Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
J Colloid Interface Sci ; 669: 537-551, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38729002

ABSTRACT

Infectious diseases, particularly those associated with biofilms, are challenging to treat due to an increased tolerance to commonly used antibiotics. This underscores the urgent need for innovative antimicrobial strategies. Here, we present an alternative simple-by-design approach focusing on the development of biocompatible and antibiotic-free nanocarriers from docosahexaenoic acid (DHA) that has the potential to combat microbial infections and phosphatidylglycerol (DOPG), which is attractive for use as a biocompatible prominent amphiphilic component of Gram-positive bacterial cell membranes. We assessed the anti-bacterial and anti-biofilm activities of these nanoformulations (hexosomes and vesicles) against S. aureus and S. epidermidis, which are the most common causes of infections on catheters and medical devices by different methods (including resazurin assay, time-kill assay, and confocal laser scanning microscopy on an in vitro catheter biofilm model). In a DHA-concentration-dependent manner, these nano-self-assemblies demonstrated strong anti-bacterial and anti-biofilm activities, particularly against S. aureus. A five-fold reduction of the planktonic and a four-fold reduction of biofilm populations of S. aureus were observed after treatment with hexosomes. The nanoparticles had a bacteriostatic effect against S. epidermidis planktonic cells but no anti-biofilm activity was detected. We discuss the findings in terms of nanoparticle-bacterial cell interactions, plausible alterations in the phospholipid membrane composition, and potential penetration of DHA into these membranes, leading to changes in their structural and biophysical properties. The implications for the future development of biocompatible nanocarriers for the delivery of DHA alone or in combination with other anti-bacterial agents are discussed, as novel treatment strategies of Gram-positive infections, including biofilm-associated infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Docosahexaenoic Acids , Microbial Sensitivity Tests , Nanoparticles , Phosphatidylglycerols , Staphylococcus aureus , Staphylococcus epidermidis , Biofilms/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/pharmacology , Staphylococcus aureus/drug effects , Nanoparticles/chemistry , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacology , Staphylococcus epidermidis/drug effects , Liquid Crystals/chemistry , Particle Size
2.
Int J Antimicrob Agents ; 62(3): 106888, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37328075

ABSTRACT

The rapid emergence and spread of multi-drug- or pan-drug-resistant bacterial pathogens, such as ESKAPE, pose a serious threat to global health. However, the development of novel antibiotics is hindered by difficulties in identifying new antibiotic targets and the rapid development of drug resistance. Drug repurposing is an effective alternative strategy for combating antibiotic resistance that both saves resources and extends the life of existing antibiotics in combination treatment regimens. Screening of a chemical compound library identified BMS-833923 (BMS), a smoothened antagonist that kills Gram-positive bacteria directly, and potentiates colistin to destroy various Gram-negative bacteria. BMS did not induce detectable antibiotic resistance in vitro, and showed effective activity against drug-resistant bacteria in vivo. Mechanistic studies revealed that BMS caused membrane disruption by targeting the membrane phospholipids phosphatidylglycerol and cardiolipin, promoting membrane dysfunction, metabolic disturbance, leakage of cellular components, and, ultimately, cell death. This study describes a potential strategy to enhance the efficacy of colistin and combat multi-drug-resistant ESKAPE pathogens.


Subject(s)
Colistin , Hedgehog Proteins , Colistin/pharmacology , Colistin/metabolism , Hedgehog Proteins/pharmacology , Phosphatidylglycerols/pharmacology , Drug Repositioning , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Gram-Negative Bacteria , Adjuvants, Immunologic , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests
3.
Immunol Rev ; 317(1): 166-186, 2023 08.
Article in English | MEDLINE | ID: mdl-37144896

ABSTRACT

The pulmonary surfactant system of the lung is a lipid and protein complex, which regulates the biophysical properties of the alveoli to prevent lung collapse and the innate immune system in the lung. Pulmonary surfactant is a lipoprotein complex consisting of 90% phospholipids and 10% protein, by weight. Two minor components of pulmonary surfactant phospholipids, phosphatidylglycerol (PG) and phosphatidylinositol (PI), exist at very high concentrations in the extracellular alveolar compartments. We have reported that one of the most dominant molecular species of PG, palmitoyl-oleoyl-phosphatidylglycerol (POPG) and PI inhibit inflammatory responses induced by multiple toll-like receptors (TLR2/1, TLR3, TLR4, and TLR2/6) by interacting with subsets of multiprotein receptor components. These lipids also exert potent antiviral effects against RSV and influenza A, in vitro, by inhibiting virus binding to host cells. POPG and PI inhibit these viral infections in vivo, in multiple animal models. Especially noteworthy, these lipids markedly attenuate SARS-CoV-2 infection including its variants. These lipids are natural compounds that already exist in the lung and, thus, are less likely to cause adverse immune responses by hosts. Collectively, these data demonstrate that POPG and PI have strong potential as novel therapeutics for applications as anti-inflammatory compounds and preventatives, as treatments for broad ranges of RNA respiratory viruses.


