Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Innate Immun ; 11(5): 432-444, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30485856

RESUMEN

Antimicrobial peptides (AMPs) are a diverse family of peptides that defend the mucosal surfaces of the oral cavity and other locations. Many AMPs have multiple functions and properties that influence aspects of innate defense and colonization by microorganisms. The human oral cavity is home to the second-most diverse microbiome, and the health of the mouth is influenced by the presence of these bacteria as well as by extrinsic factors such as periodontitis and smoking. This study hypothesized that the AMP profile is different in the presence of extrinsic factors and that this would also be reflected in the bacteria present. The AMP profile was analyzed by quantitative selected-reaction-monitoring mass spectrometry analysis and 40 bacterial species were quantified by DNA-DNA hybridization in saliva donated by 41 individuals. Periodontal status was assessed through dental examination and smoking status through medical charting. Periodontal health (in nonsmokers) was associated with a higher abundance of ribonuclease 7, protachykinin 1, ß-defensin 128, lipocalin 1, bactericidal permeability-increasing protein fold-containing family B member 3, and bone-marrow proteoglycan. Nonsmoking periodontal disease was associated with an abundance of neutrophil defensin 1 and cathelicidin. However, 7 AMPs were overabundant in periodontal disease in smokers: adrenomedullin, eosinophil peroxidase, 3 different histones, myeloperoxidase, and neutrophil defensin 1. There were no differentially abundant AMPs in smokers versus nonsmokers with periodontal health. Correlation network inference of healthy nonsmokers, healthy smokers, nonsmoking periodontitis, or smoking periodontitis donors demonstrated very different networks growing in complexity with increasing numbers of stressors. The study highlights the importance of the interaction between the oral cavity and its resident microbiota and how this may be influenced by periodontal disease and smoking.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Microbiota , Boca/microbiología , Saliva/metabolismo , Adulto , Anciano , Péptidos Catiónicos Antimicrobianos/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Microbiota/genética , Persona de Mediana Edad , Boca/metabolismo , Periodontitis/metabolismo , Periodontitis/microbiología , Fumar/efectos adversos , Suecia
2.
PLoS One ; 13(1): e0192051, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29385206

RESUMEN

Cutibacterium (Propionibacterium) acnes, considered a part of the skin microbiota, is one of the most commonly isolated anaerobic bacteria from medical implants in contact with plasma. However, the precise interaction of C. acnes with blood cells and plasma proteins has not been fully elucidated. Herein, we have investigated the molecular interaction of C. acnes with platelets and plasma proteins. We report that the ability of C. acnes to aggregate platelets is dependent on phylotype, with a significantly lower ability amongst type IB isolates, and the interaction of specific donor-dependent plasma proteins (or concentrations thereof) with C. acnes. Pretreatment of C. acnes with plasma reduces the lag time before aggregation demonstrating that pre-deposition of plasma proteins on C. acnes is an important step in platelet aggregation. Using mass spectrometry we identified several plasma proteins deposited on C. acnes, including IgG, fibrinogen and complement factors. Inhibition of IgG, fibrinogen or complement decreased C. acnes-mediated platelet aggregation, demonstrating the importance of these plasma proteins for aggregation. The interaction of C. acnes and platelets was visualized using fluorescence microscopy, verifying the presence of IgG and fibrinogen as components of the aggregates, and co-localization of C. acnes and platelets in the aggregates. Here, we have demonstrated the ability of C. acnes to activate and aggregate platelets in a bacterium and donor-specific fashion, as well as added mechanistic insights into this interaction.


Asunto(s)
Activación Plaquetaria , Agregación Plaquetaria , Propionibacterium acnes/fisiología , Proteínas Sanguíneas/metabolismo , Humanos , Espectrometría de Masas , Microscopía Fluorescente
3.
Mol Cell Proteomics ; 16(4 suppl 1): S29-S41, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28183813

RESUMEN

Sepsis is a systemic immune response responsible for considerable morbidity and mortality. Molecular modeling of host-pathogen interactions in the disease state represents a promising strategy to define molecular events of importance for the transition from superficial to invasive infectious diseases. Here we used the Gram-positive bacterium Streptococcus pyogenes as a model system to establish a mass spectrometry based workflow for the construction of a stoichiometric surface density model between the S. pyogenes surface, the surface virulence factor M-protein, and adhered human blood plasma proteins. The workflow relies on stable isotope labeled reference peptides and selected reaction monitoring mass spectrometry analysis of a wild-type strain and an M-protein deficient mutant strain, to generate absolutely quantified protein stoichiometry ratios between S. pyogenes and interacting plasma proteins. The stoichiometry ratios in combination with a novel targeted mass spectrometry method to measure cell numbers enabled the construction of a stoichiometric surface density model using protein structures available from the protein data bank. The model outlines the topology and density of the host-pathogen protein interaction network on the S. pyogenes bacterial surface, revealing a dense and highly organized protein interaction network. Removal of the M-protein from S. pyogenes introduces a drastic change in the network topology, validated by electron microscopy. We propose that the stoichiometric surface density model of S. pyogenes in human blood plasma represents a scalable framework that can continuously be refined with the emergence of new results. Future integration of new results will improve the understanding of protein-protein interactions and their importance for bacterial virulence. Furthermore, we anticipate that the general properties of the developed workflow will facilitate the production of stoichiometric surface density models for other types of host-pathogen interactions.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas Portadoras/metabolismo , Espectrometría de Masas/métodos , Proteómica/métodos , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/fisiología , Interacciones Huésped-Patógeno , Humanos , Marcaje Isotópico , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Unión Proteica , Mapas de Interacción de Proteínas , Streptococcus pyogenes/metabolismo
4.
Int J Med Microbiol ; 306(7): 504-516, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27423808

RESUMEN

Streptococcus pyogenes is an important human pathogen responsible for substantial morbidity and mortality worldwide. Although S. pyogenes is a strictly human pathogen with no other known animal reservoir, several murine infection models exist to explore different aspects of the bacterial pathogenesis. Inoculating mice with wild-type S. pyogenes strains can result in the generation of new bacterial phenotypes that are hypervirulent compared to the original inoculum. In this study, we used a serial mass spectrometry based proteomics strategy to investigate if these hypervirulent strains have an altered distribution of virulence proteins across the intracellular, surface associated and secreted bacterial compartments and if any change in compartmentalization can alter the protein-protein interaction network between bacteria and host proteins. Quantitative analysis of the S. pyogenes surface and secreted proteomes revealed that animal passaged strains are associated with significantly higher amount of virulence factors on the bacterial surface and in the media. This altered virulence factor compartmentalization results in increased binding of several mouse plasma proteins to the bacterial surface, a trend that was consistent for mouse plasma from several different mouse strains. In general, both the wild-type strain and animal passaged strain were capable of binding high amounts of human plasma proteins. However, compared to the non-passaged strains, the animal passaged strains displayed an increased ability to bind mouse plasma proteins, in particular for M protein binders, indicating that the increased affinity for mouse blood plasma proteins is a consequence of host adaptation of this pathogen to a new host. In conclusion, plotting the total amount of virulence factors against the total amount of plasma proteins associated to the bacterial surface could clearly separate out animal passaged strains from wild type strains indicating a virulence model that could predict the virulence of a S. pyogenes strain in mice and which could be used to identify key aspects of this bacteria's pathogenesis.


Asunto(s)
Proteínas Bacterianas/análisis , Interacciones Huésped-Patógeno , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/química , Streptococcus pyogenes/fisiología , Factores de Virulencia/análisis , Animales , Proteínas Sanguíneas/análisis , Modelos Animales de Enfermedad , Femenino , Espectrometría de Masas , Proteínas de la Membrana/análisis , Ratones Endogámicos BALB C , Unión Proteica , Mapas de Interacción de Proteínas , Proteoma/análisis
5.
Nat Commun ; 7: 10261, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26732734

RESUMEN

The plasma proteome is highly dynamic and variable, composed of proteins derived from surrounding tissues and cells. To investigate the complex processes that control the composition of the plasma proteome, we developed a mass spectrometry-based proteomics strategy to infer the origin of proteins detected in murine plasma. The strategy relies on the construction of a comprehensive protein tissue atlas from cells and highly vascularized organs using shotgun mass spectrometry. The protein tissue atlas was transformed to a spectral library for highly reproducible quantification of tissue-specific proteins directly in plasma using SWATH-like data-independent mass spectrometry analysis. We show that the method can determine drastic changes of tissue-specific protein profiles in blood plasma from mouse animal models with sepsis. The strategy can be extended to several other species advancing our understanding of the complex processes that contribute to the plasma proteome dynamics.


Asunto(s)
Plasma/química , Proteínas/clasificación , Proteómica/métodos , Sepsis/sangre , Infecciones Estreptocócicas/sangre , Streptococcus pyogenes , Animales , Biomarcadores , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Espectrometría de Masas/métodos , Ratones , Proteínas/química , Proteínas/metabolismo , Reproducibilidad de los Resultados , Sepsis/microbiología , Infecciones Estreptocócicas/microbiología
6.
J Proteomics ; 129: 98-107, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26381203

RESUMEN

The increasing number of bacterial genomes in combination with reproducible quantitative proteome measurements provides new opportunities to explore how genetic differences modulate proteome composition and virulence. It is challenging to combine genome and proteome data as the underlying genome influences the proteome. We present a strategy to facilitate the integration of genome data from several genetically similar bacterial strains with data-independent analysis mass spectrometry (DIA-MS) for rapid interrogation of the combined data sets. The strategy relies on the construction of a composite genome combining all genetic data in a compact format, which can accommodate the fusion with quantitative peptide and protein information determined via DIA-MS. We demonstrate the method by combining data sets from whole genome sequencing, shotgun MS and DIA-MS from 34 clinical isolates of Streptococcus pyogenes. The data structure allows for fast exploration of the data showing that undetected proteins are on average more amenable to amino acid substitution than expressed proteins. We identified several significantly differentially expressed proteins between invasive and non-invasive strains. The work underlines how integration of whole genome sequencing with accurately quantified proteomes can further advance the interpretation of the relationship between genomes, proteomes and virulence. This article is part of a Special Issue entitled: Computational Proteomics.


Asunto(s)
Proteínas Bacterianas/genética , Genoma Bacteriano/genética , Proteoma/genética , Proteómica/métodos , Análisis de Secuencia de ADN/métodos , Streptococcus pyogenes/genética , Mapeo Cromosómico/métodos , Humanos , Espectrometría de Masas
7.
Biochim Biophys Acta ; 1820(9): 1366-72, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22285770

RESUMEN

BACKGROUND: Changes in glycosylation of serum proteins are common, and various glycoforms are being explored as biomarkers in cancer and inflammation. We recently showed that glycoforms detected by endogenous galectins not only provide potential biomarkers, but also have different functions when they encounter galectins in tissue cells. Now we have explored the use of a combination of two galectins with different specificities, to further increase biomarker sensitivity and specificity. METHODS: Sera from 14 women with metastatic breast cancer, 12 healthy controls, 14 patients with IgA-nephritis (IgAN), and 12 patients with other glomerulonephritis were fractionated by affinity chromatography on immobilized human galectin-1 or galectin-8N, and the protein amounts of the bound and unbound fractions for each galectin were determined. RESULTS: Each galectin bound largely different fractions of the serum glycoproteins, including different glycoforms of haptoglobin. In the cancer sera, the level of galectin-1 bound glycoproteins was higher and galectin-8N bound glycoproteins lower compared to the other patients groups, whereas in IgAN sera the level of galectin-8N bound glycoproteins were higher. CONCLUSION: The ratio of galectin-1 bound/galectin-8N bound glycoproteins showed high discriminatory power between cancer patients and healthy, with AUC of 0.98 in ROC analysis, and thus provides an interesting novel cancer biomarker candidate. GENERAL SIGNIFICANCE: The galectin-binding ability of a glycoprotein is not only a promising biomarker candidate but may also have a specific function when the glycoprotein encounters the galectin in tissue cells, and thus be related to the pathophysiological state of the patient. This article is part of a Special Issue entitled Glycoproteomics.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Galectina 1/metabolismo , Galectinas/metabolismo , Glicoproteínas/metabolismo , Inflamación/diagnóstico , Neoplasias/diagnóstico , Suero/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Neoplasias de la Mama/sangre , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Carcinoma/sangre , Carcinoma/diagnóstico , Carcinoma/patología , Estudios de Casos y Controles , Cromatografía de Afinidad , Femenino , Glicoproteínas/sangre , Humanos , Inflamación/metabolismo , Inflamación/patología , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica/fisiología , Suero/química
8.
Am J Respir Cell Mol Biol ; 46(2): 240-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21960546

RESUMEN

Bacterial colonization of the lower respiratory tract is frequently seen in chronic obstructive pulmonary disease (COPD), and may cause exacerbations leading to disease progression. Antimicrobial peptides comprise an important part of innate lung immunity, and not least the cathelicidin human cationic antimicrobial protein-18/LL-37. Peptidylarginine deiminases (PADIs) post-translationally modify proteins by converting cationic peptidylarginine residues to neutral peptidylcitrulline. An increased presence of PADI2 and citrullinated proteins was demonstrated in the lungs of smokers. In this study, preformed PADI4, stored in granulocytes and extracellularly in the lumina of bronchi, was found in lung tissue of individuals suffering from COPD. In vitro, recombinant human PADI2 and PADI4 both caused a time- and dose-dependent citrullination of LL-37. The citrullination resulted in impaired antibacterial activity against Staphylococcus aureus, Streptococcus pneumoniae, and nontypable Haemophilus influenzae, but less so against Pseudomonas aeruginosa. Using artificial lipid bilayers, we observed discrete differences when comparing the disrupting activity of native and citrullinated LL-37, suggesting that differences in cell wall composition are important during interactions with whole bacteria. Furthermore, citrullinated LL-37 showed higher chemotactic activity against mononuclear leukocytes than did native LL-37, but was less efficient at neutralizing lipolysaccharide, and also in converting apoptotic neutrophils into a state of secondary necrosis. In addition, citrullinated LL-37 was more prone to degradation by proteases, whereas the V8 endopetidase of S. aureus cleaved the modified peptide at additional sites, compared with native LL-37. Together, these findings demonstrate novel mechanisms whereby the inflammation-dependent deiminases PADI2 and PADI4 can alter the activites of antibacterial polypeptides, affecting the course of inflammatory disorders such as COPD.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Bronquios/enzimología , Citrulina/metabolismo , Hidrolasas/metabolismo , Inflamación/enzimología , Fumar , Tráquea/enzimología , Péptidos Catiónicos Antimicrobianos/metabolismo , Dicroismo Circular , Electroforesis en Gel de Poliacrilamida , Haemophilus influenzae/fisiología , Inmunohistoquímica , Espectrometría de Masas , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica , Proteolisis , Pseudomonas aeruginosa/fisiología , Staphylococcus aureus/fisiología , Streptococcus pneumoniae/fisiología , Catelicidinas
9.
J Clin Immunol ; 32(2): 246-55, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22173878

RESUMEN

BACKGROUND: Immunoglobulin A nephritis (IgAN) is the most common primary glomerulonephritis worldwide. It is caused by accumulation of IgA1-containing immune complexes in the kidney resulting in renal failure, which is thought to be due to altered glycosylation of IgA with a decrease of 2-3-sialylated galactosides (NeuAcα2-3Gal). PURPOSE: The purpose of this study was to analyze whether altered glycosylation of IgA would lead to an altered binding to galectin-8, an endogenous lectin with strong affinity for 2-3-sialylated galactosides. Galectins are a family of ß-galactoside-binding proteins; by binding various glycoproteins, they play important roles in the regulation of cellular functions in inflammation and immunity. Hence, an altered binding of IgA to galectin-8 could lead to pathologic immune functions, such as glomerulonephritis. METHODS: Affinity chromatography of serum glycoproteins on the human sialogalactoside-binding lectin galectin-8N permitted quantitation of bound and unbound fractions, including IgA. RESULTS: Analysis of ~100 IgA nephritis sera showed that the galectin-8N unbound fraction of IgA increased compared to ~100 controls, consistent with the known loss of galactosylation. A subgroup of ~15% of the IgAN patients had a ratio of galectin-8 bound/unbound IgA <0.09, not found for any of the controls. Unexpectedly, the galectin-8N-binding fraction of serum glycoproteins other than IgA increased in the sera of IgAN patients but not in controls, suggesting a previously unrecognized change in this disease. CONCLUSION: This is the first study that relates a galectin, an endogenous lectin family, to IgA nephritis and thus should stimulate new avenues of research into the pathophysiology of the disease.


Asunto(s)
Galectinas/metabolismo , Glomerulonefritis por IGA/metabolismo , Glicoproteínas/metabolismo , Inmunoglobulina A/metabolismo , Adulto , Anciano , Progresión de la Enfermedad , Femenino , Glomerulonefritis por IGA/inmunología , Glicoproteínas/sangre , Glicoproteínas/inmunología , Humanos , Inmunoglobulina A/inmunología , Masculino , Persona de Mediana Edad , Unión Proteica , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA