Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunol Res ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38748319

RESUMEN

The gut microbiome can modulate systemic inflammation and is therefore target for immunomodulation. Immunomodulating effects of EDP1815, a bacterial commensal strain of Prevotella histicola, were studied in healthy participants. Effects on adaptive immunity were evaluated by a neo-antigen challenge with keyhole limpet haemocyanin (KLH), while effects on innate immunity were evaluated by topical toll-like receptor 7 (TLR7) agonist imiquimod. Capsules with two enteric coating levels (EC1, EC2) were compared. Thirty-six healthy participants were included and received a daily dose of 8 × 1010 cells EDP1815-EC1, EDP1815-EC2 or placebo (randomization 1:1:1) for 60 days. They received KLH vaccinations at days 8, 24 and 36, with intradermal skin challenge at day 57. KLH challenge outcomes were antibody levels, and skin blood flow and erythema after skin challenge, measured by imaging techniques. Imiquimod administration started at day 57, for 72 h. Outcomes consisted of imaging measurements similar to the KLH challenge, and the influx of inflammatory cells and cytokines in blister fluid. There was no effect of EDP1815 treatment on the KLH challenge, neither on the imaging outcomes of the imiquimod challenge. There was a consistently lower influx of inflammatory cells in the blister fluid of EDP1815-treated participants (neutrophils, p = 0.016; granulocytes, p = 0.024), more pronounced in EC1. There was a lower influx of interleukin [IL]-1ß, IL-6, IL-8, IL-10, interferon [IFN]-γ and tumour necrosis factor in blister fluid of EDP1815-treated participants. EDP1815 had immunomodulatory effects on the innate immune response driven by imiquimod, but no effect on the KLH challenge was observed. Trial registration number: NCT05682222; date: 22 July 2022.

2.
Am J Respir Crit Care Med ; 209(6): 703-715, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-37972349

RESUMEN

Rationale: Acute respiratory distress syndrome (ARDS) has an unacceptably high mortality rate (35%) and is without effective therapy. Orai1 is a Ca2+ channel involved in store-operated Ca2+ entry (SOCE), a process that exquisitely regulates inflammation. Orai1 is considered a druggable target, but no Orai1-specific inhibitors exist to date. Objectives: To evaluate whether ELD607, a first-in-class Orai1 antagonist, can treat ARDS caused by bacterial pneumonia in preclinical models. Methods: ELD607 pharmacology was evaluated in HEK293T cells and freshly isolated immune cells from patients with ARDS. A murine acute lung injury model caused by bacterial pneumonia was then used: mice were infected with Pseudomonas aeruginosa, Staphylococcus aureus, methicillin-resistant S. aureus, or multidrug-resistant P. aeruginosa and then treated with ELD607 intranasally. Measurements and Main Results: ELD607 specifically inhibited SOCE in HEK293T cells with a half-maximal inhibitory concentration of 9 nM. ELD607 was stable in ARDS airway secretions and inhibited SOCE in ARDS immune cells. In vivo, inhaled ELD607 significantly reduced neutrophilia and improved survival. Surprisingly, Orai1 inhibition by ELD607 caused a significant reduction in lung bacteria, including methicillin-resistant S. aureus. ELD607 worked as an immunomodulator that reduced cytokine levels, reduced neutrophilia, and promoted macrophage-mediated resolution of inflammation and clearance of bacteria. Indeed, when alveolar macrophages were depleted with inhaled clodronate, ELD607 was no longer able to resolve inflammation or clear bacteria. Conclusions: These data indicate that specific Orai1 inhibition by ELD607 may be a novel approach to reduce multiorgan inflammation and treat antibiotic-resistant bacteria.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Neumonía Bacteriana , Síndrome de Dificultad Respiratoria , Humanos , Ratones , Animales , Canales de Calcio/metabolismo , Canales de Calcio/farmacología , Calcio/metabolismo , Células HEK293 , Staphylococcus aureus Resistente a Meticilina/metabolismo , Señalización del Calcio , Inflamación/tratamiento farmacológico , Pulmón/metabolismo , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Neumonía Bacteriana/tratamiento farmacológico , Proteína ORAI1/metabolismo , Proteína ORAI1/farmacología
3.
J Crohns Colitis ; 18(1): 82-90, 2024 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-37616127

RESUMEN

OBJECTIVE: Central reading of endoscopy is advocated by regulatory agencies for clinical trials in ulcerative colitis [UC]. It is uncertain whether the local/site reader should be included in the reading paradigm. We explore whether using locally- and centrally-determined endoscopic Mayo subscores [eMS] provide a reliable final assessment and whether the paradigm used has an impact on effect size. METHODS: eMS data from the TURANDOT [NCT01620255] study were used to retrospectively examine seven different reading paradigms (using the scores of local readers [LR], first central readers [CR1], second central readers [CR2], and various consensus reads [ConCR]) by assessing inter-rater reliabilities and their impact on the key study endpoint, endoscopic improvement. RESULTS: More than 40% of eMS scores between two trained central readers were discordant. Central readers had wide variability in scorings at baseline (intraclass correlation coefficient [ICC] of 0.475 [0.339, 0.610] for CR1 vs CR2). Centrally-read scores had variable concordance with LR (LR vs CR1 ICC 0.682 [0.575, 0.788], and LR vs CR2 ICC 0.526 [0.399, 0.653]). Reading paradigms with LR and CR which included a consensus, enhanced ICC estimates to >0.8. At Week 12, without the consensus reads, the CR1 vs CR2 ICC estimates were 0.775 [0.710, 0.841], and with consensus reads the ICC estimates were >0.9. Consensus-based approaches were most favourable to detect a treatment difference. CONCLUSION: The ICC between the eMS of two trained and experienced central readers is unexpectedly low, which reinforces that currently used central reading processes are still associated with several weaknesses.


Asunto(s)
Colitis Ulcerosa , Humanos , Colitis Ulcerosa/diagnóstico , Reproducibilidad de los Resultados , Estudios Retrospectivos , Endoscopía Gastrointestinal , Variaciones Dependientes del Observador
4.
Int J Biol Macromol ; 256(Pt 1): 128387, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38000593

RESUMEN

Alpha amylases catalyse the hydrolysis of α-1, 4-glycosidic bonds in starch, yielding glucose, maltose, dextrin, and short oligosaccharides, vital to various industrial processes. Structural and functional insights on α-amylase from Methanocaldococcus jannaschii were computationally explored to evaluate a catalytic domain and its fusion with a small ubiquitin-like modifier (SUMO). The recombinant proteins' production, characterization, ligand binding studies, and structural analysis of the cloned amylase native full gene (MjAFG), catalytic domain (MjAD) and fusion enzymes (S-MjAD) were thoroughly analysed in this comparative study. The MjAD and S-MjAD showed 2-fold and 2.5-fold higher specific activities (µmol min-1 mg -1) than MjAFG at 95 °C at pH 6.0. Molecular modelling and MD simulation results showed that the removal of the extra loop (178 residues) at the C-terminal of the catalytic domain exposed the binding and catalytic residues near its active site, which was buried in the MjAFG enzyme. The temperature ramping and secondary structure analysis of MjAFG, MjAD and S-MjAD through CD spectrometry showed no notable alterations in the secondary structures but verified the correct folding of MjA variants. The chimeric fusion of amylases with thermostable α-glucosidases makes it a potential candidate for the starch degrading processes.


Asunto(s)
Methanocaldococcus , alfa-Amilasas , alfa-Amilasas/química , Methanocaldococcus/metabolismo , Archaea/metabolismo , Amilasas/química , Almidón/metabolismo
5.
Physiol Rep ; 10(10): e15306, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35581745

RESUMEN

Orai1 is a ubiquitously-expressed plasma membrane Ca2+ channel that is involved in store-operated Ca2+ entry (SOCE): a fundamental biological process that regulates gene expression, the onset of inflammation, secretion, and the contraction of airway smooth muscle (ASM). During SOCE, Ca2+ leaves the endoplasmic reticulum, which then stimulates a second, amplifying wave of Ca2+ influx through Orai1 into the cytoplasm. Short Palate LUng and Nasal epithelial Clone 1 (SPLUNC1; gene name BPIFA1) is a multi-functional, innate defense protein that is highly abundant in the lung. We have previously reported that SPLUNC1 was secreted from epithelia, where it bound to and inhibited Orai1, leading to reduced SOCE and ASM relaxation. However, the underlying mechanism of action is unknown. Here, we probed the SPLUNC1-Orai1 interactions in ASM and HEK293T cells using biochemical and imaging techniques. We observed that SPLUNC1 caused a conformational change in Orai1, as measured using Forster resonance energy transfer (FRET). SPLUNC1 binding also led to Nedd4-2 dependent ubiquitination of Orai1. Moreover, SPLUNC1 internalized Orai1 to lysosomes, leading to Orai1 degradation. Thus, we conclude that SPLUNC1 is an allosteric regulator of Orai1. Our data indicate that SPLUNC1-mediated Orai1 inhibition could be utilized as a therapeutic strategy to reduce SOCE.


Asunto(s)
Glicoproteínas/metabolismo , Pulmón , Músculo Liso , Fosfoproteínas/metabolismo , Calcio/metabolismo , Señalización del Calcio/fisiología , Membrana Celular/metabolismo , Células HEK293 , Humanos , Pulmón/metabolismo , Músculo Liso/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo
6.
Am J Respir Cell Mol Biol ; 66(3): 271-282, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34807800

RESUMEN

Orai1 is a plasma membrane Ca2+ channel that mediates store-operated Ca2+ entry (SOCE) and regulates inflammation. Short palate lung and nasal epithelial clone 1 (SPLUNC1) is an asthma gene modifier that inhibits Orai1 and SOCE via its C-terminal α6 region. SPLUNC1 levels are diminished in asthma patient airways. Thus, we hypothesized that inhaled α6 peptidomimetics could inhibit Orai1 and reduce airway inflammation in a murine asthma model. To evaluate α6-Orai1 interactions, we used fluorescent assays to measure Ca2+ signaling, Förster resonance energy transfer, fluorescent recovery after photobleaching, immunostaining, total internal reflection microscopy, and Western blotting. To test whether α6 peptidomimetics inhibited SOCE and decreased inflammation in vivo, wild-type and SPLUNC1-/- mice were exposed to house dust mite (HDM) extract with or without α6 peptide. We also performed nebulization, jet milling, and scanning electron microscopy to evaluate α6 for inhalation. SPLUNC1-/- mice had an exaggerated response to HDM. In BAL-derived immune cells, Orai1 levels increased after HDM exposure in SPLUNC1-/- but not wild-type mice. Inhaled α6 reduced Orai1 levels in mice regardless of genotype. In HDM-exposed mice, α6 dose-dependently reduced eosinophilia and neutrophilia. In vitro, α6 inhibited SOCE in multiple immune cell types, and α6 could be nebulized or jet milled without loss of function. These data suggest that α6 peptidomimetics may be a novel, effective antiinflammatory therapy for patients with asthma.


Asunto(s)
Asma , Peptidomiméticos , Animales , Asma/tratamiento farmacológico , Calcio/metabolismo , Glicoproteínas , Humanos , Inflamación , Pulmón/metabolismo , Ratones , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Fosfoproteínas
8.
Respir Res ; 21(1): 190, 2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32680508

RESUMEN

Cystic fibrosis (CF) patients are extremely vulnerable to Burkholderia cepacia complex (Bcc) infections. However, the underlying etiology is poorly understood. We tested the hypothesis that short palate lung and nasal epithelial clone 1 (SPLUNC1)-epithelial sodium channel (ENaC) interactions at the plasma membrane are required to reduce Bcc burden in normal airways. To determine if SPLUNC1 was needed to reduce Bcc burden in the airways, SPLUNC1 knockout mice and their wild-type littermates were infected with B. cenocepacia strain J2315. SPLUNC1 knockout mice had increased bacterial burden in the lungs compared to wild-type littermate mice. SPLUNC1-knockdown primary human bronchial epithelia (HBECs) were incubated with J2315, which resulted in increased bacterial burden compared to non-transduced HBECs. We next determined the interaction of the SPLUNC1-ENaC complex during J2315 infection. SPLUNC1 remained at the apical plasma membrane of normal HBECs but less was present at the apical plasma membrane of CF HBECs. Additionally, SPLUNC1-ßENaC complexes reduced intracellular J2315 burden. Our data indicate that (i) secreted SPLUNC1 is required to reduce J2315 burden in the airways and (ii) its interaction with ENaC prevents cellular invasion of J2315.


Asunto(s)
Infecciones por Burkholderia/microbiología , Burkholderia cenocepacia/patogenicidad , Fibrosis Quística/microbiología , Células Epiteliales/microbiología , Canales Epiteliales de Sodio/metabolismo , Glicoproteínas/metabolismo , Pulmón/microbiología , Fosfoproteínas/metabolismo , Infecciones del Sistema Respiratorio/microbiología , Adolescente , Adulto , Animales , Carga Bacteriana , Infecciones por Burkholderia/genética , Infecciones por Burkholderia/metabolismo , Estudios de Casos y Controles , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/genética , Femenino , Glicoproteínas/genética , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fosfoproteínas/genética , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/metabolismo , Adulto Joven
11.
FASEB J ; 32(5): 2478-2491, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29295861

RESUMEN

Cystic fibrosis (CF) is a common genetic disease with significantly increased mortality. CF airways exhibit ion transport abnormalities, including hyperactivity of the epithelial Na+ channel (ENaC). Short-palate lung and nasal epithelial clone 1 (SPLUNC1) is a multifunctional innate defense protein that is secreted into the airway lumen. We have previously demonstrated that SPLUNC1 binds to and inhibits ENaC to maintain fluid homeostasis in airway epithelia and that this process fails in CF airways. Despite this, how SPLUNC1 actually regulates ENaC is unknown. Here, we found that SPLUNC1 caused αγ-ENaC to internalize, whereas SPLUNC1 and ß-ENaC remained at the plasma membrane. Additional studies revealed that SPLUNC1 increased neural precursor cell-expressed developmentally down-regulated protein 4-2-dependent ubiquitination of α- but not ß- or γ-ENaC. We also labeled intracellular ENaC termini with green fluorescent protein and mCherry, and found that extracellular SPLUNC1 altered intracellular ENaC Forster resonance energy transfer. Taken together, our data indicate that SPLUNC1 is an allosteric regulator of ENaC that dissociates αßγ-ENaC to generate a new SPLUNC1-ß-ENaC complex. These data indicate a novel mode for regulating ENaC at the plasma membrane.-Kim, C. S., Ahmad, S., Wu, T., Walton, W. G., Redinbo, M. R., Tarran, R. SPLUNC1 is an allosteric modulator of the epithelial sodium channel.


Asunto(s)
Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Glicoproteínas/metabolismo , Complejos Multiproteicos/química , Mucosa Nasal/metabolismo , Fosfoproteínas/metabolismo , Regulación Alostérica/fisiología , Membrana Celular/química , Membrana Celular/genética , Células Epiteliales/química , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Transferencia Resonante de Energía de Fluorescencia , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Proteínas Luminiscentes , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mucosa Nasal/química , Fosfoproteínas/química , Fosfoproteínas/genética , Proteína Fluorescente Roja
12.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L192-L205, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982737

RESUMEN

In cystic fibrosis (CF) lungs, epithelial Na+ channel (ENaC) hyperactivity causes a reduction in airway surface liquid volume, leading to decreased mucocilliary clearance, chronic bacterial infection, and lung damage. Inhibition of ENaC is an attractive therapeutic option. However, ENaC antagonists have failed clinically because of off-target effects in the kidney. The S18 peptide is a naturally occurring short palate lung and nasal epithelial clone 1 (SPLUNC1)-derived ENaC antagonist that restores airway surface liquid height for up to 24 h in CF human bronchial epithelial cultures. However, its efficacy and safety in vivo are unknown. To interrogate the potential clinical efficacy of S18, we assessed its safety and efficacy using human airway cultures and animal models. S18-mucus interactions were tested using superresolution microscopy, quartz crystal microbalance with dissipation, and confocal microscopy. Human and murine airway cultures were used to measure airway surface liquid height. Off-target effects were assessed in conscious mice and anesthetized rats. Morbidity and mortality were assessed in the ß-ENaC-transgenic (Tg) mouse model. Restoration of normal mucus clearance was measured in cystic fibrosis transmembrane conductance regulator inhibitor 172 [CFTR(inh)-172]-challenged sheep. We found that S18 does not interact with mucus and rapidly penetrated dehydrated CF mucus. Compared with amiloride, an early generation ENaC antagonist, S18 displayed a superior ability to slow airway surface liquid absorption, reverse CFTR(inh)-172-induced reduction of mucus transport, and reduce morbidity and mortality in the ß-ENaC-Tg mouse, all without inducing any detectable signs of renal toxicity. These data suggest that S18 is the first naturally occurring ENaC antagonist to show improved preclinical efficacy in animal models of CF with no signs of renal toxicity.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Glicoproteínas/metabolismo , Enfermedades Pulmonares/tratamiento farmacológico , Péptidos/farmacología , Fosfoproteínas/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Animales , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Transporte Iónico , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Masculino , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Mucosa Respiratoria/metabolismo
13.
Antimicrob Agents Chemother ; 60(10): 6003-12, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27458217

RESUMEN

The opportunistic bacteria of the Burkholderia cepacia complex (Bcc) are extremely pathogenic to cystic fibrosis (CF) patients, and acquisition of Bcc bacteria is associated with a significant increase in mortality. Treatment of Bcc infections is difficult because the bacteria are multidrug resistant and able to survive in biofilms. Short palate, lung, and nasal epithelial clone 1 (SPLUNC1) is an innate defense protein that is secreted by the upper airways and pharynx. While SPLUNC1 is known to have antimicrobial functions, its effects on Bcc strains are unclear. We therefore tested the hypothesis that SPLUNC1 is able to impair Bcc growth and biofilm formation. We found that SPLUNC1 exerted bacteriostatic effects against several Bcc clinical isolates, including B. cenocepacia strain J2315 (50% inhibitory concentration [IC50] = 0.28 µM), and reduced biofilm formation and attachment (IC50 = 0.11 µM). We then determined which domains of SPLUNC1 are responsible for its antimicrobial activity. Deletions of SPLUNC1's N terminus and α6 helix did not affect its function. However, deletion of the α4 helix attenuated antimicrobial activity, while the corresponding α4 peptide displayed antimicrobial activity. Chronic neutrophilia is a hallmark of CF lung disease, and neutrophil elastase (NE) cleaves SPLUNC1. However, we found that the ability of SPLUNC1 to disrupt biofilm formation was significantly potentiated by NE pretreatment. While the impact of CF on SPLUNC1-Bcc interactions is not currently known, our data suggest that understanding this interaction may have important implications for CF lung disease.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Complejo Burkholderia cepacia/efectos de los fármacos , Glicoproteínas/farmacología , Elastasa de Leucocito/química , Fosfoproteínas/farmacología , Antibacterianos/química , Antibacterianos/metabolismo , Sitios de Unión , Biopelículas/crecimiento & desarrollo , Complejo Burkholderia cepacia/crecimiento & desarrollo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad
14.
Front Microbiol ; 7: 696, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27252681

RESUMEN

Francisella (F.) philomiragia is a Gram-negative bacterium with a preference for brackish environments that has been implicated in causing bacterial infections in near-drowning victims. The purpose of this study was to characterize the ability of F. philomiragia to infect cultured mammalian cells, a commonly used invertebrate model, and, finally, to characterize the ability of F. philomiragia to infect BALB/c mice via the pulmonary (intranasal) route of infection. This study shows that F. philomiragia infects J774A.1 murine macrophage cells, HepG2 cells and A549 human Type II alveolar epithelial cells. However, replication rates vary depending on strain at 24 h. F. philomiragia infection after 24 h was found to be cytotoxic in human U937 macrophage-like cells and J774A.1 cells. This is in contrast to the findings that F. philomiragia was non-cytotoxic to human hepatocellular carcinoma cells, HepG2 cells and A549 cells. Differential cytotoxicity is a point for further study. Here, it was demonstrated that F. philomiragia grown in host-adapted conditions (BHI, pH 6.8) is sensitive to levofloxacin but shows increased resistance to the human cathelicidin LL-37 and murine cathelicidin mCRAMP when compared to related the Francisella species, F. tularensis subsp. novicida and F. tularensis subsp. LVS. Previous findings that LL-37 is strongly upregulated in A549 cells following F. tularensis subsp. novicida infection suggest that the level of antimicrobial peptide expression is not sufficient in cells to eradicate the intracellular bacteria. Finally, this study demonstrates that F. philomiragia is lethal in two in vivo models; Galleria mellonella via hemocoel injection, with a LD50 of 1.8 × 10(3), and BALB/c mice by intranasal infection, with a LD50 of 3.45 × 10(3). In conclusion, F. philomiragia may be a useful model organism to study the genus Francisella, particularly for those researchers with interest in studying microbial ecology or environmental strains of Francisella. Additionally, the Biosafety level 2 status of F. philomiragia makes it an attractive model for virulence and pathogenesis studies.

15.
Biochemistry ; 55(21): 2979-91, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27145151

RESUMEN

SPLUNC1 is an abundantly secreted innate immune protein in the mammalian respiratory tract that exerts bacteriostatic and antibiofilm effects, binds to lipopolysaccharide (LPS), and acts as a fluid-spreading surfactant. Here, we unravel the structural elements essential for the surfactant and antimicrobial functions of human SPLUNC1 (short palate lung nasal epithelial clone 1). A unique α-helix (α4) that extends from the body of SPLUNC1 is required for the bacteriostatic, surfactant, and LPS binding activities of this protein. Indeed, we find that mutation of just four leucine residues within this helical motif to alanine is sufficient to significantly inhibit the fluid spreading abilities of SPLUNC1, as well as its bacteriostatic actions against Gram-negative pathogens Burkholderia cenocepacia and Pseudomonas aeruginosa. Conformational flexibility in the body of SPLUNC1 is also involved in the bacteriostatic, surfactant, and LPS binding functions of the protein as revealed by disulfide mutants introduced into SPLUNC1. In addition, SPLUNC1 exerts antibiofilm effects against Gram-negative bacteria, although α4 is not involved in this activity. Interestingly, though, the introduction of surface electrostatic mutations away from α4 based on the unique dolphin SPLUNC1 sequence, and confirmed by crystal structure, is shown to impart antibiofilm activity against Staphylococcus aureus, the first SPLUNC1-dependent effect against a Gram-positive bacterium reported to date. Together, these data pinpoint SPLUNC1 structural motifs required for the antimicrobial and surfactant actions of this protective human protein.


Asunto(s)
Antiinfecciosos/farmacología , Bronquios/efectos de los fármacos , Burkholderia cenocepacia/efectos de los fármacos , Glicoproteínas/química , Glicoproteínas/metabolismo , Lipopolisacáridos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Secuencia de Aminoácidos , Biopelículas/efectos de los fármacos , Bronquios/citología , Burkholderia cenocepacia/inmunología , Células Cultivadas , Cristalización , Cristalografía por Rayos X , Glicoproteínas/genética , Humanos , Inmunidad Innata/efectos de los fármacos , Fosfoproteínas/genética , Conformación Proteica , Pseudomonas aeruginosa/inmunología , Surfactantes Pulmonares/química , Surfactantes Pulmonares/metabolismo
16.
Trop Doct ; 42(2): 94-6, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22328602

RESUMEN

Our study compares the risk factors, clinical presentations and outcomes of pulmonary infections caused by Nocardia asteroides and non-asteroides species. We performed a retrospective cohort study comparing pulmonary infections by both species in patients presenting to a tertiary care hospital in Karachi, Pakistan. Forty-one patients were identified with pulmonary nocardiosis, with 58.5% belonging to the N. asteroids complex. The most common clinical findings were fever and a cough for both groups, with lobar infiltrates being the most common finding on chest radiographs. In vitro testing showed a sensitivity of all species to trimethoprim-sulfamethoxazole (TMP-SMZ), aminoglycosides, ceftriaxone and imipenem. The majority of the patients were treated with TMP-SMZ in combination with other drugs. The results of our study suggest that there is no significant difference in the risk factors, presentations and outcomes of pulmonary infections by N. asteroides and non-asteroides species. Immunocompromised patients are more likely to have unfavorable outcomes.


Asunto(s)
Enfermedades Pulmonares/fisiopatología , Nocardiosis/fisiopatología , Nocardia asteroides/patogenicidad , Nocardia/patogenicidad , Adulto , Anciano , Antibacterianos/uso terapéutico , Estudios de Cohortes , Femenino , Hospitales Universitarios/estadística & datos numéricos , Humanos , Huésped Inmunocomprometido , Enfermedades Pulmonares/epidemiología , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Masculino , Persona de Mediana Edad , Nocardia/clasificación , Nocardiosis/epidemiología , Nocardiosis/microbiología , Nocardiosis/patología , Pakistán/epidemiología , Estudios Retrospectivos , Factores de Riesgo , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico
17.
BMC Microbiol ; 10: 123, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20416090

RESUMEN

BACKGROUND: Macrolide antibiotics are commonly administered for bacterial respiratory illnesses. Azithromycin (Az) is especially noted for extremely high intracellular concentrations achieved within macrophages which is far greater than the serum concentration. Clinical strains of Type B Francisella (F.) tularensis have been reported to be resistant to Az, however our laboratory Francisella strains were found to be sensitive. We hypothesized that different strains/species of Francisella (including Type A) may have different susceptibilities to Az, a widely used and well-tolerated antibiotic. RESULTS: In vitro susceptibility testing of Az confirmed that F. tularensis subsp. holarctica Live Vaccine Strain (LVS) (Type B) was not sensitive while F. philomiragia, F. novicida, and Type A F. tularensis (NIH B38 and Schu S4 strain) were susceptible. In J774A.1 mouse macrophage cells infected with F. philomiragia, F. novicida, and F. tularensis LVS, 5 microg/ml Az applied extracellularly eliminated intracellular Francisella infections. A concentration of 25 microg/ml Az was required for Francisella-infected A549 human lung epithelial cells, suggesting that macrophages are more effective at concentrating Az than epithelial cells. Mutants of RND efflux components (tolC and ftlC) in F. novicida demonstrated less sensitivity to Az by MIC than the parental strain, but the tolC disc-inhibition assay demonstrated increased sensitivity, indicating a complex role for the outer-membrane transporter. Mutants of acrA and acrB mutants were less sensitive to Az than the parental strain, suggesting that AcrAB is not critical for the efflux of Az in F. novicida. In contrast, F. tularensis Schu S4 mutants DeltaacrB and DeltaacrA were more sensitive than the parental strain, indicating that the AcrAB may be important for Az efflux in F. tularensis Schu S4. F. novicida LPS O-antigen mutants (wbtN, wbtE, wbtQ and wbtA) were found to be less sensitive in vitro to Az compared to the wild-type. Az treatment prolonged the survival of Galleria (G.) mellonella infected with Francisella. CONCLUSION: These studies demonstrate that Type A Francisella strains, as well as F. novicida and F. philomiragia, are sensitive to Az in vitro. Francisella LPS and the RND efflux pump may play a role in Az sensitivity. Az also has antimicrobial activity against intracellular Francisella, suggesting that the intracellular concentration of Az is high enough to be effective against multiple strains/species of Francisella, especially in macrophages. Az treatment prolonged survival an in vivo model of Francisella-infection.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Células Epiteliales/microbiología , Francisella/efectos de los fármacos , Macrófagos/microbiología , Animales , Antibacterianos/uso terapéutico , Azitromicina/uso terapéutico , Línea Celular , Humanos , Lepidópteros/microbiología , Ratones , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Análisis de Supervivencia
18.
J Immunol ; 181(12): 8298-307, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19050246

RESUMEN

Insulin is a major target for the autoimmune-mediated destruction of pancreatic beta cells during the pathogenesis of type I diabetes. A plasmid DNA vaccine encoding mouse proinsulin II reduced the incidence of diabetes in a mouse model of type I diabetes when administered to hyperglycemic (therapeutic mode) or normoglycemic (prophylactic mode) NOD mice. Therapeutic administration of proinsulin DNA was accompanied by a rapid decrease in the number of insulin-specific IFN-gamma-producing T cells, whereas prophylactic treatment was accompanied by enhanced IFN-gamma-secreting cells and a decrease in insulin autoantibodies. Adoptive transfer experiments demonstrated that the protection was not mediated by induction of CD25(+)/CD4(+) T regulatory cells. The efficacy of the DNA vaccine was enhanced by increasing the level of expression of the encoded Ag, more frequent dosing, increasing dose level, and localization of the protein product to the intracellular compartment. The efficacy data presented in this study demonstrate that Ag-specific plasmid DNA therapy is a viable strategy for preventing progression of type I diabetes and defines critical parameters of the dosing regime that influences tolerance induction.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Hiperglucemia/prevención & control , Tolerancia Inmunológica , Líquido Intracelular/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Secuencia de Aminoácidos , Animales , Autoantígenos/administración & dosificación , Autoantígenos/biosíntesis , Autoantígenos/genética , Autoantígenos/inmunología , Línea Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/prevención & control , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Hiperglucemia/genética , Hiperglucemia/inmunología , Hiperglucemia/patología , Tolerancia Inmunológica/genética , Líquido Intracelular/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Datos de Secuencia Molecular , Plásmidos/administración & dosificación , Plásmidos/genética , Plásmidos/inmunología , Estado Prediabético/genética , Estado Prediabético/inmunología , Estado Prediabético/patología , Estado Prediabético/terapia , Proinsulina/administración & dosificación , Proinsulina/biosíntesis , Proinsulina/genética , Proinsulina/inmunología , Ensayos Clínicos Controlados Aleatorios como Asunto , Vacunas de ADN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...