Subject(s)
COVID-19 , Pulmonary Surfactants , Animals , Humans , Phospholipids/metabolism , Pulmonary Surfactants/therapeutic use , Pulmonary Surfactants/chemistry , Pulmonary Surfactants/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Toll-Like Receptor 2 , SARS-CoV-2 , Lung/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Phosphatidylglycerols/therapeutic use , Phosphatidylglycerols/pharmacology
4.
Int J Mol Sci ; 24(6)2023 Mar 19.
Article in English | MEDLINE | ID: mdl-36982926

ABSTRACT

Our previous work shows that dioleoylphosphatidylglycerol (DOPG) accelerates corneal epithelial healing in vitro and in vivo by unknown mechanisms. Prior data demonstrate that DOPG inhibits toll-like receptor (TLR) activation and inflammation induced by microbial components (pathogen-associated molecular patterns, PAMPs) and by endogenous molecules upregulated in psoriatic skin, which act as danger-associated molecular patterns (DAMPs) to activate TLRs and promote inflammation. In the injured cornea, sterile inflammation can result from the release of the DAMP molecule, heat shock protein B4 (HSPB4), to contribute to delayed wound healing. Here, we show in vitro that DOPG inhibits TLR2 activation induced in response to HSPB4, as well as DAMPs that are elevated in diabetes, a disease that also slows corneal wound healing. Further, we show that the co-receptor, cluster of differentiation-14 (CD14), is necessary for PAMP/DAMP-induced activation of TLR2, as well as of TLR4. Finally, we simulated the high-glucose environment of diabetes to show that elevated glucose levels enhance TLR4 activation by a DAMP known to be upregulated in diabetes. Together, our results demonstrate the anti-inflammatory actions of DOPG and support further investigation into its development as a possible therapy for corneal injury, especially in diabetic patients at high risk of vision-threatening complications.


Subject(s)
HMGB1 Protein , Toll-Like Receptor 2 , Humans , Adaptor Proteins, Signal Transducing/metabolism , Alarmins , Antigens, CD19 , Glucose , Heat-Shock Proteins/metabolism , HMGB1 Protein/metabolism , Inflammation/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Phosphatidylglycerols/pharmacology
5.
J Biol Chem ; 299(5): 104659, 2023 05.
Article in English | MEDLINE | ID: mdl-36997087

ABSTRACT

Decarboxylation of phosphatidylserine (PS) to form phosphatidylethanolamine by PS decarboxylases (PSDs) is an essential process in most eukaryotes. Processing of a malarial PSD proenzyme into its active alpha and beta subunits is by an autoendoproteolytic mechanism regulated by anionic phospholipids, with PS serving as an activator and phosphatidylglycerol (PG), phosphatidylinositol, and phosphatidic acid acting as inhibitors. The biophysical mechanism underlying this regulation remains unknown. We used solid phase lipid binding, liposome-binding assays, and surface plasmon resonance to examine the binding specificity of a processing-deficient Plasmodium PSD (PkPSDS308A) mutant enzyme and demonstrated that the PSD proenzyme binds strongly to PS and PG but not to phosphatidylethanolamine and phosphatidylcholine. The equilibrium dissociation constants (Kd) of PkPSD with PS and PG were 80.4 nM and 66.4 nM, respectively. The interaction of PSD with PS is inhibited by calcium, suggesting that the binding mechanism involves ionic interactions. In vitro processing of WT PkPSD proenzyme was also inhibited by calcium, consistent with the conclusion that PS binding to PkPSD through ionic interactions is required for the proenzyme processing. Peptide mapping identified polybasic amino acid motifs in the proenzyme responsible for binding to PS. Altogether, the data demonstrate that malarial PSD maturation is regulated through a strong physical association between PkPSD proenzyme and anionic lipids. Inhibition of the specific interaction between the proenzyme and the lipids can provide a novel mechanism to disrupt PSD enzyme activity, which has been suggested as a target for antimicrobials, and anticancer therapies.


Subject(s)
Carboxy-Lyases , Malaria , Phospholipids , Plasmodium , Amino Acid Motifs , Calcium/metabolism , Calcium/pharmacology , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , Enzyme Precursors/metabolism , Liposomes , Phosphatidic Acids/metabolism , Phosphatidic Acids/pharmacology , Phosphatidylcholines/metabolism , Phosphatidylcholines/pharmacology , Phosphatidylethanolamines/metabolism , Phosphatidylethanolamines/pharmacology , Phosphatidylglycerols/metabolism , Phosphatidylglycerols/pharmacology , Phosphatidylinositols/metabolism , Phosphatidylinositols/pharmacology , Phosphatidylserines/metabolism , Phosphatidylserines/pharmacology , Phospholipids/chemistry , Phospholipids/metabolism , Phospholipids/pharmacology , Protein Binding , Malaria/parasitology , Proteolysis/drug effects , Surface Plasmon Resonance , Plasmodium/enzymology
6.
Eur J Clin Microbiol Infect Dis ; 41(2): 331-333, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34671843

