Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Physiol ; 12: 645650, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746781

RESUMEN

α1-microglobulin (A1M) is found in all vertebrates including humans. A1M was, together with retinol-binding protein and ß-lactoglobulin, one of the three original lipocalins when the family first was proposed in 1985. A1M is described as an antioxidant and tissue cleaning protein with reductase, heme- and radical-binding activities. These biochemical properties are driven by a strongly electronegative surface-exposed thiol group, C34, on loop 1 of the open end of the lipocalin barrel. A1M has been shown to have protective effects in vitro and in vivo in cell-, organ-, and animal models of oxidative stress-related medical conditions. The gene coding for A1M is unique among lipocalins since it is flanked downstream by four exons coding for another non-lipocalin protein, bikunin, and is consequently named α1-microglobulin-bikunin precursor gene (AMBP). The precursor is cleaved in the Golgi, and A1M and bikunin are secreted from the cell separately. Recent publications have suggested novel physiological roles of A1M in regulation of endoplasmic reticulum activities and erythrocyte homeostasis. This review summarizes the present knowledge of the structure and functions of the lipocalin A1M and presents a current model of its biological role(s).

2.
Redox Biol ; 41: 101892, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33607500

RESUMEN

Heparin and heparan sulfate (HS) are linear sulfated disaccharide polymers. Heparin is found mainly in mast cells, while heparan sulfate is found in connective tissue, extracellular matrix and on cell membranes in most tissues. α1-microglobulin (A1M) is a ubiquitous protein with thiol-dependent antioxidant properties, protecting cells and matrix against oxidative damage due to its reductase activities and radical- and heme-binding properties. In this work, it was shown that A1M binds to heparin and HS and can be purified from human plasma by heparin affinity chromatography and size exclusion chromatography. The binding strength is inversely dependent of salt concentration and proportional to the degree of sulfation of heparin and HS. Potential heparin binding sites, located on the outside of the barrel-shaped A1M molecule, were determined using hydrogen deuterium exchange mass spectrometry (HDX-MS). Immunostaining of endothelial cells revealed pericellular co-localization of A1M and HS and the staining of A1M was almost completely abolished after treatment with heparinase. A1M and HS were also found to be co-localized in vivo in the lungs, aorta, kidneys and skin of mice. The redox-active thiol group of A1M was unaffected by the binding to HS, and the cell protection and heme-binding abilities of A1M were slightly affected. The discovery of the binding of A1M to heparin and HS provides new insights into the biological role of A1M and represents the basis for a novel method for purification of A1M from plasma.


Asunto(s)
Células Endoteliales , Heparina , alfa-Globulinas , Animales , Sitios de Unión , Heparitina Sulfato , Humanos , Ratones , Unión Proteica
3.
Free Radic Biol Med ; 162: 149-159, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32092412

RESUMEN

During red blood cell (RBC) lysis hemoglobin and heme leak out of the cells and cause damage to the endothelium and nearby tissue. Protective mechanisms exist; however, these systems are not sufficient in diseases with increased extravascular hemolysis e.g. hemolytic anemia. α1-microglobulin (A1M) is a ubiquitous reductase and radical- and heme-binding protein with antioxidation properties. Although present in the circulation in micromolar concentrations, its function in blood is unclear. Here, we show that A1M provides RBC stability. A1M-/- mice display abnormal RBC morphology, reminiscent of macrocytic anemia conditions, i.e. fewer, larger and more heterogeneous cells. Recombinant human A1M (rA1M) reduced in vitro hemolysis of murine RBC against spontaneous, osmotic and heme-induced stress. Moreover, A1M is taken up by human RBCs both in vitro and in vivo. Similarly, rA1M also protected human RBCs against in vitro spontaneous, osmotic, heme- and radical-induced hemolysis as shown by significantly reduced leakage of hemoglobin and LDH. Addition of rA1M resulted in decreased hemolysis compared to addition of the heme-binding protein hemopexin and the radical-scavenging and reducing agents ascorbic acid and Trolox (vitamin E). Furthermore, rA1M significantly reduced spontaneous and heme-induced fetal RBC cell death. Addition of A1M to human whole blood resulted in a significant reduction of hemolysis, whereas removal of A1M from whole blood resulted in increased hemolysis. We conclude that A1M has a protective function in reducing hemolysis which is neither specific to the origin of hemolytic insult, nor species specific.


Asunto(s)
Anemia Macrocítica , Hemólisis , alfa-Globulinas , Animales , Muerte Celular , Eritrocitos , Humanos , Ratones , Ratones Noqueados , Fenotipo
4.
ACS Sens ; 4(3): 717-725, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30758943

RESUMEN

In this study, a surface plasmon resonance (SPR) biosensor was developed for the detection and quantification of a secreted bacterial factor (RoxP) from skin. A molecular imprinting method was used for the preparation of sensor chips and five different monomer-cross-linker compositions were evaluated for sensitivity, selectivity, affinity, and kinetic measurements. The most promising molecularly imprinted polymer (MIP) was characterized by using scanning electron microscopy, atomic force microscopy, and cyclic voltammetry. Limit of detection (LOD) value was calculated as 0.23 nM with an affinity constant of 3.3 × 10-9 M for the promising MIP. Besides being highly sensitive, the developed system was also very selective for the template protein RoxP, proven by the calculated selectivity coefficients. Finally, absolute concentrations of RoxP in several skin swabs were analyzed by using the developed MIP-SPR biosensor and compared to a competitive ELISA. Consequently, the developed system offers a very efficient tool for the detection and quantification of RoxP as an early indicator for some oxidative skin diseases especially when they are present in low-abundance levels (e.g., skin samples).


Asunto(s)
Impresión Molecular , Polímeros/síntesis química , Piel/metabolismo , Resonancia por Plasmón de Superficie/métodos , Factores de Virulencia/metabolismo , Animales , Humanos , Límite de Detección
5.
Antioxid Redox Signal ; 30(4): 489-504, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29471681

RESUMEN

AIMS: Human α1-microglobulin (A1M) is an endogenous reductase and radical- and heme-binding protein with physiological antioxidant protective functions. Recombinant human A1M (rA1M) has been shown to have therapeutic properties in animal models of preeclampsia, a pregnancy disease associated with oxidative stress. Recombinant A1M, however, lacks glycosylation, and shows lower solubility and stability than A1M purified from human plasma. The aims of this work were to (i) use site-directed mutagenesis to improve the physicochemical properties of rA1M, (ii) demonstrate that the physicochemically improved rA1M displays full in vitro cell protective effects as recombinant wild-type A1M (rA1M-wt), and (iii) show its therapeutic potential in vivo against acute kidney injury (AKI), another disease associated with oxidative stress. RESULTS: A novel recombinant A1M-variant (rA1M-035) with three amino acid substitutions was constructed, successfully expressed, and purified. rA1M-035 had improved solubility and stability compared with rA1M-wt, and showed intact in vitro heme-binding, reductase, antioxidation, and cell protective activities. Both rA1M-035 and rA1M-wt showed, for the first time, potential in vivo protective effects on kidneys using a mouse rhabdomyolysis glycerol injection model of AKI. INNOVATION: A novel recombinant A1M-variant, rA1M-035, was engineered. This protein showed improved solubility and stability compared with rA1M-wt, full in vitro functional activity, and potential protection against AKI in an in vivo rhabdomyolysis mouse model. CONCLUSION: The new rA1M-035 is a better drug candidate than rA1M-wt for treatment of AKI and preeclampsia in human patients.


Asunto(s)
Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/terapia , alfa-Globulinas/metabolismo , Rabdomiólisis/metabolismo , Lesión Renal Aguda/metabolismo , alfa-Globulinas/genética , Animales , Femenino , Humanos , Células K562 , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/metabolismo
6.
Sci Rep ; 6: 36412, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27805044

RESUMEN

The role of the skin microbiota in human health is poorly understood. Here, we identified and characterized a novel antioxidant enzyme produced by the skin microbiota, designated RoxP for radical oxygenase of Propionibacterium acnes. RoxP is uniquely produced by the predominant skin bacterium P. acnes, with no homologs in other bacteria; it is highly expressed and strongly secreted into culture supernatants. We show that RoxP binds heme, reduces free radicals, and can protect molecules from oxidation. Strikingly, RoxP is crucial for the survival of P. acnes in oxic conditions and for skin colonization of P. acnes ex vivo. Taken together, our study strongly suggests that RoxP facilitates P. acnes' survival on human skin, and is an important beneficial factor for the host-commensal interaction. Thus, RoxP is the first described skin microbiota-derived mutualistic factor that potentially can be exploited for human skin protection.


Asunto(s)
Antioxidantes/metabolismo , Proteínas Bacterianas/metabolismo , Oxigenasas/metabolismo , Propionibacterium acnes/aislamiento & purificación , Piel/microbiología , Antioxidantes/química , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Hemo/metabolismo , Humanos , Microbiota , Mutagénesis , Oxidación-Reducción , Oxigenasas/clasificación , Oxigenasas/genética , Filogenia , Propionibacterium acnes/genética , Unión Proteica , ARN Bacteriano/química , ARN Bacteriano/aislamiento & purificación , ARN Bacteriano/metabolismo , Análisis de Secuencia de ARN
7.
Future Microbiol ; 11: 721-36, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27200457

RESUMEN

AIM: The aim of this study was to identify and characterize EndoS-like enzymes in Streptococcus dysgalactiae subspecies dysgalactiae (SDSD). MATERIALS & METHODS: PCR, DNA sequencing, recombinant protein expression, lectin blot, ultra high performance liquid chromatography analysis and a chitinase assay were used to identify ndoS-like genes and characterize EndoSd. RESULTS: EndoSd were found in four SDSD strains. EndoSd hydrolyzes the chitobiose core of the glycan on IgG. The amino acid sequence of EndoSd is 70% identical to EndoS in S. pyogenes, but it has a unique C-terminal sequence. EndoSd secretion is influenced by the carbohydrate composition of the growth medium. CONCLUSION: Our findings indicate that IgG glycan hydrolyzing activity is present in SDSD, and that the activity can be attributed to the here identified enzyme EndoSd.


Asunto(s)
Acetilglucosaminidasa/metabolismo , Proteínas Bacterianas/metabolismo , Inmunoglobulina G/metabolismo , Polisacáridos/metabolismo , Streptococcus/enzimología , Acetilglucosaminidasa/química , Acetilglucosaminidasa/genética , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Disacáridos/metabolismo , Humanos , Hidrólisis , Inmunoglobulina G/química , Filogenia , Polisacáridos/química , Streptococcus/química , Streptococcus/clasificación , Streptococcus/genética , Especificidad por Sustrato
8.
Glycobiology ; 25(10): 1053-63, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26156869

RESUMEN

Enzymes that affect glycoproteins of the human immune system, and thereby modulate defense responses, are abundant among bacterial pathogens. Two endoglycosidases from the human pathogen Streptococcus pyogenes, EndoS and EndoS2, have recently been shown to hydrolyze N-linked glycans of human immunoglobulin G. However, detailed characterization and comparison of the hydrolyzing activities have not been performed. In the present study, we set out to characterize the enzymes by comparing the activities of EndoS and EndoS2 on a selection of therapeutic monoclonal antibodies (mAbs), cetuximab, adalimumab, panitumumab and denosumab. By analyzing the glycans hydrolyzed by EndoS and EndoS2 from the antibodies using matrix-assisted laser desorption ionization time of flight, we found that both the enzymes cleaved complex glycans and that EndoS2 hydrolyzed hybrid and oligomannose structures to a greater extent compared with EndoS. A comparison of ultra-high-performance liquid chromatography (LC) profiles of the glycan pool of cetuximab hydrolyzed with EndoS and EndoS2 showed that EndoS2 hydrolyzed hybrid and oligomannose glycans, whereas these peaks were missing in the EndoS chromatogram. We utilized this difference in glycoform selectivity, in combination with the IdeS protease, and developed a LC separation method to quantify high mannose content in the Fc fragments of the selected mAbs. We conclude that EndoS and EndoS2 hydrolyze different glycoforms from the Fc-glycosylation site on therapeutic mAbs and that this can be used for rapid quantification of high mannose content.


Asunto(s)
Proteínas Bacterianas/química , Glicósido Hidrolasas/química , Fragmentos Fc de Inmunoglobulinas/química , Mananos/análisis , Adalimumab/química , Anticuerpos Monoclonales/química , Cetuximab/química , Denosumab/química , Hidrólisis , Mananos/química , Panitumumab , Polisacáridos/química , Especificidad por Sustrato
9.
Biochem J ; 455(1): 107-18, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23865566

RESUMEN

Many bacteria have evolved ways to interact with glycosylation functions of the immune system of their hosts. Streptococcus pyogenes [GAS (group A Streptococcus)] secretes the enzyme EndoS that cleaves glycans on human IgG and impairs the effector functions of the antibody. The ndoS gene, encoding EndoS, has, until now, been thought to be conserved throughout the serotypes. However, in the present study, we identify EndoS2, an endoglycosidase in serotype M49 GAS strains. We characterized EndoS2 and the corresponding ndoS2 gene using sequencing, bioinformatics, phylogenetic analysis, recombinant expression and LC-MS analysis of glycosidic activity. This revealed that EndoS2 is present exclusively, and highly conserved, in serotype M49 of GAS and is only 37% identical with EndoS. EndoS2 showed endo-ß-N-acetylglucosaminidase activity on all N-linked glycans of IgG and on biantennary and sialylated glycans of AGP (α1-acid glycoprotein). The enzyme was found to act only on native IgG and AGP and to be specific for free biantennary glycans with or without terminal sialylation. GAS M49 expression of EndoS2 was monitored in relation to carbohydrates present in the culture medium and was linked to the presence of sucrose. We conclude that EndoS2 is a unique endoglycosidase in serotype M49 and differs from EndoS of other GAS strains by targeting both IgG and AGP. EndoS2 expands the repertoire of GAS effectors that modify key glycosylated molecules of host defence.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glicósido Hidrolasas/metabolismo , Inmunoglobulina G/metabolismo , Orosomucoide/metabolismo , Streptococcus pyogenes/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Secuencia de Carbohidratos , Secuencia Conservada , Escherichia coli/genética , Escherichia coli/metabolismo , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Interacciones Huésped-Patógeno , Humanos , Inmunoglobulina G/química , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Datos de Secuencia Molecular , Orosomucoide/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Streptococcus pyogenes/química , Streptococcus pyogenes/genética , Especificidad por Sustrato , Sacarosa/metabolismo
10.
J Virol ; 86(22): 12039-52, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22933282

RESUMEN

The resistance of human immunodeficiency virus type 1 (HIV-1) to antibody-mediated immunity often prevents the detection of antibodies that neutralize primary isolates of HIV-1. However, conventional assays for antibody functions other than neutralization are suboptimal. Current methods for measuring the killing of virus-infected cells by antibody-dependent cell-mediated cytotoxicity (ADCC) are limited by the number of natural killer (NK) cells obtainable from individual donors, donor-to-donor variation, and the use of nonphysiological targets. We therefore developed an ADCC assay based on NK cell lines that express human or macaque CD16 and a CD4(+) T-cell line that expresses luciferase from a Tat-inducible promoter upon HIV-1 or simian immunodeficiency virus (SIV) infection. NK cells and virus-infected targets are mixed in the presence of serial plasma dilutions, and ADCC is measured as the dose-dependent loss of luciferase activity. Using this approach, ADCC titers were measured in plasma samples from HIV-infected human donors and SIV-infected macaques. For the same plasma samples paired with the same test viruses, this assay was approximately 2 orders of magnitude more sensitive than optimized assays for neutralizing antibodies-frequently allowing the measurement of ADCC in the absence of detectable neutralization. Although ADCC correlated with other measures of Env-specific antibodies, neutralizing and gp120 binding titers did not consistently predict ADCC activity. Hence, this assay affords a sensitive method for measuring antibodies capable of directing ADCC against HIV- or SIV-infected cells expressing native conformations of the viral envelope glycoprotein and reveals incomplete overlap of the antibodies that direct ADCC and those measured in neutralization and binding assays.


Asunto(s)
Anticuerpos Antivirales/química , VIH-1/metabolismo , Pruebas de Neutralización/métodos , Virus de la Inmunodeficiencia de los Simios/metabolismo , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Linfocitos T CD4-Positivos/virología , Línea Celular , Ensayo de Inmunoadsorción Enzimática/métodos , Proteína gp120 de Envoltorio del VIH/metabolismo , Humanos , Inmunoglobulina G/química , Células Asesinas Naturales/virología , Macaca , Unión Proteica , Receptores de IgG/biosíntesis , Reproducibilidad de los Resultados , Virología/métodos
11.
Arthritis Rheum ; 64(8): 2698-706, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22392566

RESUMEN

OBJECTIVE: Systemic lupus erythematosus (SLE) is an autoimmune disease with chronic or episodic inflammation in several organ systems, related to the presence of circulating and tissue-deposited immune complexes (ICs) that stimulate leukocytes through Fcγ receptors (FcγR) with subsequent inflammation. Treatment with endoglycosidase S (EndoS), an IgG glycan-hydrolyzing bacterial enzyme from Streptococcus pyogenes, has shown beneficial effects in several experimental animal models of chronic inflammatory disease. This study was undertaken to investigate whether EndoS affects the proinflammatory properties of ICs and has the potential to be developed as a therapy for SLE. METHODS: ICs purified from SLE patients or RNA-containing ICs formed in vitro were treated with EndoS and used in several assays reflecting different important features of SLE pathogenesis, such as phagocytosis by polymorphonuclear cells (PMNs) and plasmacytoid dendritic cells (PDCs), complement activation, and interferon-α (IFNα) production by PDCs. RESULTS: EndoS treatment abolished all proinflammatory properties of the ICs investigated. This included FcγR-mediated phagocytosis by PDCs (P = 0.001) and subsequent production of IFNα (P = 0.002), IC-induced classical pathway of complement activation (P = 0.008), chemotaxis, and oxidative burst activity of PMNs (P = 0.002). EndoS treatment also had a direct effect on the molecular structure of ICs, causing decreased IC size and glycosylation. CONCLUSION: Our findings indicate that EndoS treatment has prominent effects on several pathogenetically important IC-mediated events, and suggest that EndoS has the potential to be developed as a novel therapy for SLE.


Asunto(s)
Complejo Antígeno-Anticuerpo/efectos de los fármacos , Proteínas Bacterianas/farmacología , Glicósido Hidrolasas/farmacología , Inmunoglobulina G/metabolismo , Inflamación/inmunología , Lupus Eritematoso Sistémico/inmunología , Polisacáridos/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Complejo Antígeno-Anticuerpo/metabolismo , Quimiotaxis/fisiología , Células Dendríticas/metabolismo , Femenino , Humanos , Hidrólisis/efectos de los fármacos , Inflamación/metabolismo , Inflamación/patología , Interferón-alfa/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Masculino , Persona de Mediana Edad , Neutrófilos/fisiología , Fagocitosis/fisiología , Receptores de IgG/fisiología , Adulto Joven
12.
Antioxid Redox Signal ; 17(5): 813-46, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22324321

RESUMEN

Hemoglobin (Hb) is the major oxygen (O(2))-carrying system of the blood but has many potentially dangerous side effects due to oxidation and reduction reactions of the heme-bound iron and O(2). Extracellular Hb, resulting from hemolysis or exogenous infusion, is shown to be an important pathogenic factor in a growing number of diseases. This review briefly outlines the oxidative/reductive toxic reactions of Hb and its metabolites. It also describes physiological protection mechanisms that have evolved against extracellular Hb, with a focus on the most recently discovered: the heme- and radical-binding protein α(1)-microglobulin (A1M). This protein is found in all vertebrates, including man, and operates by rapidly clearing cytosols and extravascular fluids of heme groups and free radicals released from Hb. Five groups of pathological conditions with high concentrations of extracellular Hb are described: hemolytic anemias and transfusion reactions, the pregnancy complication pre-eclampsia, cerebral intraventricular hemorrhage of premature infants, chronic inflammatory leg ulcers, and infusion of Hb-based O(2) carriers as blood substitutes. Finally, possible treatments of these conditions are discussed, giving a special attention to the described protective effects of A1M.


Asunto(s)
alfa-Globulinas/metabolismo , Hemoglobinas/metabolismo , Humanos
13.
J Neurochem ; 120(2): 279-91, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22043816

RESUMEN

Paraquat is a widely used herbicide that is structurally similar to the known dopaminergic neurotoxicant 1-methyl-4-phenyl-pyridine and acts as a potential etiologic factor for the development of Parkinson's disease. In this study, we investigated the protective roles of lipocalin-type prostaglandin (PG) D synthase (L-PGDS) against paraquat-mediated apoptosis of human neuronal SH-SY5Y cells. The treatment of SH-SY5Y cells with paraquat decreased the intracellular GSH level, and enhanced the cell death with elevation of the caspase activities. L-PGDS was expressed in SH-SY5Y cells, and its expression was enhanced with the peak at 2 h after the initiation of the treatment with paraquat. Inhibition of PGD2 synthesis and exogenously added PGs showed no effects regarding the paraquat-mediated apoptosis. SiRNA-mediated suppression of L-PGDS expression in the paraquat-treated cells increased the cell death and caspase activities. Moreover, over-expression of L-PGDS suppressed the cell death and caspase activities in the paraquat-treated cells. The results of a promoter-luciferase assay demonstrated that paraquat-mediated elevation of L-PGDS gene expression occurred through the NF-κB element in the proximal promoter region of the L-PGDS gene in SH-SY5Y cells. These results indicate that L-PGDS protected against the apoptosis in the paraquat-treated SH-SY5Y cells through the up-regulation of L-PGDS expression via the NF-κB element. Thus, L-PGDS might potentially serve as an agent for prevention of human neurodegenerative diseases caused by oxidative stress and apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Paraquat/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , FN-kappa B/metabolismo , Neuroblastoma , Prostaglandina D2/metabolismo , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Estadísticas no Paramétricas
14.
PLoS One ; 6(11): e27505, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22096585

RESUMEN

During bleeding the skin is subjected to oxidative insults from free heme and radicals, generated from extracellular hemoglobin. The lipocalin α(1)-microglobulin (A1M) was recently shown to have reductase properties, reducing heme-proteins and other substrates, and to scavenge heme and radicals. We investigated the expression and localization of A1M in skin and the possible role of A1M in the protection of skin tissue from damage induced by heme and reactive oxygen species. Skin explants, keratinocyte cultures and purified collagen I were exposed to heme, reactive oxygen species, and/or A1M and investigated by biochemical methods and electron microscopy. The results demonstrate that A1M is localized ubiquitously in the dermal and epidermal layers, and that the A1M-gene is expressed in keratinocytes and up-regulated after exposure to heme and reactive oxygen species. A1M inhibited the heme- and reactive oxygen species-induced ultrastructural damage, up-regulation of antioxidation and cell cycle regulatory genes, and protein carbonyl formation in skin and keratinocytes. Finally, A1M bound to purified collagen I (K(d) = 0.96×10(-6) M) and could inhibit and repair the destruction of collagen fibrils by heme and reactive oxygen species. The results suggest that A1M may have a physiological role in protection of skin cells and matrix against oxidative damage following bleeding.


Asunto(s)
alfa-Globulinas/metabolismo , Hemo/farmacología , Especies Reactivas de Oxígeno/farmacología , Piel/metabolismo , alfa-Globulinas/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Humanos , Inmunohistoquímica , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/ultraestructura , Microscopía Electrónica de Transmisión , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Radioinmunoensayo , Reacción en Cadena en Tiempo Real de la Polimerasa , Piel/ultraestructura
15.
Blood ; 116(11): 1951-7, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20538795

RESUMEN

Patients with systemic lupus erythematosus (SLE) have a markedly increased risk to develop cardiovascular disease, and traditional cardiovascular risk factors fail to account for this increased risk. We used microarray to probe the platelet transcriptome in patients with SLE and healthy controls, and the gene and protein expression of a subset of differentially expressed genes was further investigated and correlated to platelet activation status. Real-time PCR was used to confirm a type I interferon (IFN) gene signature in patients with SLE, and the IFN-regulated proteins PRKRA, IFITM1 and CD69 (P < .0001) were found to be up-regulated in platelets from SLE patients compared with healthy volunteers. Notably, patients with a history of vascular disease had increased expression of type I IFN-regulated proteins as well as more activated platelets compared with patients without vascular disease. We suggest that interferogenic immune complexes stimulate production of IFNα that up-regulates the megakaryocytic type I IFN-regulated genes and proteins. This could affect platelet activation and contribute to development of vascular disease in SLE. In addition, platelets with type I IFN signature could be a novel marker for vascular disease in SLE.


Asunto(s)
Plaquetas/metabolismo , Perfilación de la Expresión Génica/métodos , Lupus Eritematoso Sistémico/sangre , Proteómica/métodos , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/metabolismo , Western Blotting , Estudios de Cohortes , Femenino , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba , Enfermedades Vasculares/sangre , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Adulto Joven
16.
Blood ; 115(24): 5080-8, 2010 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-20357243

RESUMEN

EndoS from Streptococcus pyogenes is an immunomodulating enzyme that specifically hydrolyzes glycans from human immunoglobulin G and thereby affects antibody effector functions. Autoimmune hemolytic anemia is caused by antibody-mediated red blood cell (RBC) destruction and often resists treatment with corticosteroids that also cause frequent adverse effects. We show here that anti-RhD (anti-D) and rabbit anti-human-RBC antibodies (anti-RBC) mediated destruction of RBC, ie, phagocytosis, complement activation, and hemolysis in vitro and in vivo was inhibited by EndoS. Phagocytosis by monocytes in vitro was inhibited by pretreatment of anti-D with EndoS before sensitization of RBCs and abrogated by direct addition of EndoS to blood containing sensitized RBCs. The toxic effects of monocytes stimulated with anti-D-sensitized RBCs, as measured by interleukin-8 secretion and oxygen metabolite production, was restrained by EndoS. Agglutination of RBCs and complement-mediated hemolysis in vitro in whole human blood caused by rabbit anti-RBCs was inhibited by EndoS. Development of anemia in mice caused by a murine anti-RBC immunoglobulin G2a monoclonal autoantibody and complement activation and erythrophagocytosis by Kupffer cells in the liver were reduced by EndoS. Our data indicate that EndoS is a potential therapeutic agent that might be evaluated as an alternative to current treatment regimens against antibody-mediated destruction of RBCs.


Asunto(s)
Anemia Hemolítica Autoinmune , Proteínas Bacterianas/farmacología , Glicósido Hidrolasas/farmacología , Hemólisis/efectos de los fármacos , Hemólisis/inmunología , Inmunoglobulina G/metabolismo , Anemia Hemolítica Autoinmune/tratamiento farmacológico , Anemia Hemolítica Autoinmune/inmunología , Anemia Hemolítica Autoinmune/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Complemento C1q/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/inmunología , Eritrocitos/metabolismo , Glicósido Hidrolasas/metabolismo , Humanos , Inmunoglobulina G/inmunología , Interleucina-8/metabolismo , Isoanticuerpos/inmunología , Isoanticuerpos/metabolismo , Ratones , Ratones Endogámicos BALB C , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Oxígeno/metabolismo , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Conejos , Globulina Inmune rho(D)
17.
Ann N Y Acad Sci ; 1173: 664-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19758213

RESUMEN

The enzyme EndoS from Streptococcus pyogenes is an immunomodulatory molecule hydrolyzing the conserved glycans in the effector part of immunoglobulin G (IgG). EndoS is remarkably specific for IgG, and hydrolysis has profound effects on IgG effector functions. EndoS pretreatment of IgG, or direct administration to animals with experimental antibody-mediated autoimmune diseases, inhibits development of disease or cures animals from established disease. The properties of EndoS make it a unique experimental tool and an attractive alternative to current therapies of conditions involving pathogenic antibodies. This review describes the discovery of EndoS, the effects of EndoS on IgG effector functions in vitro and in vivo, the biotechnological potential of EndoS, and the outcomes of EndoS treatment in animal models of autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Proteínas Bacterianas/metabolismo , Glicósido Hidrolasas/metabolismo , Inmunoglobulina G/metabolismo , Polisacáridos/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Autoinmunidad/efectos de los fármacos , Proteínas Bacterianas/administración & dosificación , Modelos Animales de Enfermedad , Glicósido Hidrolasas/administración & dosificación , Humanos , Inmunoglobulina G/química , Conformación Proteica , Especificidad por Sustrato
18.
Aging Cell ; 7(4): 506-15, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18419796

RESUMEN

Many nervous system pathologies are associated with increased levels of apolipoprotein D (ApoD), a lipocalin also expressed during normal development and aging. An ApoD homologous gene in Drosophila, Glial Lazarillo, regulates resistance to stress, and neurodegeneration in the aging brain. Here we study for the first time the protective potential of ApoD in a vertebrate model organism. Loss of mouse ApoD function increases the sensitivity to oxidative stress and the levels of brain lipid peroxidation, and impairs locomotor and learning abilities. Human ApoD overexpression in the mouse brain produces opposite effects, increasing survival and preventing the raise of brain lipid peroxides after oxidant treatment. These observations, together with its transcriptional up-regulation in the brain upon oxidative insult, identify ApoD as an acute response protein with a protective and therefore beneficial function mediated by the control of peroxidated lipids.


Asunto(s)
Apolipoproteínas D/metabolismo , Estrés Oxidativo , Envejecimiento/efectos de los fármacos , Animales , Apolipoproteínas D/genética , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Aprendizaje/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Lipocalinas/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/patología , Estrés Oxidativo/efectos de los fármacos , Paraquat/farmacología , Análisis de Supervivencia , Transgenes , Regulación hacia Arriba/efectos de los fármacos
19.
BMC Microbiol ; 8: 3, 2008 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-18182097

RESUMEN

BACKGROUND: The endoglycosidase EndoS and the cysteine proteinase SpeB from the human pathogen Streptococcus pyogenes are functionally related in that they both hydrolyze IgG leading to impairment of opsonizing antibodies and thus enhance bacterial survival in human blood. In this study, we further investigated the relationship between EndoS and SpeB by examining their in vitro temporal production and stability and activity of EndoS. Furthermore, theoretical structure modeling of EndoS combined with site-directed mutagenesis and chemical blocking of amino acids was used to identify amino acids required for the IgG glycan-hydrolyzing activity of EndoS. RESULTS: We could show that during growth in vitro S. pyogenes secretes the IgG glycan-hydrolyzing endoglycosidase EndoS prior to the cysteine proteinase SpeB. Upon maturation SpeB hydrolyzes EndoS that then loses its IgG glycan-hydrolyzing activity. Sequence analysis and structural homology modeling of EndoS provided a basis for further analysis of the prerequisites for IgG glycan-hydrolysis. Site-directed mutagenesis and chemical modification of amino acids revealed that glutamic acid 235 is an essential catalytic residue, and that tryptophan residues, but not the abundant lysine or the single cysteine residues, are important for EndoS activity. CONCLUSION: We present novel information about the amino acid requirements for IgG glycan-hydrolyzing activity of the immunomodulating enzyme EndoS. Furthermore, we show that the cysteine proteinase SpeB processes/degrades EndoS and thus emphasize the importance of the SpeB as a degrading/processing enzyme of proteins from the bacterium itself.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Ácido Glutámico/metabolismo , Glicósido Hidrolasas/metabolismo , Inmunoglobulina G/metabolismo , Polisacáridos/metabolismo , Triptófano/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Western Blotting , Cisteína Endopeptidasas/genética , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Hidrólisis , Inmunoglobulina G/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Polisacáridos/química , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido , Streptococcus pyogenes/enzimología , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Homología Estructural de Proteína , Relación Estructura-Actividad
20.
PLoS One ; 3(1): e1413, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18183294

RESUMEN

BACKGROUND: The human pathogen Streptococcus pyogenes produces an endoglycosidase, EndoS that hydrolyzes the chitobiose core of the asparagine-linked glycan on the heavy chain of human IgG. IgG-binding to Fc gamma receptors (Fc gamma R) on leukocytes triggers effector functions including phagocytosis, oxidative burst and the release of inflammatory mediators. The interactions between Fc gamma R and the Fc domain of IgG depend on the IgG glycosylation state. METHODOLOGY/PRINCIPAL FINDINGS: Here we show for the first time that EndoS hydrolyzes the heavy chain glycan of all four human IgG subclasses (IgG1-4), in purified form and in a plasma environment. An inactive form of EndoS, obtained by site-directed mutagenesis, binds IgG with high affinity, in contrast to wild type EndoS that only transiently interacts with IgG, as shown by Slot-blotting and surface plasmon resonance technology. Furthermore, EndoS hydrolysis of the IgG glycan influences the binding of IgG to immobilized soluble Fc gamma R and to an erythroleukemic cell line, K562, expressing Fc gamma RIIa. Incubation of whole blood with EndoS results in a dramatic decrease of IgG binding to activated monocytes as analyzed by flow cytometry. Moreover, the IgG bound to K562 cells dissociates when cells are treated with EndoS. Likewise, IgG bound to immobilized Fc gamma RIIa and subsequently treated with EndoS, dissociates from the receptor as analyzed by surface plasmon resonance and Western blot. CONCLUSIONS/SIGNIFICANCE: We provide novel information about bacterial enzymatic modulation of the IgG/Fc gamma R interaction that emphasizes the importance of glycosylation for antibody effector functions. Moreover, EndoS could be used as a biochemical tool for specific IgG N-glycan hydrolysis and IgG purification/detection, or as a potential immunosuppressing agent for treatment of antibody-mediated pathological processes.


Asunto(s)
Inmunoglobulina G/metabolismo , Polisacáridos/metabolismo , Receptores de IgG/metabolismo , Streptococcus/metabolismo , Western Blotting , Línea Celular , Humanos , Hidrólisis , Mutagénesis Sitio-Dirigida , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA