Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Immunity ; 55(3): 423-441.e9, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35139355

RESUMO

Cell death plays an important role during pathogen infections. Here, we report that interferon-γ (IFNγ) sensitizes macrophages to Toll-like receptor (TLR)-induced death that requires macrophage-intrinsic death ligands and caspase-8 enzymatic activity, which trigger the mitochondrial apoptotic effectors, BAX and BAK. The pro-apoptotic caspase-8 substrate BID was dispensable for BAX and BAK activation. Instead, caspase-8 reduced pro-survival BCL-2 transcription and increased inducible nitric oxide synthase (iNOS), thus facilitating BAX and BAK signaling. IFNγ-primed, TLR-induced macrophage killing required iNOS, which licensed apoptotic caspase-8 activity and reduced the BAX and BAK inhibitors, A1 and MCL-1. The deletion of iNOS or caspase-8 limited SARS-CoV-2-induced disease in mice, while caspase-8 caused lethality independent of iNOS in a model of hemophagocytic lymphohistiocytosis. These findings reveal that iNOS selectively licenses programmed cell death, which may explain how nitric oxide impacts disease severity in SARS-CoV-2 infection and other iNOS-associated inflammatory conditions.


Assuntos
COVID-19/imunologia , Caspase 8/metabolismo , Interferon gama/metabolismo , Linfo-Histiocitose Hemofagocítica/imunologia , Macrófagos/imunologia , Mitocôndrias/metabolismo , SARS-CoV-2/fisiologia , Animais , Caspase 8/genética , Células Cultivadas , Citotoxicidade Imunológica , Humanos , Interferon gama/genética , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/metabolismo , Moléculas com Motivos Associados a Patógenos/imunologia , Transdução de Sinais , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
2.
Small ; 20(6): e2305052, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37798622

RESUMO

The rapid increase and spread of Gram-negative bacteria resistant to many or all existing treatments threaten a return to the preantibiotic era. The presence of bacterial polysaccharides that impede the penetration of many antimicrobials and protect them from the innate immune system contributes to resistance and pathogenicity. No currently approved antibiotics target the polysaccharide regions of microbes. Here, describe monolaurin-based niosomes, the first lipid nanoparticles that can eliminate bacterial polysaccharides from hypervirulent Klebsiella pneumoniae, are described. Their combination with polymyxin B shows no cytotoxicity in vitro and is highly effective in combating K. pneumoniae infection in vivo. Comprehensive mechanistic studies have revealed that antimicrobial activity proceeds via a multimodal mechanism. Initially, lipid nanoparticles disrupt polysaccharides, then outer and inner membranes are destabilized and destroyed by polymyxin B, resulting in synergistic cell lysis. This novel lipidic nanoparticle system shows tremendous promise as a highly effective antimicrobial treatment targeting multidrug-resistant Gram-negative pathogens.


Assuntos
Nanopartículas , Polimixina B , Polimixina B/farmacologia , Lipossomos/farmacologia , Antibacterianos/farmacologia , Bactérias Gram-Negativas , Klebsiella pneumoniae , Polissacarídeos Bacterianos/farmacologia , Testes de Sensibilidade Microbiana , Farmacorresistência Bacteriana Múltipla
3.
Artigo em Inglês | MEDLINE | ID: mdl-32660985