ABSTRACT

The emergence of high-level daptomycin (DAP)-resistant (HLDR) Corynebacterium striatum has been reported as a result of loss-of-function point mutations or premature stop codon mutations in a responsible gene, pgsA2. We herein describe the novel detection of an HLDR C. striatum clinical isolate, in which IS30-insertion was corroborated to cause destruction of pgsA2 gene. We isolated an HLDR C. striatum from a critically ill patient with underlying mycosis fungoides who had been treated with DAP for 10 days. With a sequence investigation, IS30-insertion was discovered to split pgsA2 in the HLDR C. striatum strain, which may cause disrupted phospholipid phosphatidylglycerol (PG) production. Future studies should survey the prevalence of IS-mediated gene inactivation among HLDR C. striatum clinical isolates.


Subject(s)
Corynebacterium/enzymology , Corynebacterium/genetics , Drug Resistance, Bacterial/genetics , Mutation , Transferases (Other Substituted Phosphate Groups)/genetics , Anti-Bacterial Agents/pharmacology , Corynebacterium/drug effects , Corynebacterium Infections , Daptomycin/pharmacology , Drug Resistance, Bacterial/drug effects , Female , Genes, Bacterial/genetics , Humans , Middle Aged , Phosphatidylglycerols/pharmacology , Phospholipids/pharmacology
7.
J Biol Chem ; 297(1): 100813, 2021 07.
Article in English | MEDLINE | ID: mdl-34023384

ABSTRACT

Niemann-Pick C (NPC) is an autosomal recessive disorder characterized by mutations in the NPC1 or NPC2 genes encoding endolysosomal lipid transport proteins, leading to cholesterol accumulation and autophagy dysfunction. We have previously shown that enrichment of NPC1-deficient cells with the anionic lipid lysobisphosphatidic acid (LBPA; also called bis(monoacylglycerol)phosphate) via treatment with its precursor phosphatidylglycerol (PG) results in a dramatic decrease in cholesterol storage. However, the mechanisms underlying this reduction are unknown. In the present study, we showed using biochemical and imaging approaches in both NPC1-deficient cellular models and an NPC1 mouse model that PG incubation/LBPA enrichment significantly improved the compromised autophagic flux associated with NPC1 disease, providing a route for NPC1-independent endolysosomal cholesterol mobilization. PG/LBPA enrichment specifically enhanced the late stages of autophagy, and effects were mediated by activation of the lysosomal enzyme acid sphingomyelinase. PG incubation also led to robust and specific increases in LBPA species with polyunsaturated acyl chains, potentially increasing the propensity for membrane fusion events, which are critical for late-stage autophagy progression. Finally, we demonstrated that PG/LBPA treatment efficiently cleared cholesterol and toxic protein aggregates in Purkinje neurons of the NPC1I1061T mouse model. Collectively, these findings provide a mechanistic basis supporting cellular LBPA as a potential new target for therapeutic intervention in NPC disease.


Subject(s)
Autophagy , Cholesterol/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Lysophospholipids/metabolism , Lysosomes/metabolism , Monoglycerides/metabolism , Animals , Autophagy/drug effects , Endosomes/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells , Homeostasis/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mutation/genetics , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Phosphatidylglycerols/pharmacology , Purkinje Cells/drug effects , Purkinje Cells/metabolism , Sequestosome-1 Protein/metabolism , Sphingomyelin Phosphodiesterase/metabolism
8.
FASEB J ; 35(2): e21318, 2021 02.
Article in English | MEDLINE | ID: mdl-33508158

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects cognition and memory. Recent advances have helped identify many clinical sub-types in AD. Mounting evidence point toward structural polymorphism among fibrillar aggregates of amyloid-ß (Aß) to being responsible for the phenotypes and clinical manifestations. In the emerging paradigm of polymorphism and prion-like propagation of aggregates in AD, the role of low molecular weight soluble oligomers, which are long known to be the primary toxic agents, in effecting phenotypes remains inconspicuous. In this study, we present the characterization of three soluble oligomers of Aß42, namely 14LPOs, 16LPOs, and GM1Os with discreet biophysical and biochemical properties generated using lysophosphatidyl glycerols and GM1 gangliosides. The results indicate that the oligomers share some biophysical similarities but display distinctive differences with GM1Os. Unlike the other two, GM1Os were observed to be complexed with the lipid upon isolation. It also differs mainly in detection by conformation-sensitive dyes and conformation-specific antibodies, temperature and enzymatic stability, and in the ability to propagate morphologically-distinct fibrils. GM1Os also show distinguishable biochemical behavior with pronounced neuronal toxicity. Furthermore, all the oligomers induce cerebral amyloid angiopathy (CAA) and plaque burden in transgenic AD mice, which seems to be a consistent feature among all lipid-derived oligomers, but 16LPOs and GM1Os displayed significantly higher effect than the others. These results establish a correlation between molecular features of Aß42 oligomers and their distinguishable effects in transgenic AD mice attuned by lipid characteristics, and therefore help bridge the knowledge gap in understanding how oligomer conformers could elicit AD phenotypes.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Lipids/pharmacology , Amyloid/drug effects , Animals , Cell Line, Tumor , Cell Survival/physiology , Circular Dichroism , Dynamic Light Scattering , G(M1) Ganglioside/pharmacology , Immunohistochemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Transgenic , Microscopy, Atomic Force , Phosphatidylglycerols/pharmacology , Plaque, Amyloid/metabolism , Spectrometry, Mass, Electrospray Ionization , Spectroscopy, Fourier Transform Infrared
9.
Mol Pharmacol ; 97(5): 324-335, 2020 05.
Article in English | MEDLINE | ID: mdl-32173651

