resolution of inflammation, in which proresolving lipid mediators (LMs) play key roles. Multiple mechanisms underlying the hMSC immunosuppressive effect has been elucidated; however, the impact of LMs and EVs in the resolution is poorly understood. In this study, we supplemented hMSCs with polyunsaturated fatty acids (PUFAs); arachidonic acid, eicosapentaenoic acid, and docosahexaenoic acid, which serve as precursors for multiple LMs. We then determined the consequent compositional modifications in the fatty acid, phospholipid, and LM profiles. Mass spectrometric analyses revealed that the supplemented PUFAs were incorporated into the main membrane phospholipid classes with different dynamics, with phosphatidylcholine serving as the first acceptor. Most importantly, the PUFA modifications were transferred into hMSC-EVs, which are known to mediate hMSC immunomodulation. Furthermore, the membrane-incorporated PUFAs influenced the LM profile by increasing the production of downstream prostaglandin E2 and proresolving LMs, including Resolvin E2 and Resolvin D6. The production of LMs was further enhanced by a highly proinflammatory stimulus, which resulted in an increase in a number of mediators, most notably prostaglandins, while other stimulatory conditions had less a pronounced impact after a 48-h incubation. The current findings suggest that PUFA manipulations of hMSCs exert significant immunomodulatory effects via EVs and proresolving LMs, the composition of which can be modified to potentiate the therapeutic impact of hMSCs.


Subject(s)
Extracellular Vesicles/metabolism , Fatty Acids, Unsaturated/metabolism , Inflammation Mediators/metabolism , Mesenchymal Stem Cells/metabolism , Cells, Cultured , Dinoprostone/metabolism , Fatty Acids/metabolism , Humans , Phospholipids/metabolism
10.
EMBO Mol Med ; 11(6)2019 06.
Article in English | MEDLINE | ID: mdl-31068339

ABSTRACT

The current clinical care of glioblastomas leaves behind invasive, radio- and chemo-resistant cells. We recently identified mammary-derived growth inhibitor (MDGI/FABP3) as a biomarker for invasive gliomas. Here, we demonstrate a novel function for MDGI in the maintenance of lysosomal membrane integrity, thus rendering invasive glioma cells unexpectedly vulnerable to lysosomal membrane destabilization. MDGI silencing impaired trafficking of polyunsaturated fatty acids into cells resulting in significant alterations in the lipid composition of lysosomal membranes, and subsequent death of the patient-derived glioma cells via lysosomal membrane permeabilization (LMP). In a preclinical model, treatment of glioma-bearing mice with an antihistaminergic LMP-inducing drug efficiently eradicated invasive glioma cells and secondary tumours within the brain. This unexpected fragility of the aggressive infiltrating cells to LMP provides new opportunities for clinical interventions, such as re-positioning of an established antihistamine drug, to eradicate the inoperable, invasive, and chemo-resistant glioma cells from sustaining disease progression and recurrence.


Subject(s)
Biomarkers, Tumor/metabolism , Fatty Acid Binding Protein 3/metabolism , Glioblastoma , Intracellular Membranes , Lysosomes , Neoplasm Proteins/metabolism , Animals , Cell Line, Tumor , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Heterografts , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/pathology , Lysosomes/metabolism , Lysosomes/pathology , Male , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Permeability , Xenograft Model Antitumor Assays
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(8): 1168-1182, 2019 08.
Article in English | MEDLINE | ID: mdl-30980920

ABSTRACT

Platelets are collected for transfusion to patients with different haematological disorders, and for logistical reasons, platelets are stored as concentrates. Despite carefully controlled conditions, platelets become activated during storage, and platelet concentrates (PlaCs) may cause adverse inflammatory reactions in recipients. The time-dependent changes in the lipidome of clinical PlaCs, platelets isolated from PlaCs, and extracellular vesicles (EVs) thereof were examined by mass spectrometry. The relative amount of arachidonic acid containing glycerophospholipids, especially those in the phosphatidylethanolamine and phosphatidylserine classes during storage, but the relative amount of other polyunsaturated fatty acid containing glycerophospholipids remained stable in all sample types. These changes were not directly translated to lipid mediator (LM) profile since the levels of arachidonic acid-derived proinflammatory LMs were not specifically elevated. Instead, several monohydroxy pathway markers and functionally relevant LMs, both proinflammatory and proresolving, were detected in the PlaCs and the EVs, and some representatives of both kind clearly accumulated during storage. By Western blot, the key enzymes of these pathways were shown to be present in platelets, and in many cases, EVs. Since the EVs were enriched in the fatty acid precursors of LMs in their (phospholipid) membranes, harboured LM-producing enzymes, contained the related monohydroxy pathway markers, and secreted the final LM products, PlaC-derived EVs could participate in the regulation of inflammation and healing, and thereby aid the platelets in exerting their essential physiological functions.


Subject(s)
Blood Platelets/cytology , Blood Preservation , Extracellular Vesicles/chemistry , Glycerophospholipids/analysis , Cell Membrane/chemistry , Extracellular Vesicles/physiology , Humans , Inflammation Mediators/analysis , Mass Spectrometry/methods , Platelet Transfusion/adverse effects , Platelet Transfusion/standards
12.
Front Immunol ; 9: 771, 2018.
Article in English | MEDLINE | ID: mdl-29706969

ABSTRACT

Resolution-phase macrophage population orchestrates active dampening of the inflammation by secreting anti-inflammatory and proresolving products including interleukin (IL)-10 and lipid mediators (LMs). We investigated the effects of both human bone marrow-derived mesenchymal stromal cells (MSCs) and MSC-derived extracellular vesicles (MSC-EVs) on mature human regulatory macrophages (Mregs). The cytokines and LMs were determined from cell culture media of Mregs cultivated with MSCs and MSC-EVs. In addition, the alterations in the expression of cell surface markers and the phagocytic ability of Mregs were investigated. Our novel findings indicate that both MSC coculture and MSC-EVs downregulated the production of IL-23 and IL-22 enhancing the anti-inflammatory phenotype of Mregs and amplifying proresolving properties. The levels of prostaglandin E2 (PGE2) were substantially upregulated in MSC coculture media, which may endorse proresolving LM class switching. In addition, our results manifest, for the first time, that MSC-EVs mediate the Mreg phenotype change via PGE2. These data suggest that both human MSC and MSC-EVs may potentiate tolerance-promoting proresolving phenotype of human Mregs.


Subject(s)
Extracellular Vesicles/immunology , Interleukin-23/biosynthesis , Interleukins/biosynthesis , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Down-Regulation , Humans , Immune Tolerance/immunology , Inflammation/immunology , Inflammation/metabolism , Macrophages/metabolism , Phenotype , Interleukin-22
13.
J Lipid Res ; 58(1): 92-110, 2017 01.
Article in English | MEDLINE | ID: mdl-27856675

ABSTRACT

High arachidonic acid (20:4n-6) and low n-3 PUFA levels impair the capacity of cultured human bone marrow mesenchymal stromal cells (hBMSCs) to modulate immune functions. The capacity of the hBMSCs to modify PUFA structures was found to be limited. Therefore, different PUFA supplements given to the cells resulted in very different glycerophospholipid (GPL) species profiles and substrate availability for phospholipases, which have preferences for polar head group and acyl chains when liberating PUFA precursors for production of lipid mediators. When supplemented with 20:4n-6, the cells increased prostaglandin E2 secretion. However, they elongated 20:4n-6 to the less active precursor, 22:4n-6, and also incorporated it into triacylglycerols, which may have limited the proinflammatory signaling. The n-3 PUFA precursor, 18:3n-3, had little potency to reduce the GPL 20:4n-6 content, while the eicosapentaenoic (20:5n-3) and docosahexaenoic (22:6n-3) acid supplements efficiently displaced the 20:4n-6 acyls, and created diverse GPL species substrate pools allowing attenuation of inflammatory signaling. The results emphasize the importance of choosing appropriate PUFA supplements for in vitro hBMSC expansion and suggests that for optimal function they require an exogenous fatty acid source providing 20:5n-3 and 22:6n-3 sufficiently, but 20:4n-6 moderately, which calls for specifically designed optimal PUFA supplements for the cultures.


Subject(s)
Fatty Acids, Omega-3/metabolism , Fatty Acids, Unsaturated/metabolism , Inflammation/metabolism , Mesenchymal Stem Cells/metabolism , Phospholipids/metabolism , Arachidonic Acid/metabolism , Bone Marrow Cells/metabolism , Cell Line , Dietary Supplements , Dinoprostone/genetics , Dinoprostone/metabolism , Eicosapentaenoic Acid/metabolism , Fatty Acids, Unsaturated/genetics , Glycerophospholipids/metabolism , Humans , Immunomodulation/genetics , Inflammation/pathology , Mass Spectrometry , Phospholipids/genetics , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...