RESUMO

Innate immunity is crucial for the host to defend against infections, and understanding the effect of polymyxins on innate immunity is important for optimizing their clinical use. In this study, we investigated the potential toxicity of polymyxins on human macrophage-like THP-1 and neutrophil-like HL-60 cells. Differentiated THP-1 human macrophages (THP-1-dMs) and HL-60 human neutrophils (HL-60-dNs) were employed. Flow cytometry was used to measure the concentration-dependent effects (100 to 2,500 µM for THP-1-dMs and 5 to 2,500 µM for HL-60-dNs) and time-dependent effects (1,000 µM for THP-1-dMs and 300 µM for HL-60-dNs) of polymyxin B over 24 h. Effects of polymyxin B on mitochondrial activity, activation of caspase-3, caspase-8, and caspase-9, and Fas ligand (FasL) expression in both cell lines were examined using fluorescence imaging, colorimetric, and fluorometric assays. In both cell lines, polymyxin B induced concentration- and time-dependent loss of viability at 24 h with 50% effective concentration (EC50) values of 751.8 µM (95% confidence interval [CI], 692.1 to 816.6 µM; Hill slope, 3.09 to 5.64) for THP-1-dM cells and 175.4 µM (95% CI, 154.8 to 198.7 µM; Hill slope, 1.42 to 2.21) for HL-60-dN cells. A concentration-dependent loss of mitochondrial membrane potential and generation of mitochondrial superoxide was also observed. Polymyxin B-induced apoptosis was associated with concentration-dependent activation of all three tested caspases. The death receptor apoptotic pathway activation was demonstrated by a concentration-dependent increase of FasL expression. For the first time, our results reveal that polymyxin B induced concentration- and time-dependent cell death in human macrophage-like THP-1 and neutrophil-like HL-60 cells associated with mitochondrial and death receptor apoptotic pathways.


Assuntos
Apoptose , Macrófagos , Neutrófilos , Polimixinas , Células HL-60 , Humanos , Células THP-1
4.
PLoS Pathog ; 14(3): e1006945, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29601598

RESUMO

Neisseria gonorrhoeae causes the sexually transmitted disease gonorrhoea by evading innate immunity. Colonizing the mucosa of the reproductive tract depends on the bacterial outer membrane porin, PorB, which is essential for ion and nutrient uptake. PorB is also targeted to host mitochondria and regulates apoptosis pathways to promote infections. How PorB traffics from the outer membrane of N. gonorrhoeae to mitochondria and whether it modulates innate immune cells, such as macrophages, remains unclear. Here, we show that N. gonorrhoeae secretes PorB via outer membrane vesicles (OMVs). Purified OMVs contained primarily outer membrane proteins including oligomeric PorB. The porin was targeted to mitochondria of macrophages after exposure to purified OMVs and wild type N. gonorrhoeae. This was associated with loss of mitochondrial membrane potential, release of cytochrome c, activation of apoptotic caspases and cell death in a time-dependent manner. Consistent with this, OMV-induced macrophage death was prevented with the pan-caspase inhibitor, Q-VD-PH. This shows that N. gonorrhoeae utilizes OMVs to target PorB to mitochondria and to induce apoptosis in macrophages, thus affecting innate immunity.


Assuntos
Apoptose , Membrana Celular/metabolismo , Gonorreia/patologia , Macrófagos/patologia , Mitocôndrias/patologia , Neisseria gonorrhoeae/patogenicidade , Porinas/metabolismo , Animais , Gonorreia/microbiologia , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/microbiologia , Porinas/genética
5.
Cell Microbiol ; 18(4): 466-74, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26833712

RESUMO

Macrophages can respond to microbial infections with programmed cell death. The major cell death pathways of apoptosis, pyroptosis and necroptosis are tightly regulated to ensure adequate immune reactions to virulent and persistent invaders. Macrophage death eliminates the replicative niche of intracellular pathogens and induces immune attack. Not surprisingly, successful pathogens have evolved strategies to modulate macrophage cell death pathways to enable microbial survival and replication. Uncontrolled macrophage death can also lead to tissue damage, which may augment bacterial dissemination and pathology. In this review, we highlight how pathogens hijack macrophage cell death signals to promote microbial survival and immune evasion.


Assuntos
Morte Celular , Doenças Transmissíveis/imunologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Macrófagos/microbiologia , Macrófagos/fisiologia , Animais , Doenças Transmissíveis/patologia , Humanos
6.
Eur Biophys J ; 44(8): 697-708, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26271933