ABSTRACT

Skin serves not only as a protective barrier to microbial entry into the body but also as an immune organ. The outer layer, the epidermis, is composed predominantly of keratinocytes, which can be stimulated to produce proinflammatory mediators. Although some inflammation is useful to defend against infection, excessive or persistent inflammation can lead to the development of inflammatory skin diseases, such as psoriasis, a common skin disorder affecting approximately 2% of the US population. We have previously found that phosphatidylglycerol (PG) derived from soy can inhibit inflammation in a contact irritant ear edema mouse model. Here, we investigated the ability of soy PG to inhibit inflammatory mediator expression in response to activators of the pattern recognition receptors, toll-like receptor-2 (TLR2) and -4 (TLR4). We found that in epidermal keratinocytes, soy PG inhibited TLR2 and TLR4 activation and inflammatory mediator expression in response to a synthetic triacylated lipopeptide and lipopolysaccharide, respectively, as well as an endogenous danger-associated molecular pattern. However, at higher concentrations, soy PG alone enhanced the expression of some proinflammatory cytokines, suggesting a narrow therapeutic window for this lipid. Dioleoylphosphatidylglycerol (DOPG), but not dioleoylphosphatidylcholine, exerted a similar inhibitory effect, completely blocking keratinocyte inflammatory mediator expression induced by TLR2 and TLR4 activators as well as NFκB activation in a macrophage cell line (RAW264.7); however, DOPG was not itself proinflammatory even at high concentrations. Furthermore, DOPG had no effect on NFκB activation in response to a TLR7/8 agonist. Our results suggest that DOPG could be used to inhibit excessive skin inflammation. SIGNIFICANCE STATEMENT: Although inflammation is beneficial for clearing an infection, in some cases, the infection can be excessive and/or become chronic, thereby resulting in considerable tissue damage and pathological conditions. We show here that the phospholipid phosphatidylglycerol can inhibit the activation of toll-like receptors 2 and 4 of the innate immune system as well as the downstream inflammatory mediator expression in response to microbial component-mimicking agents in epidermal keratinocytes that form the physical barrier of the skin.


Subject(s)
Inflammation Mediators/metabolism , Keratinocytes/metabolism , Pathogen-Associated Molecular Pattern Molecules/pharmacology , Phosphatidylglycerols/pharmacology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Calgranulin B/pharmacology , Humans , Imidazoles/pharmacology , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , NF-kappa B/metabolism , RAW 264.7 Cells , Receptors, Pattern Recognition/metabolism , Recombinant Proteins/pharmacology , Glycine max/chemistry
10.
Invest Ophthalmol Vis Sci ; 61(3): 29, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32186673

ABSTRACT

Purpose: In contact with the external environment, the cornea can easily be injured. Although corneal wounds generally heal rapidly, the pain and increased risk of infection associated with a damaged cornea, as well as the impaired healing observed in some individuals, emphasize the need for novel treatments to accelerate corneal healing. We previously demonstrated in epidermal keratinocytes that the glycerol channel aquaporin-3 (AQP3) interacts with phospholipase D2 (PLD2) to produce the signaling phospholipid phosphatidylglycerol (PG), which has been shown to accelerate skin wound healing in vivo. We hypothesized that the same signaling pathway might be operational in corneal epithelial cells. Methods: We used co-immunoprecipitation, immunohistochemistry, scratch wound healing assays in vitro, and corneal epithelial wound healing assays in vivo to determine the role of the AQP3/PLD2/PG signaling pathway in corneal epithelium. Results: AQP3 was present in human corneas in situ, and AQP3 and PLD2 were co-immunoprecipitated from corneal epithelial cell lysates. The two proteins could also be co-immunoprecipitated from insect cells simultaneously infected with AQP3- and PLD2-expressing baculoviruses, suggesting a likely direct interaction. A particular PG, dioleoylphosphatidylglycerol (DOPG), enhanced scratch wound healing of a corneal epithelial monolayer in vitro. DOPG also accelerated corneal epithelial wound healing in vivo, both in wild-type mice and in a mouse model exhibiting impaired corneal wound healing (AQP3 knockout mice). Conclusions: These results indicate the importance of the AQP3/PLD2/PG signaling pathway in corneal epithelial cells and suggest the possibility of developing DOPG as a pharmacologic therapy to enhance corneal wound healing in patients.


Subject(s)
Epithelium, Corneal/drug effects , Limbus Corneae/drug effects , Phosphatidylglycerols/pharmacology , Wound Healing/physiology , Animals , Aquaporin 3/metabolism , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Epithelium, Corneal/metabolism , Humans , Immunoprecipitation , Limbus Corneae/metabolism , Male , Mice , Mice, Knockout , Microscopy, Fluorescence , Phospholipase D/metabolism , Sf9 Cells/metabolism , Signal Transduction/physiology , Transfection
11.
J Biol Chem ; 295(6): 1704-1715, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31882535

ABSTRACT

The influenza A (H1N1)pdm09 outbreak in 2009 exemplified the problems accompanying the emergence of novel influenza A virus (IAV) strains and their unanticipated virulence in populations with no pre-existing immunity. Neuraminidase inhibitors (NAIs) are currently the drugs of choice for intervention against IAV outbreaks, but there are concerns that NAI-resistant viruses can transmit to high-risk populations. These issues highlight the need for new approaches that address the annual influenza burden. In this study, we examined whether palmitoyl-oleoyl-phosphatidylglycerol (POPG) and phosphatidylinositol (PI) effectively antagonize (H1N1)pdm09 infection. POPG and PI markedly suppressed cytopathic effects and attenuated viral gene expression in (H1N1)pdm09-infected Madin-Darby canine kidney cells. POPG and PI bound to (H1N1)pdm09 with high affinity and disrupted viral spread from infected to noninfected cells in tissue culture and also reduced (H1N1)pdm09 propagation by a factor of 102 after viral infection was established in vitro In a mouse infection model of (H1N1)pdm09, POPG and PI significantly reduced lung inflammation and viral burden. Of note, when mice were challenged with a typically lethal dose of 1000 plaque-forming units of (H1N1)pdm09, survival after 10 days was 100% (14 of 14 mice) with the POPG treatment compared with 0% (0 of 14 mice) without this treatment. POPG also significantly reduced inflammatory infiltrates and the viral burden induced by (H1N1)pdm09 infection in a ferret model. These findings indicate that anionic phospholipids potently and efficiently disrupt influenza infections in animal models.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Phosphatidylglycerols/therapeutic use , Phosphatidylinositols/therapeutic use , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Dogs , Female , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Phosphatidylglycerols/pharmacology , Phosphatidylinositols/pharmacology , Pulmonary Surfactants/pharmacology , Pulmonary Surfactants/therapeutic use
12.
Nanomedicine ; 23: 102096, 2020 01.
Article in English | MEDLINE | ID: mdl-31669855

ABSTRACT

Phosphatidylserine (PS) and phosphatidylglycerol (PG) are endogenous phospholipids with putative anti-inflammatory potential. However, studies comparing PS and PG are rare and were mainly conducted with phospholipid-dispersions of large size and broad distributions. Thus, we prepared small-sized PS- and PG-loaded liposomes exhibiting narrow distribution, and additionally studied the impact of liposome-pegylation on the reduction of the TNFα-production caused by the PS- and PG-liposomes. These PS- and PG-containing nanodispersions had a small size around 100nm and a narrow distribution (PDI<0.1). The liposome-dispersions showed no toxicity in NHDF- and 3T3-cells and virtually no hemolytic activity. They decreased the TNFα-production of LPS-(lipopolysaccharide)-stimulated mouse peritoneal macrophages in vitro. PG-liposomes always decreased the TNFα-levels more potently than PS-liposomes. Pegylation of PS- and PG-liposomes caused different Zeta potentials, but did not change biological activity. The results of the current study indicate a high potential of the tested formulations for phospholipid-based anti-inflammatory therapies.


Subject(s)
Macrophages, Peritoneal/metabolism , Nanoparticles , Phosphatidylglycerols , Phosphatidylserines , 3T3 Cells , Animals , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Liposomes , Macrophages, Peritoneal/pathology , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/pharmacology , Phosphatidylserines/chemistry , Phosphatidylserines/pharmacology
13.
Mar Drugs ; 17(2)2019 02 09.
Article in English | MEDLINE | ID: mdl-30744121

ABSTRACT

An unprecedented phosphatidylmonogalactosyldiacylglycerol pool (PGDG, 1) rich in polyunsaturated fatty acids was isolated from the marine diatoms Thalassiosira weissflogii. Here we report for the first time the NMR characterization of this rare lipid from marine organisms along with a synthetic strategy for the preparation of a PGDG analog (2). PGDG 1 exhibited immunostimulatory activity in human dendritic cells (DCs) and the synthetic PGDG 2 was prepared to explore its mechanism of action. A Toll-like receptor-4 (TLR-4) agonistic activity was evidenced in human and murine DCs underlying the antigen-specific T-cell activation of this class of molecules.


Subject(s)
Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Diatoms/chemistry , Glycolipids/chemistry , Glycolipids/pharmacology , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/pharmacology , Toll-Like Receptor 4/agonists , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/isolation & purification , Animals , Carbon-13 Magnetic Resonance Spectroscopy , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Glycolipids/chemical synthesis , Glycolipids/isolation & purification , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylglycerols/chemical synthesis , Phosphatidylglycerols/isolation & purification , Toll-Like Receptor 4/immunology
14.
J Invest Dermatol ; 139(4): 868-877, 2019 04.
Article in English | MEDLINE | ID: mdl-30391260

ABSTRACT

Psoriasis is a common skin disorder characterized by hyperproliferation and aberrant differentiation of epidermal keratinocytes and inflammation. We previously showed that phosphatidylglycerol (PG) can regulate keratinocyte function and suppress skin inflammation. Based on data suggesting that PG can inhibit toll-like receptor (TLR) activation induced by microorganisms and their components, we determined whether PG can inhibit TLR activation in response to antimicrobial peptides. These peptides, which are up-regulated in psoriasis, are known to function as danger-associated molecular patterns (i.e., DAMPs) to activate TLRs and the innate immune system. Because S100A9 is elevated in psoriatic skin and in animal models of psoriasis, we selected S100A9 as a representative antimicrobial peptide DAMP. We showed that in primary keratinocytes and a macrophage cell line, PG suppressed inflammatory mediator production induced by recombinant S100A9 functioning through both TLR2 and TLR4. In addition, PG, but not phosphatidylcholine, inhibited downstream S100A9-elicited TLR2 and NF-κB activation. These results, to our knowledge previously unreported, show PG's ability to inhibit DAMP-induced TLR activation, thereby reducing inflammatory signals. In addition, topical PG ameliorated skin lesions and inflammation in a mouse model of psoriasis. Together, these results suggest the possibility of developing PG as a therapy for psoriasis.


Subject(s)
Alarmins/metabolism , Gene Expression Regulation , Phosphatidylglycerols/pharmacology , Psoriasis/genetics , RNA/genetics , Toll-Like Receptors/genetics , Animals , Animals, Newborn , Blotting, Western , Calgranulin B/biosynthesis , Calgranulin B/drug effects , Calgranulin B/genetics , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Psoriasis/metabolism , Psoriasis/pathology , Signal Transduction , Toll-Like Receptors/biosynthesis
15.
Eur J Pharm Sci ; 128: 43-53, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30471412

ABSTRACT

The mechanisms of membrane defence by lysylphosphatidylglycerol (LPG), were investigated using synthetic biomimetic mono- and bilayer models of methicillin resistant S. aureus ST239 TW, based on its lipid composition in both pH 7.4 (28% LPG) and pH 5.5 (51% LPG) cultures. These models incorporated a stable synthetic analogue of LPG (3adLPG) to facilitate long-duration biophysical studies, which were previously limited by the lability native LPG. Both increased 3adLPG content and full headgroup ionization at pH 5.5, increased bilayer order and dampened overall charge, via the formation of neutral ion pairs with anionic lipids. Ion pair formation in air/liquid interface lipid monolayers elicited a significant condensing effect, which correlated with the inhibition of subphase-injected magainin 2 F5W partitioning. In fluid phase lipid vesicles, increasing the proportion of 3adLPG from 28 to 51 mol% completely inhibited the adoption of the membrane-active α­helical conformation of the peptide, without the need for full headgroup ionization. Neutron reflectivity measurements performed on biomimetic PG/3adLPG fluid floating bilayers, showed a significant ordering effect of mild acidity on a bilayer containing 30 mol% 3adLPG, whilst peptide binding/partitioning was only fully inhibited in a bilayer with 55 mol% 3adLPG at pH 5.5. These findings are discussed with respect to the roles of LPG in resistance to human epithelial defences in S. aureus and the continued evolution of this opportunistic pathogen's virulence.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Cell Membrane/physiology , Staphylococcus aureus , Adaptation, Biological , Anti-Bacterial Agents , Antimicrobial Cationic Peptides/metabolism , Biological Transport , Drug Resistance, Bacterial , Hydrogen-Ion Concentration , Lipid Metabolism , Molecular Structure , Phosphatidylglycerols/chemical synthesis , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/pharmacology
16.
Sci Rep ; 8(1): 17704, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30531805

ABSTRACT

Peptidoglycan (PG) biosynthesis and assembly are needed for bacterial cell wall formation. Lipid II is the precursor in the PG biosynthetic pathway and carries a nascent PG unit that is processed by glycosyltransferases. Despite its immense therapeutic value as a target of several classes of antibiotics, the conformational ensemble of lipid II in bacterial membranes and its interactions with membrane-anchored enzymes remain elusive. In this work, lipid II and its elongated forms (lipid VI and lipid XII) were modeled and simulated in bilayers of POPE (palmitoyl-oleoyl-phosphatidyl-ethanolamine) and POPG (palmitoyl-oleoyl-phosphatidyl-glycerol) that mimic the prototypical composition of Gram-negative cytoplasmic membranes. In addition, penicillin-binding protein 1b (PBP1b) from Escherichia coli was modeled and simulated in the presence of a nascent PG to investigate their interactions. Trajectory analysis reveals that as the glycan chain grows, the non-reducing end of the nascent PG displays much greater fluctuation along the membrane normal and minimally interacts with the membrane surface. In addition, dihedral angles within the pyrophosphate moiety are determined by the length of the PG moiety and its surrounding environment. When a nascent PG is bound to PBP1b, the stem peptide remains in close contact with PBP1b by structural rearrangement of the glycan chain. Most importantly, the number of nascent PG units required to reach the transpeptidase domain are determined to be 7 or 8. Our findings complement experimental results to further understand how the structure of nascent PG can dictate the assembly of the PG scaffold.


Subject(s)
Cell Membrane/metabolism , Peptidoglycan/biosynthesis , Peptidoglycan/metabolism , Cell Membrane/drug effects , Cell Wall/drug effects , Cell Wall/metabolism , Diphosphates/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , Penicillin-Binding Proteins/metabolism , Peptidyl Transferases/metabolism , Phosphatidylethanolamines/pharmacology , Phosphatidylglycerols/pharmacology , Polysaccharides/metabolism , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives , Uridine Diphosphate N-Acetylmuramic Acid/metabolism
17.
Vet Microbiol ; 224: 1-7, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30269782

ABSTRACT

Prion diseases are characterized by the conformational conversion of the cellular prion protein (PrPC) to the pathogenic isoform (PrPSc). Lipids have been found to interact with PrPC and contribute to the efficient formation of PrPSc. Non-mammalian PrPs are not readily to undergo the conversion process into an infectious isoform, yet the effect of lipid on the conformational conversion of non-mammalian PrPC remains to be explored. Herein, the effects of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol (POPG) on full-length recombinant chicken PrP (ChPrP) 24-249 and murine PrP (MoPrP) 23-230 were investigated. Firstly, it was found that in the presence of chemical denaturant, POPG remarkably inhibited MoPrP amyloid fibril growth, while had slight effect on that of ChPrP as estimated by amyloid fibril growth and transmissible electronic microscope assays. Secondly, under physiological condition, POPG induced conformation changes in both MoPrP and ChPrP using Thioflavin T (ThT) fluorescence, circular dichroism, proteinase K digestion and transmission electron microscopy assays. With a POPG:PrP molar ratio of 30:1, the ThT fluorescence of MoPrP was found to be lower than that of ChPrP, however, the POPG-induced MoPrP had higher ß-sheet content and was more proteinase K-resistant than POPG-induced ChPrP. In summary, the present results suggested that the effects of POPG on conformational conversion of MoPrP and ChPrP were different under both denaturation and physiological conditions.


Subject(s)
Phosphatidylglycerols/pharmacology , Prion Proteins/chemistry , Prion Proteins/drug effects , Amyloid/drug effects , Amyloid/physiology , Animals , Chickens , Mice , Microscopy, Electron, Transmission , Prion Proteins/genetics , Protein Conformation , Recombinant Proteins/drug effects
18.
J Pharmacol Exp Ther ; 366(1): 1-8, 2018 07.
Article in English | MEDLINE | ID: mdl-29695409

ABSTRACT

We have previously shown that phosphatidylglycerol (PG) regulates the function of keratinocytes, the predominant cells that compose the epidermis, inhibiting the proliferation of rapidly dividing keratinocytes. In particular, soy PG, a PG mixture with a high proportion of polyunsaturated fatty acids, is efficacious at inhibiting these proliferating keratinocytes. Psoriasis is a skin disorder characterized by hyperproliferation of keratinocytes and inflammation. Data in the lung suggest that PG in pulmonary surfactant inhibits inflammation. To investigate the possibility of using PG containing polyunsaturated fatty acids for the treatment of psoriasis, we examined the effect of soy PG on inflammation induced by the application of 12-O-tetradecanoylphorbol 13-acetate (TPA), a contact irritant, to mouse ears in vivo. We monitored ear thickness and weight as a measure of ear edema, as well as CD45-positive immune cell infiltration. Our results indicate that soy PG when applied together with 1,25-dihydroxyvitamin D3 (vitamin D), an agent known to acutely disrupt the skin barrier, suppressed ear edema and inhibited the infiltration of CD45-positive immune cells. On the other hand, neither PG nor vitamin D alone was effective. The combination also decreased tumor necrosis factor-α (TNFα) levels. This result suggested the possibility that PG was not permeating the skin barrier efficiently. Therefore, in a further study we applied PG in a penetration-enhancing vehicle and found that it inhibited inflammation induced by the phorbol ester and decreased CD45-positive immune cell infiltration. Our results suggest the possibility of using soy PG as a topical treatment option for psoriasis.


Subject(s)
Edema/chemically induced , Edema/drug therapy , Glycine max/chemistry , Irritants/adverse effects , Phosphatidylglycerols/pharmacology , Animals , Disease Models, Animal , Edema/immunology , Edema/pathology , Inflammation/drug therapy , Keratinocytes/drug effects , Male , Mice , Phosphatidylglycerols/therapeutic use , Tetradecanoylphorbol Acetate/adverse effects
19.
Cell Death Dis ; 9(2): 245, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445138

ABSTRACT

Engineered nanoparticles are finding a wide spectrum of biomedical applications, including drug delivery and capacity to trigger cytotoxic phenomena, potentially useful against tumor cells. The full understanding of their biosafety and interactions with cell processes is mandatory. Using microglial (BV-2) and alveolar basal epithelial (A549) cells, in this study we determined the effects of engineered carbon nanodiamonds (ECNs) on cell viability, nitric oxide (NO) and reactive oxygen species (ROS) production, as well as on energy metabolism. Particularly, we initially measured decrease in cell viability as a function of increasing ECNs doses, finding similar cytotoxic ECN effects in the two cell lines. Subsequently, using apparently non-cytotoxic ECN concentrations (2 µg/mL causing decrease in cell number < 5%) we determined NO and ROS production, and measured the concentrations of compounds related to energy metabolism, mitochondrial functions, oxido-reductive reactions, and antioxidant defences. We found that in both cell lines non-cytotoxic ECN concentrations increased NO and ROS production with sustained oxidative/nitrosative stress, and caused energy metabolism imbalance (decrease in high energy phosphates and nicotinic coenzymes) and mitochondrial malfunctioning (decrease in ATP/ADP ratio).These results underline the importance to deeply investigate the molecular and biochemical changes occurring upon the interaction of ECNs (and nanoparticles in general) with living cells, even at apparently non-toxic concentration. Since the use of ECNs in biomedical field is attracting increasing attention the complete evaluation of their biosafety, toxicity and/or possible side effects both in vitro and in vivo is mandatory before these highly promising tools might find the correct application.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/pharmacology , Mitochondria/drug effects , Nanodiamonds/chemistry , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Phosphatidylglycerols/pharmacology , 1,2-Dipalmitoylphosphatidylcholine/chemistry , A549 Cells , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cell Line, Transformed , Energy Metabolism/drug effects , Humans , Mice , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Mitochondria/metabolism , NAD/metabolism , NADP/metabolism , Nitric Oxide/agonists , Nitric Oxide/metabolism , Phosphatidylglycerols/chemistry , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism
20.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L32-L53, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28860142

ABSTRACT

The biological and immune-protective properties of surfactant-derived phospholipids and phospholipid subfractions in the context of neonatal inflammatory lung disease are widely unknown. Using a porcine neonatal triple-hit acute respiratory distress syndrome (ARDS) model (repeated airway lavage, overventilation, and LPS instillation into airways), we assessed whether the supplementation of surfactant (S; poractant alfa) with inositol derivatives [inositol 1,2,6-trisphosphate (IP3) or phosphatidylinositol 3,5-bisphosphate (PIP2)] or phosphatidylglycerol subfractions [16:0/18:1-palmitoyloleoyl-phosphatidylglycerol (POPG) or 18:1/18:1-dioleoyl-phosphatidylglycerol (DOPG)] would result in improved clinical parameters and sought to characterize changes in key inflammatory pathways behind these improvements. Within 72 h of mechanical ventilation, the oxygenation index (S+IP3, S+PIP2, and S+POPG), the ventilation efficiency index (S+IP3 and S+POPG), the compliance (S+IP3 and S+POPG) and resistance (S+POPG) of the respiratory system, and the extravascular lung water index (S+IP3 and S+POPG) significantly improved compared with S treatment alone. The inositol derivatives (mainly S+IP3) exerted their actions by suppressing acid sphingomyelinase activity and dependent ceramide production, linked with the suppression of the inflammasome nucleotide-binding domain, leucine-rich repeat-containing protein-3 (NLRP3)-apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-caspase-1 complex, and the profibrotic response represented by the cytokines transforming growth factor-ß1 and IFN-γ, matrix metalloproteinase (MMP)-1/8, and elastin. In addition, IκB kinase activity was significantly reduced. S+POPG and S+DOPG treatment inhibited polymorphonuclear leukocyte activity (MMP-8 and myeloperoxidase) and the production of interleukin-6, maintained alveolar-capillary barrier functions, and reduced alveolar epithelial cell apoptosis, all of which resulted in reduced pulmonary edema. S+DOPG also limited the profibrotic response. We conclude that highly concentrated inositol derivatives and phosphatidylglycerol subfractions in surfactant preparations mitigate key inflammatory pathways in inflammatory lung disease and that their clinical application may be of interest for future treatment of the acute exudative phase of neonatal ARDS.


Subject(s)
Disease Models, Animal , Inositol/pharmacology , Phosphatidylglycerols/pharmacology , Pulmonary Edema/drug therapy , Pulmonary Surfactants/pharmacology , Respiratory Distress Syndrome, Newborn/drug therapy , Animals , Animals, Newborn , Apoptosis , Bronchoalveolar Lavage Fluid , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Male , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Gas Exchange , Random Allocation , Respiration, Artificial , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Swine , Translational Research, Biomedical , Vitamin B Complex/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...