RESUMO

Annexin V is of crucial importance for detection of the phosphatidylserine of apoptotic cell membranes. However, the manner in which different amounts of phosphatidylserine at the membrane surface at different stages of apoptosis contribute to binding of annexin V is unclear. We have used a quartz crystal microbalance combined with dissipative monitoring (QCM-D) and neutron reflectivity to characterize binding of human annexin V to supported bilayers of different phospholipid composition. We created model apoptotic bilayers of 1-palmitoyl-2-oleoyl-sn-glycerophosphocholine and 1-palmitoyl-2-oleoyl-sn-glycerophosphoserine (POPS) in the ratios 19:1, 9:1, 6.7:1, 4:1, 3:1, and 2:1 (w/w) in the presence of 2.5 mM CaCl2. QCM-D data revealed that annexin V bound less to supported fluid lipid bilayers with higher POPS content (>25 % POPS). Neutron reflectivity was used to further characterize the detailed composition of lipid bilayers with membrane-bound annexin V. Analysis confirmed less annexin V binding with higher POPS content, that bound annexin V formed a discrete layer above the lipid bilayer with little effect on the overall structure of the membrane, and that the thickness and volume fraction of the annexin V layer varied with POPS content. From these results we show that the POPS content of the outer surface of lipid bilayers affects the structure of membrane-bound annexin V.


Assuntos
Anexina A5/química , Apoptose , Bicamadas Lipídicas/química , Sequência de Aminoácidos , Anexina A5/metabolismo , Sítios de Ligação , Humanos , Bicamadas Lipídicas/metabolismo , Dados de Sequência Molecular , Ligação Proteica
7.
Life Sci Alliance ; 6(6)2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36977592

RESUMO

Staphylococcus aureus causes severe infections such as pneumonia and sepsis depending on the pore-forming toxin Panton-Valentine leukocidin (PVL). PVL kills and induces inflammation in macrophages and other myeloid cells by interacting with the human cell surface receptor, complement 5a receptor 1 (C5aR1). C5aR1 expression is tighly regulated and may thus modulate PVL activity, although the mechanisms involved remain incompletely understood. Here, we used a genome-wide CRISPR/Cas9 screen and identified F-box protein 11 (FBXO11), an E3 ubiquitin ligase complex member, to promote PVL toxicity. Genetic deletion of FBXO11 reduced the expression of C5aR1 at the mRNA level, whereas ectopic expression of C5aR1 in FBXO11-/- macrophages, or priming with LPS, restored C5aR1 expression and thereby PVL toxicity. In addition to promoting PVL-mediated killing, FBXO11 dampens secretion of IL-1ß after NLRP3 activation in response to bacterial toxins by reducing mRNA levels in a BCL-6-dependent and BCL-6-independent manner. Overall, these findings highlight that FBXO11 regulates C5aR1 and IL-1ß expression and controls macrophage cell death and inflammation following PVL exposure.


Assuntos
Toxinas Bacterianas , Proteínas F-Box , Humanos , Neutrófilos/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Exotoxinas/toxicidade , Inflamação/genética , Inflamação/metabolismo , Macrófagos/metabolismo , Morte Celular/genética , Leucocidinas/farmacologia , Leucocidinas/toxicidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo
8.
Methods Mol Biol ; 2523: 239-252, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35759201

RESUMO

The NLRP3 inflammasome senses the activity of pore-forming toxins secreted by Staphylococcus aureus. The bacterial toxins compromise plasma membrane integrity which activates the NLRP3 inflammasome to induce host pore-forming proteins and cellular suicide, termed pyroptosis. Host cell death rates are routinely determined at pre-defined time points and on whole cell populations. To capture the dynamic interactions between bacterial pore-forming toxins and host cell death factors, we have applied live-cell imaging techniques capable of analyzing single cell events in real time. Here, we describe methods using live-cell imaging to determine the host responses, such as plasma membrane integrity, mitochondrial health, and apoptotic caspases, towards pore-forming toxins.


Assuntos
Toxinas Bacterianas , Inflamassomos , Toxinas Bacterianas/metabolismo , Sobrevivência Celular , Humanos , Inflamassomos/metabolismo , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , Transdução de Sinais
9.
Microlife ; 3: uqac013, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37223348

RESUMO

Neisseria gonorrhoeae causes the sexually transmitted disease gonorrhoea. The treatment of gonorrhoea is becoming increasingly challenging, as N. gonorrhoeae has developed resistance to antimicrobial agents routinely used in the clinic. Resistance to penicillin is wide-spread partly due to the acquisition of ß-lactamase genes. How N. gonorrhoeae survives an initial exposure to ß-lactams before acquiring resistance genes remains to be understood. Here, using a panel of clinical isolates of N. gonorrhoeae we show that the ß-lactamase enzyme is packaged into outer membrane vesicles (OMVs) by strains expressing blaTEM-1B or blaTEM-106, which protects otherwise susceptible clinical isolates from the ß-lactam drug amoxycillin. We characterized the phenotypes of these clinical isolates of N. gonorrhoeae and the time courses over which the cross-protection of the strains is effective. Imaging and biochemical assays suggest that OMVs promote the transfer of proteins and lipids between bacteria. Thus, N. gonorrhoeae strains secret antibiotic degrading enzymes via OMVs enabling survival of otherwise susceptible bacteria.

10.
ACS Appl Mater Interfaces ; 14(33): 37369-37379, 2022 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-35951370

RESUMO

Nitric oxide (NO)-releasing nanoparticles are effective nanomedicines with diverse therapeutic advantages compared with small molecule-based NO donors. Here, we report a new class of furoxan-based NO-releasing nanoparticles using a simple, creative yet facile coassembly approach. This is the first time we demonstrated that the coassembled NO-releasing nanoparticles with poly(ethylene glycol)101-block-poly(propylene glycol)56-block-poly(ethylene glycol)101 (Pluronic F127) had potent antimicrobial efficacies against methicillin-resistant Staphylococcus aureus (MRSA) strains. Nanoparticles obtained from the coassembly of either 4-(1-(3-methylpentan-5-ol)oxyl)(3-phenylsulfonyl) furoxan (compound 1) or 4-methoxy(3-phenylsulfonyl) furoxan (compound 2) with Pluronic F127 exhibit 4-fold improved antimicrobial activities compared to their self-assembled counterparts without Pluronic F127. 5(6)-Carboxylfluorescein (CF) leakage experiments further reveal that both coassembled NO-releasing nanoparticles show stronger interactions with lipid bilayers than those self-assembled alone. Subsequently, their strong plasma membrane-damaging capabilities are confirmed under both high-resolution optical microscopy and scanning electron microscopy characterizations. This coassembly approach could be readily applied to other small molecule-based antimicrobials, providing new solutions and important insights to further antimicrobial recipe design.


Assuntos
Anti-Infecciosos , Staphylococcus aureus Resistente à Meticilina , Nanopartículas , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Testes de Sensibilidade Microbiana , Óxido Nítrico , Poloxâmero , Polietilenoglicóis
11.
Nat Commun ; 13(1): 343, 2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-35039508

RESUMO

A depleted antimicrobial drug pipeline combined with an increasing prevalence of Gram-negative 'superbugs' has increased interest in nano therapies to treat antibiotic resistance. As cubosomes and polymyxins disrupt the outer membrane of Gram-negative bacteria via different mechanisms, we herein examine the antimicrobial activity of polymyxin-loaded cubosomes and explore an alternative strategy via the polytherapy treatment of pathogens with cubosomes in combination with polymyxin. The polytherapy treatment substantially increases antimicrobial activity compared to polymyxin B-loaded cubosomes or polymyxin and cubosomes alone. Confocal microscopy and neutron reflectometry suggest the superior polytherapy activity is achieved via a two-step process. Firstly, electrostatic interactions between polymyxin and lipid A initially destabilize the outer membrane. Subsequently, an influx of cubosomes results in further membrane disruption via a lipid exchange process. These findings demonstrate that nanoparticle-based polytherapy treatments may potentially serve as improved alternatives to the conventional use of drug-loaded lipid nanoparticles for the treatment of "superbugs".


Assuntos
Farmacorresistência Bacteriana Múltipla , Nanopartículas/química , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Quimioterapia Combinada , Células HEK293 , Humanos , Bicamadas Lipídicas/química , Testes de Sensibilidade Microbiana , Microscopia Confocal , Polimixina B/farmacologia
12.
Acta Biomater ; 126: 433-444, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33774200

RESUMO

Glaucoma, a major cause of irreversible blindness worldwide, is associated with elevated intraocular pressure (IOP) and progressive loss of retinal ganglion cells (RGCs) that undergo apoptosis. A mechanism for RGCs injury involves impairment of neurotrophic support and exogenous supply of neurotrophic factors has been shown to be beneficial. However, neurotrophic factors can have widespread effects on neuronal tissues, thus targeting neurotrophic support to injured neurons may be a better neuroprotective strategy. In this study, we have encapsulated LM22A-4, a small neurotrophic factor mimetic, into Annexin V-conjugated cubosomes (L4-ACs) for targeted delivery to injured RGCs in a model of acute IOP elevation, which is induced by acute IOP elevation. We have tested cubosomes formulations that encapsulate from 9% to 33% LM22A-4. Our data indicated that cubosomes encapsulating 9% and 17% LM22A-4 exhibited a mixture of Pn3m/Im3m cubic phase, whereas 23% and 33% showed a pure Im3m cubic phase. We found that 17% L4-ACs with Pn3m/Im3m symmetries showed better in-situ and in-vitro lipid membrane interactions than the 23% and 33% L4-ACs with Im3m symmetry. In vivo experiments showed that 17% L4-ACs targeted the posterior retina and the optic nerve head, which prevented RGCs loss and improved functional outcomes in a mouse model of acute IOP elevation. These results provide evidence that Annexin V-conjugated cubosomes-based LM22A-4 delivery may be a useful targeted approach to prevent the progression of RGCs loss in glaucoma. STATEMENT OF SIGNIFICANCE: Recent studies suggest that the therapy of effectively delivering neurotrophic factors to the injured retinal ganglion cells (RGCs) could promote the survival of RGCs in glaucoma. Our present work has for the first time used cubosomes as an active targeted delivery system and have successfully delivered a neuroprotective drug to the damaged RGCs in vivo. Our new cubosomal formulation can protect apoptotic cell death in vitro and in vivo, showing that cubosomes are a promising drug carrier system for ocular drug delivery and glaucoma treatment. We have further found that by controlling cubosomes in Pn3m phase we can facilitate delivery of neuroprotective drug through apoptotic membranes. This data, we believe, has important implications for future design and formulation of cubosomes for therapeutic applications.


Assuntos
Glaucoma , Disco Óptico , Animais , Benzamidas , Modelos Animais de Doenças , Glaucoma/tratamento farmacológico , Pressão Intraocular , Camundongos , Células Ganglionares da Retina
13.
Nat Microbiol ; 5(11): 1418-1427, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32807891

RESUMO

Sensing of microbes activates the innate immune system, depending on functional mitochondria. However, pathogenic bacteria inhibit mitochondrial activity by delivering toxins via outer membrane vesicles (OMVs). How macrophages respond to pathogenic microbes that target mitochondria remains unclear. Here, we show that macrophages exposed to OMVs from Neisseria gonorrhoeae, uropathogenic Escherichia coli and Pseudomonas aeruginosa induce mitochondrial apoptosis and NLRP3 inflammasome activation. OMVs and toxins that cause mitochondrial dysfunction trigger inhibition of host protein synthesis, which depletes the unstable BCL-2 family member MCL-1 and induces BAK-dependent mitochondrial apoptosis. In parallel with caspase-11-mediated pyroptosis, mitochondrial apoptosis and potassium ion efflux activate the NLRP3 inflammasome after OMV exposure in vitro. Importantly, in the in vivo setting, the activation and release of interleukin-1ß in response to N. gonorrhoeae OMVs is regulated by mitochondrial apoptosis. Our data highlight how innate immune cells sense infections by monitoring mitochondrial health.


Assuntos
Apoptose , Membrana Externa Bacteriana/metabolismo , Bactérias Gram-Negativas/metabolismo , Mitocôndrias/patologia , Animais , Vesículas Extracelulares , Bactérias Gram-Negativas/patogenicidade , Infecções por Bactérias Gram-Negativas/imunologia , Inflamação , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Biossíntese de Proteínas , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
14.
J Leukoc Biol ; 108(3): 967-981, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32531864

RESUMO

Staphylococcus aureus causes necrotizing pneumonia by secreting toxins such as leukocidins that target front-line immune cells. The mechanism by which leukocidins kill innate immune cells and trigger inflammation during S. aureus lung infection, however, remains unresolved. Here, we explored human-induced pluripotent stem cell-derived macrophages (hiPSC-dMs) to study the interaction of the leukocidins Panton-Valentine leukocidin (PVL) and LukAB with lung macrophages, which are the initial leukocidin targets during S. aureus lung invasion. hiPSC-dMs were susceptible to the leukocidins PVL and LukAB and both leukocidins triggered NLPR3 inflammasome activation resulting in IL-1ß secretion. hiPSC-dM cell death after LukAB exposure, however, was only temporarily dependent of NLRP3, although NLRP3 triggered marked cell death after PVL treatment. CRISPR/Cas9-mediated deletion of the PVL receptor, C5aR1, protected hiPSC-dMs from PVL cytotoxicity, despite the expression of other leukocidin receptors, such as CD45. PVL-deficient S. aureus had reduced ability to induce lung IL-1ß levels in human C5aR1 knock-in mice. Unexpectedly, inhibiting NLRP3 activity resulted in increased wild-type S. aureus lung burdens. Our findings suggest that NLRP3 induces macrophage death and IL-1ß secretion after PVL exposure and controls S. aureus lung burdens.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Toxinas Bacterianas/antagonistas & inibidores , Exotoxinas/antagonistas & inibidores , Células-Tronco Pluripotentes Induzidas/citologia , Leucocidinas/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Receptor da Anafilatoxina C5a/efeitos dos fármacos , Staphylococcus aureus , Animais , Antígeno CD11b/imunologia , Sistemas CRISPR-Cas , Diferenciação Celular , Células Cultivadas , Exotoxinas/deficiência , Técnicas de Introdução de Genes , Humanos , Interleucina-1beta/metabolismo , Antígenos Comuns de Leucócito/fisiologia , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Fragmentos de Peptídeos/imunologia , Pneumonia Estafilocócica/imunologia , Subunidades Proteicas , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/fisiologia , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/fisiologia
15.
J Mater Chem B ; 6(46): 7652-7661, 2018 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-32254887

RESUMO

New drug delivery materials targeting damaged ocular tissues are of particular interest. In this work, we have formulated annexin/phosphatidylserine/phytantriol and annexin/phosphatidylserine/monoolein cubosomes based on incorporation of 1,2-dipalmitoyl-sn-glycero-3-phospho-l-serine (PS) lipid and annexin V (ANX) protein with phytantriol (Phy) and monoolein (MO) respectively. The incorporation of ANX is important because it can be used as a diagnostic tool for in vivo apoptosis detection due to its high affinity to phosphatidylserine in the presence of Ca2+. We have also prepared PS-Phy and PS-MO cubosomes without ANX as a comparison, and characterized them using dynamic light scattering, cryo-TEM images and small-angle X-ray scattering, showing that PS-Phy cubosomes have greater chemical stability, and that ANX-PS-Phy cubosomes have the potential for in vivo drug delivery. In addition, we have reconstituted an apoptotic biomimetic membrane on a surface to gain insights into cubosome-bilayer interactions using a quartz-crystal microbalance and neutron reflectometry. The neutron reflectivity data reveal that there is exchange of materials between the biomimetic apoptotic bilayer and ANX-PS-Phy cubosomes, with an accumulation of ANX between the membrane and cubosomes possibly being the reason for the reduced cytotoxicity of ANX-PS-Phy cubosomes. A rat model of laser-induced choroidal neovascularization showed that ANX-PS-Phy cubosomes specifically targeted apoptotic cells in vivo. We propose that ANX-PS-Phy cubosomes are a potential candidate for ocular drug delivery for eye diseases.

16.
ACS Chem Biol ; 13(1): 121-130, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29182311

RESUMO

Polymyxins are last-line antibiotics against life-threatening multidrug-resistant Gram-negative bacteria. Unfortunately, polymyxin resistance is increasingly reported, leaving a total lack of therapies. Using lipidomics and transcriptomics, we discovered that polymyxin B induced lipid A deacylation via pagL in both polymyxin-resistant and -susceptible Pseudomonas aeruginosa. Our results demonstrated that the deacylation of lipid A is an "innate immunity" response to polymyxins and a key compensatory mechanism to the aminoarabinose modification to confer high-level polymyxin resistance in P. aeruginosa. Furthermore, cutting-edge neutron reflectometry studies revealed that an assembled outer membrane (OM) with the less hydrophobic penta-acylated lipid A decreased polymyxin B penetration, compared to the hexa-acylated form. Polymyxin analogues with enhanced hydrophobicity displayed superior penetration into the tail regions of the penta-acylated lipid A OM. Our findings reveal a previously undiscovered mechanism of polymyxin resistance, wherein polymyxin-induced lipid A remodeling affects the OM packing and hydrophobicity, perturbs polymyxin penetration, and thereby confers high-level resistance.


Assuntos
Antibacterianos/farmacologia , Lipídeo A/metabolismo , Polimixinas/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Acilação , Amino Açúcares/química , Amino Açúcares/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Farmacorresistência Bacteriana/efeitos dos fármacos , Lipídeo A/química , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
17.
PLoS One ; 11(10): e0164035, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27711151

RESUMO

Infrared (IR) spectroscopy has been used to quantify chemical and structural characteristics of a wide range of materials including biological tissues. In this study, we examined spatial changes in the chemical characteristics of rat retina in response to intraocular pressure (IOP) elevation using synchrotron infrared microscopy (SIRM), a non-destructive imaging approach. IOP elevation was induced by placing a suture around the eye of anaesthetised rats. Retinal sections were collected onto transparent CaF2 slides 10 days following IOP elevation. Using combined SIRM spectra and chemical mapping approaches it was possible to quantify IOP induced changes in protein conformation and chemical distribution in various layers of the rat retina. We showed that 10 days following IOP elevation there was an increase in lipid and protein levels in the inner nuclear layer (INL) and ganglion cell layer (GCL). IOP elevation also resulted in an increase in nucleic acids in the INL. Analysis of SIRM spectra revealed a shift in amide peaks to lower vibrational frequencies with a more prominent second shoulder, which is consistent with the presence of cell death in specific layers of the retina. These changes were more substantial in the INL and GCL layers compared with those occurring in the outer nuclear layer. These outcomes demonstrate the utility of SIRM to quantify the effect of IOP elevation on specific layers of the retina. Thus SIRM may be a useful tool for the study of localised tissue changes in glaucoma and other eye diseases.


Assuntos
Raios Infravermelhos , Pressão Intraocular , Microscopia/instrumentação , Retina/fisiologia , Síncrotrons , Animais , Conformação Proteica , Ratos , Retina/metabolismo
18.
Nat Microbiol ; 1: 15034, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-27572165

RESUMO

Human pathogenic Legionella replicate in alveolar macrophages and cause a potentially lethal form of pneumonia known as Legionnaires' disease(1). Here, we have identified a host-directed therapeutic approach to eliminate intracellular Legionella infections. We demonstrate that the genetic deletion, or pharmacological inhibition, of the host cell pro-survival protein BCL-XL induces intrinsic apoptosis of macrophages infected with virulent Legionella strains, thereby abrogating Legionella replication. BCL-XL is essential for the survival of Legionella-infected macrophages due to bacterial inhibition of host-cell protein synthesis, resulting in reduced levels of the short-lived, related BCL-2 pro-survival family member, MCL-1. Consequently, a single dose of a BCL-XL-targeted BH3-mimetic therapy, or myeloid cell-restricted deletion of BCL-XL, limits Legionella replication and prevents lethal lung infections in mice. These results indicate that repurposing BH3-mimetic compounds, originally developed to induce cancer cell apoptosis, may have efficacy in treating Legionnaires' and other diseases caused by intracellular microbes.


Assuntos
Apoptose , Legionella/crescimento & desenvolvimento , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/fisiologia , Proteína bcl-X/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Deleção de Genes , Legionelose/prevenção & controle , Camundongos , Proteína bcl-X/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA