Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 17, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38178089

RESUMEN

BACKGROUND: Hemolysis is a cardinal feature of hemolytic uremic syndrome (HUS) and during hemolysis excess arginase 1 is released from red blood cells. Increased arginase activity leads to reduced L-arginine, as it is converted to urea and L-ornithine, and thereby reduced nitric oxide bioavailability, with secondary vascular injury. The objective of this study was to investigate arginase release in HUS patients and laboratory models and correlate arginase levels to hemolysis and kidney injury. METHODS: Two separate cohorts of patients (n = 47 in total) with HUS associated with Shiga toxin-producing enterohemorrhagic E. coli (EHEC) and pediatric controls (n = 35) were investigated. Two mouse models were used, in which mice were either challenged intragastrically with E. coli O157:H7 or injected intraperitoneally with Shiga toxin 2. An in vitro model of thrombotic microangiopathy was developed in which Shiga toxin 2- and E. coli O157 lipopolysaccharide-stimulated human blood cells combined with ADAMTS13-deficient plasma were perfused over glomerular endothelial cells. Two group statistical comparisons were performed using the Mann-Whitney test, multiple groups were compared using the Kruskal-Wallis test followed by Dunn's procedure, the Wilcoxon signed rank test was used for paired data, or linear regression for continuous variables. RESULTS: HUS patients had excessively high plasma arginase 1 levels and activity (conversion of L-arginine to urea and L-ornithine) during the acute phase, compared to remission and controls. Arginase 1 levels correlated with lactate dehydrogenase activity, indicating hemolysis, as well as the need for dialysis treatment. Patients also exhibited high levels of plasma alpha-1-microglobulin, a heme scavenger. Both mouse models exhibited significantly elevated plasma arginase 1 levels and activity. Plasma arginase 1 levels correlated with lactate dehydrogenase activity, alpha-1-microglobulin and urea levels, the latter indicative of kidney dysfunction. In the in vitro model of thrombotic microangiopathy, bioactive arginase 1 was released and levels correlated to the degree of hemolysis. CONCLUSIONS: Elevated red blood cell-derived arginase was demonstrated in HUS patients and in relevant in vivo and in vitro models. The excessively high arginase levels correlated to the degree of hemolysis and kidney dysfunction. Thus, arginase inhibition should be investigated in HUS.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli O157 , Síndrome Hemolítico-Urémico , Insuficiencia Renal , Microangiopatías Trombóticas , Humanos , Niño , Animales , Ratones , Toxina Shiga II , Células Endoteliales , Hemólisis , Arginasa , Síndrome Hemolítico-Urémico/complicaciones , Síndrome Hemolítico-Urémico/terapia , Eritrocitos , Microangiopatías Trombóticas/complicaciones , Urea , Arginina , Ornitina , Lactato Deshidrogenasas , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/terapia
2.
Pediatr Nephrol ; 39(4): 1105-1111, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37955705

RESUMEN

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) can be associated with mutations, deletions, or hybrid genes in factor H-related (FHR) proteins. METHODS: A child with aHUS was investigated. Genetics was assessed by Sanger and next generation sequencing. Serum FHR5 was evaluated by immunoblotting, ELISA, and by induction of rabbit red blood cell hemolysis in the presence/absence of recombinant human rFHR5. Mutagenesis was performed in HEK cells. RESULTS: A heterozygous genetic variant in factor H-related protein 5 (CFHR5), M514R, was found in the child, who also had a homozygous deletion of CFHR3/CFHR1, and antibodies to factor H, as well as low levels of C3. Patient serum exhibited low levels of FHR5. In the presence of rabbit red blood cells, patient serum induced hemolysis which decreased when rFHR5 was added at physiological concentrations. Similar results were obtained using serum from the father, bearing the CFHR5 variant without factor H antibodies. Patient FHR5 formed normal dimers. The CFHR5 M514R variant was expressed in HEK cells and minimal secretion was detected whereas the protein level was elevated in cell lysates. CONCLUSIONS: Decreased secretion of the product of the mutant allele could explain the low FHR5 levels in patient serum. Reduced hemolysis when rFHR5 was added to serum suggests a regulatory role regarding complement activation on red blood cells. As such, low levels of FHR5, as demonstrated in the patient, may contribute to complement activation.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Niño , Animales , Humanos , Conejos , Síndrome Hemolítico Urémico Atípico/genética , Factor H de Complemento/genética , Hemólisis , Homocigoto , Eliminación de Secuencia , Proteínas del Sistema Complemento , Anticuerpos
3.
Kidney Int ; 94(4): 689-700, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29884545

RESUMEN

Certain kidney diseases are associated with complement activation although a renal triggering factor has not been identified. Here we demonstrated that renin, a kidney-specific enzyme, cleaves C3 into C3b and C3a, in a manner identical to the C3 convertase. Cleavage was specifically blocked by the renin inhibitor aliskiren. Renin-mediated C3 cleavage and its inhibition by aliskiren also occurred in serum. Generation of C3 cleavage products was demonstrated by immunoblotting, detecting the cleavage product C3b, by N-terminal sequencing of the cleavage product, and by ELISA for C3a release. Functional assays showed mast cell chemotaxis towards the cleavage product C3a and release of factor Ba when the cleavage product C3b was combined with factor B and factor D. The renin-mediated C3 cleavage product bound to factor B. In the presence of aliskiren this did not occur, and less C3 deposited on renin-producing cells. The effect of aliskiren was studied in three patients with dense deposit disease and this demonstrated decreased systemic and renal complement activation (increased C3, decreased C3a and C5a, decreased renal C3 and C5b-9 deposition and/or decreased glomerular basement membrane thickness) over a follow-up period of four to seven years. Thus, renin can trigger complement activation, an effect inhibited by aliskiren. Since renin concentrations are higher in renal tissue than systemically, this may explain the renal propensity of complement-mediated disease in the presence of complement mutations or auto-antibodies.


Asunto(s)
Amidas/farmacología , Activación de Complemento/efectos de los fármacos , Complemento C3/química , Fumaratos/farmacología , Glomerulonefritis Membranoproliferativa/metabolismo , Glomerulonefritis Membranoproliferativa/terapia , Renina/química , Amidas/uso terapéutico , Quimiotaxis/efectos de los fármacos , Niño , Complemento C3/metabolismo , Complemento C3a/química , Complemento C3a/metabolismo , Complemento C3b/química , Complemento C3b/metabolismo , Complemento C4/química , Complemento C5a/química , Complemento C5a/metabolismo , Complemento C5b/química , Complemento C5b/metabolismo , Factor B del Complemento/química , Factor D del Complemento/química , Femenino , Fumaratos/uso terapéutico , Membrana Basal Glomerular/patología , Glomerulonefritis Membranoproliferativa/patología , Humanos , Mastocitos/fisiología , Renina/antagonistas & inhibidores , Renina/metabolismo
4.
J Immunol ; 197(4): 1276-86, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27421478

RESUMEN

Complement activation occurs during enterohemorrhagic Escherichia coli (EHEC) infection and may exacerbate renal manifestations. In this study, we show glomerular C5b-9 deposits in the renal biopsy of a child with EHEC-associated hemolytic uremic syndrome. The role of the terminal complement complex, and its blockade as a therapeutic modality, was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice were treated with monoclonal anti-C5 i.p. on day 3 or 6 after intragastric inoculation and monitored for clinical signs of disease and weight loss for 14 d. All infected untreated mice (15 of 15) or those treated with an irrelevant Ab (8 of 8) developed severe illness. In contrast, only few infected mice treated with anti-C5 on day 3 developed symptoms (three of eight, p < 0.01 compared with mice treated with the irrelevant Ab on day 3) whereas most mice treated with anti-C5 on day 6 developed symptoms (six of eight). C6-deficient C57BL/6 mice were also inoculated with E. coli O157:H7 and only 1 of 14 developed disease, whereas 10 of 16 wild-type mice developed weight loss and severe disease (p < 0.01). Complement activation via the terminal pathway is thus involved in the development of disease in murine EHEC infection. Early blockade of the terminal complement pathway, before the development of symptoms, was largely protective, whereas late blockade was not. Likewise, lack of C6, and thereby deficient terminal complement complex, was protective in murine E. coli O157:H7 infection.


Asunto(s)
Complemento C6/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/inmunología , Animales , Preescolar , Complemento C6/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Escherichia coli Enterohemorrágica , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
5.
J Am Soc Nephrol ; 28(8): 2472-2481, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28289183

RESUMEN

The kinin system is activated during vasculitis and may contribute to chronic inflammation. C1-inhibitor is the main inhibitor of the kinin system. In this study, we investigated the presence of the kinin B1 receptor on endothelial microvesicles and its contribution to the inflammatory process. Compared with controls (n=15), patients with acute vasculitis (n=12) had markedly higher levels of circulating endothelial microvesicles, identified by flow cytometry analysis, and significantly more microvesicles that were positive for the kinin B1 receptor (P<0.001). Compared with microvesicles from wild-type cells, B1 receptor-positive microvesicles derived from transfected human embryonic kidney cells induced a significant neutrophil chemotactic effect, and a B1 receptor antagonist blocked this effect. Likewise, patient plasma induced neutrophil chemotaxis, an effect decreased by reduction of microvesicle levels and by blocking the B1 receptor. We used a perfusion system to study the effect of patient plasma (n=6) and control plasma (n=6) on the release of microvesicles from glomerular endothelial cells. Patient samples induced the release of significantly more B1 receptor-positive endothelial microvesicles than control samples, an effect abrogated by reduction of the microvesicles in the perfused samples. Perfusion of C1-inhibitor-depleted plasma over glomerular endothelial cells promoted excessive release of B1 receptor-positive endothelial microvesicles compared with normal plasma, an effect significantly decreased by addition of C1-inhibitor or B1 receptor-antagonist. Thus, B1 receptor-positive endothelial microvesicles may contribute to chronic inflammation by inducing neutrophil chemotaxis, and the reduction of these microvesicles by C1-inhibitor should be explored as a potential treatment for neutrophil-induced inflammation.


Asunto(s)
Micropartículas Derivadas de Células/fisiología , Proteínas Inactivadoras del Complemento 1/fisiología , Vasculitis/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Quimiotaxis , Niño , Proteína Inhibidora del Complemento C1 , Endotelio Vascular/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
6.
PLoS Pathog ; 11(2): e1004619, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25719452

RESUMEN

Shiga toxin (Stx) is the main virulence factor of enterohemorrhagic Escherichia coli, which are non-invasive strains that can lead to hemolytic uremic syndrome (HUS), associated with renal failure and death. Although bacteremia does not occur, bacterial virulence factors gain access to the circulation and are thereafter presumed to cause target organ damage. Stx was previously shown to circulate bound to blood cells but the mechanism by which it would potentially transfer to target organ cells has not been elucidated. Here we show that blood cell-derived microvesicles, shed during HUS, contain Stx and are found within patient renal cortical cells. The finding was reproduced in mice infected with Stx-producing Escherichia coli exhibiting Stx-containing blood cell-derived microvesicles in the circulation that reached the kidney where they were transferred into glomerular and peritubular capillary endothelial cells and further through their basement membranes followed by podocytes and tubular epithelial cells, respectively. In vitro studies demonstrated that blood cell-derived microvesicles containing Stx undergo endocytosis in glomerular endothelial cells leading to cell death secondary to inhibited protein synthesis. This study demonstrates a novel virulence mechanism whereby bacterial toxin is transferred within host blood cell-derived microvesicles in which it may evade the host immune system.


Asunto(s)
Toxinas Bacterianas/metabolismo , Células Sanguíneas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Escherichia coli Enterohemorrágica/metabolismo , Infecciones por Escherichia coli/metabolismo , Adolescente , Adulto , Animales , Células Sanguíneas/microbiología , Micropartículas Derivadas de Células/microbiología , Células Cultivadas , Niño , Preescolar , Infecciones por Escherichia coli/patología , Femenino , Interacciones Huésped-Patógeno , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas
7.
J Immunol ; 194(5): 2309-18, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25637016

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli (STEC) cause hemolytic uremic syndrome (HUS). This study investigated whether Stx2 induces hemolysis and whether complement is involved in the hemolytic process. RBCs and/or RBC-derived microvesicles from patients with STEC-HUS (n = 25) were investigated for the presence of C3 and C9 by flow cytometry. Patients exhibited increased C3 deposition on RBCs compared with controls (p < 0.001), as well as high levels of C3- and C9-bearing RBC-derived microvesicles during the acute phase, which decreased after recovery. Stx2 bound to P1 (k) and P2 (k) phenotype RBCs, expressing high levels of the P(k) Ag (globotriaosylceramide), the known Stx receptor. Stx2 induced the release of hemoglobin and lactate dehydrogenase in whole blood, indicating hemolysis. Stx2-induced hemolysis was not demonstrated in the absence of plasma and was inhibited by heat inactivation, as well as by the terminal complement pathway Ab eculizumab, the purinergic P2 receptor antagonist suramin, and EDTA. In the presence of whole blood or plasma/serum, Stx2 induced the release of RBC-derived microvesicles coated with C5b-9, a process that was inhibited by EDTA, in the absence of factor B, and by purinergic P2 receptor antagonists. Thus, complement-coated RBC-derived microvesicles are elevated in HUS patients and induced in vitro by incubation of RBCs with Stx2, which also induced hemolysis. The role of complement in Stx2-mediated hemolysis was demonstrated by its occurrence only in the presence of plasma and its abrogation by heat inactivation, EDTA, and eculizumab. Complement activation on RBCs could play a role in the hemolytic process occurring during STEC-HUS.


Asunto(s)
Vesículas Cubiertas/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Infecciones por Escherichia coli/sangre , Escherichia coli O157/patogenicidad , Síndrome Hemolítico-Urémico/sangre , Toxina Shiga/toxicidad , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/farmacología , Niño , Preescolar , Vesículas Cubiertas/química , Vesículas Cubiertas/inmunología , Activación de Complemento/efectos de los fármacos , Complemento C3/química , Complemento C9/química , Complejo de Ataque a Membrana del Sistema Complemento/química , Ácido Edético/farmacología , Eritrocitos/química , Eritrocitos/inmunología , Eritrocitos/patología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Escherichia coli O157/inmunología , Escherichia coli O157/metabolismo , Femenino , Expresión Génica , Hemólisis/efectos de los fármacos , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Humanos , Lactante , L-Lactato Deshidrogenasa/metabolismo , Masculino , Persona de Mediana Edad , Antagonistas del Receptor Purinérgico P2/farmacología , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/inmunología , Toxina Shiga/química , Toxina Shiga/inmunología , Suramina/farmacología , Trihexosilceramidas/inmunología
8.
J Immunol ; 193(1): 317-26, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24850720

RESUMEN

IgA nephropathy (IgAN) is characterized by mesangial cell proliferation and extracellular matrix expansion associated with immune deposits consisting of galactose-deficient polymeric IgA1 and C3. We have previously shown that IgA-binding regions of streptococcal M proteins colocalize with IgA in mesangial immune deposits in patients with IgAN. In the present study, the IgA-binding M4 protein from group A Streptococcus was found to bind to galactose-deficient polymeric IgA1 with higher affinity than to other forms of IgA1, as shown by surface plasmon resonance and solid-phase immunoassay. The M4 protein was demonstrated to bind to mesangial cells not via the IgA-binding region but rather via the C-terminal region, as demonstrated by flow cytometry. IgA1 enhanced binding of M4 to mesangial cells, but not vice versa. Costimulation of human mesangial cells with M4 and galactose-deficient polymeric IgA1 resulted in a significant increase in IL-6 secretion compared with each stimulant alone. Galactose-deficient polymeric IgA1 alone, but not M4, induced C3 secretion from the cells, and costimulation enhanced this effect. Additionally, costimulation enhanced mesangial cell proliferation compared with each stimulant alone. These results indicate that IgA-binding M4 protein binds preferentially to galactose-deficient polymeric IgA1 and that these proteins together induce excessive proinflammatory responses and proliferation of human mesangial cells. Thus, tissue deposition of streptococcal IgA-binding M proteins may contribute to the pathogenesis of IgAN.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Complemento C3/inmunología , Glomerulonefritis por IGA/inmunología , Inmunoglobulina A/inmunología , Interleucina-6/inmunología , Células Mesangiales/inmunología , Streptococcus/inmunología , Adolescente , Femenino , Glomerulonefritis por IGA/patología , Humanos , Masculino , Células Mesangiales/patología , Persona de Mediana Edad
9.
J Immunol ; 191(5): 2184-93, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23878316

RESUMEN

This study addressed the contribution of ADAMTS13 deficiency to complement activation in thrombotic thrombocytopenic purpura (TTP). Renal tissue and blood samples were available from 12 TTP patients. C3 and C5b-9 deposition were demonstrated in the renal cortex of two TTP patients, by immunofluorescence and immunohistochemistry, respectively. C3 was also demonstrated in the glomeruli of Shiga toxin-2-treated Adamts13(-/-) mice (n = 6 of 7), but less in mice that were not Shiga toxin-2 treated (n = 1 of 8, p < 0.05) or wild-type mice (n = 0 of 7). TTP patient plasma (n = 9) contained significantly higher levels of complement-coated endothelial microparticles than control plasma (n = 13), as detected by flow cytometry. Exposure of histamine-stimulated primary glomerular endothelial cells to platelet-rich plasma from patients, or patient platelet-poor plasma combined with normal platelets, in a perfusion system, under shear, induced C3 deposition on von Willebrand factor-platelet strings (on both von Willebrand factor and platelets) and on endothelial cells. Complement activation occurred via the alternative pathway. No C3 was detected when cells were exposed to TTP plasma that was preincubated with EDTA or heat-inactivated, or to control plasma. In the perfusion system, patient plasma induced more release of C3- and C9-coated endothelial microparticles compared with control plasma. The results indicate that the microvascular process induced by ADAMTS13 deficiency triggers complement activation on platelets and the endothelium, which may contribute to formation of thrombotic microangiopathy.


Asunto(s)
Proteínas ADAM/deficiencia , Activación de Complemento/inmunología , Microangiopatías Trombóticas/inmunología , Microangiopatías Trombóticas/metabolismo , Proteínas ADAM/inmunología , Proteína ADAMTS13 , Animales , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Metaloendopeptidasas/deficiencia , Metaloendopeptidasas/inmunología , Ratones , Ratones Noqueados
10.
Adv Exp Med Biol ; 865: 19-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26306441

RESUMEN

The complement system is activated in the vasculature during thrombotic and inflammatory conditions. Activation may be associated with chronic inflammation on the endothelial surface leading to complement deposition. Complement mutations allow uninhibited complement activation to occur on platelets, neutrophils, monocytes, and aggregates thereof, as well as on red blood cells and endothelial cells. Furthermore, complement activation on the cells leads to the shedding of cell derived-microvesicles that may express complement and tissue factor thus promoting inflammation and thrombosis. Complement deposition on red blood cells triggers hemolysis and the release of red blood cell-derived microvesicles that are prothrombotic. Microvesicles are small membrane vesicles ranging from 0.1 to 1 µm, shed by cells during activation, injury and/or apoptosis that express components of the parent cell. Microvesicles are released during inflammatory and vascular conditions. The repertoire of inflammatory markers on endothelial cell-derived microvesicles shed during inflammation is large and includes complement. These circulating microvesicles may reflect the ongoing inflammatory process but may also contribute to its propagation. This overview will describe complement activation on blood and endothelial cells and the release of microvesicles from these cells during hemolytic uremic syndrome, thrombotic thrombocytopenic purpura and vasculitis, clinical conditions associated with enhanced thrombosis and inflammation.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Síndrome Hemolítico-Urémico/metabolismo , Púrpura Trombocitopénica Trombótica/metabolismo , Trombosis/metabolismo , Vasculitis/metabolismo , Factores de Coagulación Sanguínea/inmunología , Factores de Coagulación Sanguínea/metabolismo , Plaquetas/inmunología , Plaquetas/metabolismo , Plaquetas/patología , Micropartículas Derivadas de Células/inmunología , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/patología , Proteínas del Sistema Complemento/inmunología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Eritrocitos/inmunología , Eritrocitos/metabolismo , Eritrocitos/patología , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/patología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos/patología , Púrpura Trombocitopénica Trombótica/inmunología , Púrpura Trombocitopénica Trombótica/patología , Trombosis/inmunología , Trombosis/patología , Vasculitis/inmunología , Vasculitis/patología
11.
J Immunol ; 188(4): 2030-7, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22250080

RESUMEN

Atypical hemolytic uremic syndrome has been associated with dysregulation of the alternative complement pathway. In this study, a novel heterozygous C3 mutation was identified in a factor B-binding region in exon 41, V1636A (4973 T > C). The mutation was found in three family members affected with late-onset atypical hemolytic uremic syndrome and symptoms of glomerulonephritis. All three patients exhibited increased complement activation detected by decreased C3 levels and glomerular C3 deposits. Platelets from two of the patients had C3 and C9 deposits on the cell surface. Patient sera exhibited more C3 cleavage and higher levels of C3a. The C3 mutation resulted in increased C3 binding to factor B and increased net formation of the C3 convertase, even after decay induced by decay-accelerating factor and factor H, as assayed by surface plasmon resonance. Patient sera incubated with washed human platelets induced more C3 and C9 deposition on the cell surface in comparison with normal sera. More C3a was released into serum over time when washed platelets were exposed to patient sera. Results regarding C3 and C9 deposition on washed platelets were confirmed using purified patient C3 in C3-depleted serum. The results indicated enhanced convertase formation leading to increased complement activation on cell surfaces. Previously described C3 mutations showed loss of function with regard to C3 binding to complement regulators. To our knowledge, this study presents the first known C3 mutation inducing increased formation of the C3 convertase, thus explaining enhanced activation of the alternative pathway of complement.


Asunto(s)
Convertasas de Complemento C3-C5/biosíntesis , Complemento C3a/genética , Complemento C3a/metabolismo , Complemento C3b/genética , Complemento C3b/metabolismo , Síndrome Hemolítico-Urémico/inmunología , Animales , Síndrome Hemolítico Urémico Atípico , Células COS , Chlorocebus aethiops , Activación de Complemento , Convertasas de Complemento C3-C5/genética , Convertasas de Complemento C3-C5/metabolismo , Complemento C3a/biosíntesis , Complemento C3b/biosíntesis , Complemento C9/biosíntesis , Complemento C9/metabolismo , Factor B del Complemento/metabolismo , Factor H de Complemento/metabolismo , Vía Alternativa del Complemento/genética , Glomerulonefritis/genética , Síndrome Hemolítico-Urémico/genética , Humanos , Masculino , Persona de Mediana Edad
12.
Front Immunol ; 15: 1476204, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39247189

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2023.1254759.].

13.
Clin Kidney J ; 17(8): sfae213, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39135939

RESUMEN

Background: Genetic variants in SEC61A1 are associated with autosomal dominant tubulointerstitial kidney disease. SEC61A1 is a translocon in the endoplasmic reticulum membrane and variants affect biosynthesis of renin and uromodulin. Methods: A patient is described that presented at 1 year of age with failure-to-thrive, kidney failure (glomerular filtration rate, GFR, 18 ml/min/1.73m2), hyperkalemia and acidosis. Genetic evaluation was performed by whole genome sequencing. Results: The patient has a novel de novo heterozygous SEC61A1 variant, Phe458Val. Plasma renin was low or normal, aldosterone was low or undetectable and uromodulin was low. Kidney biopsy at 2 years exhibited subtle changes suggestive of tubular dysgenesis without tubulocystic or glomerulocystic lesions and with renin staining of the juxtaglomerular cells. The patient experienced extreme fatigue due to severe hypotension attributed to hypoaldosteronism and at 8 years of age fludrocortisone treatment was initiated with marked improvement in her well-being. Blood pressure and potassium normalized. Biopsy at 9 years showed extensive glomerulosclerosis and mild tubulointerstitial fibrosis, as well as tubular mitochondrial abnormalities, without specific diagnostic changes. Her GFR improved to 54 ml/min/1.73m2. Conclusions: As the renin-angiotensin system promotes aldosterone release, and the patient had repeatedly undetectable aldosterone levels, the SEC61A1 variant presumably contributed to severe hypotension. Treatment with a mineralocorticoid had a beneficial effect and corrected the electrolyte and acid-base disorder. We suggest that the increased blood pressure hemodynamically improved the patient's kidney function.

14.
Nephrol Dial Transplant ; 28(11): 2899-907, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24009284

RESUMEN

BACKGROUND: Atypical haemolytic uraemic syndrome (aHUS) is associated with dysfunction of the alternative pathway of complement. Disease activity subsides as renal failure progresses but recurs upon renal transplantation, indicating that viable renal tissue contributes to disease activity. We present evidence of cerebrovascular occlusive disease indicating that vascular injury may occur in the absence of kidneys. METHODS: A currently 12-year-old girl developed renal failure at the age of 20 months. She underwent bilateral nephrectomy and renal transplantation but lost the transplant due to recurrences. She was on haemodialysis for 7 years. At 10 years of age she developed a transient ischaemic attack. Imaging, genetic investigation and mutation characterization were performed. RESULTS: Imaging demonstrated occlusion and stenosis of the carotid arteries. Two complement mutations, a novel mutation in factor B and a previously described mutation in factor I, and the H3-factor H haplotype, were identified. The factor B mutation, L433S, did not induce excessive complement activation in vitro. Measurement of C3 degradation products indicated ongoing complement activation. In spite of the patient being anephric, treatment was initiated with eculizumab, a humanized anti-C5 antibody that blocks terminal complement activation. She underwent a successful kidney transplant 9 months later and has not developed a recurrence or progression of vascular stenosis 1 year later. CONCLUSIONS: The course of disease in this patient with aHUS suggests that complement-mediated vascular injury may occur in the total absence of renal tissue and overt recurrences. To our knowledge, this is the first description of eculizumab treatment in an anephric aHUS patient.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Trastornos Cerebrovasculares/tratamiento farmacológico , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Trasplante de Riñón , Nefrectomía/efectos adversos , Insuficiencia Renal/complicaciones , Síndrome Hemolítico Urémico Atípico , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/patología , Niño , Complemento C5/metabolismo , Factor B del Complemento/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Síndrome Hemolítico-Urémico/etiología , Síndrome Hemolítico-Urémico/patología , Humanos , Imagen por Resonancia Magnética , Mutación/genética , Reacción en Cadena de la Polimerasa , Pronóstico , Insuficiencia Renal/cirugía , Resonancia por Plasmón de Superficie
15.
Front Immunol ; 14: 1254759, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37744338

RESUMEN

Background: Complement activation in atypical hemolytic uremic syndrome (aHUS), C3 glomerulonephropathy (C3G) and immune complex-mediated membranoproliferative glomerulonephritis (IC-MPGN) may be associated with rare genetic variants. Here we describe gene variants in the Swedish and Norwegian populations. Methods: Patients with these diagnoses (N=141) were referred for genetic screening. Sanger or next-generation sequencing were performed to identify genetic variants in 16 genes associated with these conditions. Nonsynonymous genetic variants are described when they have a minor allele frequency of <1% or were previously reported as being disease-associated. Results: In patients with aHUS (n=94, one also had IC-MPGN) 68 different genetic variants or deletions were identified in 60 patients, of which 18 were novel. Thirty-two patients had more than one genetic variant. In patients with C3G (n=40) 29 genetic variants, deletions or duplications were identified in 15 patients, of which 9 were novel. Eight patients had more than one variant. In patients with IC-MPGN (n=7) five genetic variants were identified in five patients. Factor H variants were the most frequent in aHUS and C3 variants in C3G. Seventeen variants occurred in more than one condition. Conclusion: Genetic screening of patients with aHUS, C3G and IC-MPGN is of paramount importance for diagnostics and treatment. In this study, we describe genetic assessment of Nordic patients in which 26 novel variants were found.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Enfermedades Renales , Humanos , Proteínas del Sistema Complemento/genética , Activación de Complemento/genética , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/genética , Frecuencia de los Genes
17.
Front Immunol ; 13: 807959, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250980

RESUMEN

Shiga toxin-producing Escherichia coli O157:H7 is a virulent strain causing severe gastrointestinal infection, hemolytic uremic syndrome and death. To date there are no specific therapies to reduce progression of disease. Here we investigated the effect of pooled immunoglobulins (IgG) on the course of disease in a mouse model of intragastric E. coli O157:H7 inoculation. Intraperitoneal administration of murine IgG on day 3, or both on day 3 and 6, post-inoculation improved survival and decreased intestinal and renal pathology. When given on both day 3 and 6 post-inoculation IgG treatment also improved kidney function in infected mice. Murine and human commercially available IgG preparations bound to proteins in culture filtrates from E. coli O157:H7. Bound proteins were extracted from membranes and peptide sequences were identified by mass spectrometry. The findings showed that murine and human IgG bound to E. coli extracellular serine protease P (EspP) in the culture filtrate, via the IgG Fc domain. These results were confirmed using purified recombinant EspP and comparing culture filtrates from the wild-type E. coli O157:H7 strain to a deletion mutant lacking espP. Culture filtrates from wild-type E. coli O157:H7 exhibited enzymatic activity, specifically associated with the presence of EspP and demonstrated as pepsin cleavage, which was reduced in the presence of murine and human IgG. EspP is a virulence factor previously shown to promote colonic cell injury and the uptake of Shiga toxin by intestinal cells. The results presented here suggest that IgG binds to EspP, blocks its enzymatic activity, and protects the host from E. coli O157:H7 infection, even when given post-inoculation.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli O157 , Proteínas de Escherichia coli , Inmunoglobulina G , Serina Proteasas , Animales , Proteínas de Escherichia coli/metabolismo , Inmunoglobulina G/metabolismo , Ratones , Serina/metabolismo , Serina Endopeptidasas/metabolismo , Serina Proteasas/metabolismo
18.
Gut Microbes ; 14(1): 2122667, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36138514

RESUMEN

Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC) cause gastrointestinal infection and, in severe cases, hemolytic uremic syndrome which may lead to death. There is, to-date, no therapy for this infection. Stx induces ATP release from host cells and ATP signaling mediates its cytotoxic effects. Apyrase cleaves and neutralizes ATP and its effect on Stx and EHEC infection was therefore investigated. Apyrase decreased bacterial RecA and dose-dependently decreased toxin release from E. coli O157:H7 in vitro, demonstrated by reduced phage DNA and protein levels. The effect was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice infected with Stx2-producing E. coli O157:H7 were treated with apyrase intraperitoneally, on days 0 and 2 post-infection, and monitored for 11 days. Apyrase-treated mice developed disease two days later than untreated mice. Untreated infected mice lost significantly more weight than those treated with apyrase. Apyrase-treated mice exhibited less colonic goblet cell depletion and apoptotic cells, as well as lower fecal ATP and Stx2, compared to untreated mice. Apyrase also decreased platelet aggregation induced by co-incubation of human platelet-rich-plasma with Stx2 and E. coli O157 lipopolysaccharide in the presence of collagen. Thus, apyrase had multiple protective effects, reducing RecA levels, stx2 and toxin release from EHEC, reducing fecal Stx2 and protecting mouse intestinal cells, as well as decreasing platelet activation, and could thereby delay the development of disease.


Asunto(s)
Bacteriófagos , Infecciones por Escherichia coli , Escherichia coli O157 , Microbioma Gastrointestinal , Adenosina Trifosfato/metabolismo , Animales , Apirasa/metabolismo , Apirasa/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Escherichia coli O157/genética , Humanos , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos BALB C , Toxina Shiga/metabolismo , Toxina Shiga/farmacología , Toxina Shiga II/genética , Toxina Shiga II/metabolismo , Toxina Shiga II/farmacología
19.
Nephrol Dial Transplant ; 26(10): 3399-403, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21810760

RESUMEN

A 4-year-old boy presented with proteinuria and developed progressive renal failure over 6 years. In the patient's family, five individuals were affected with atypical haemolytic uraemic syndrome (aHUS) but not the patient. Renal biopsies (n = 3) showed glomerular basement membrane thickening with double contours, endothelial swelling and deposits of C3 and C1q. Electron microscopy revealed mesangial and subendothelial electron-dense deposits. Complement mutations in membrane cofactor protein (Y155D) and C3 (R713W and G1094R) were detected in all affected family members. The patient also had transient autoantibodies to factor H. The findings suggest that aHUS and glomerulopathy resembling membranoproliferative glomerulonephritis may have a common molecular background.


Asunto(s)
Glomerulonefritis Membranoproliferativa/diagnóstico , Fallo Renal Crónico/etiología , Proteinuria/complicaciones , Microangiopatías Trombóticas/diagnóstico , Preescolar , Complemento C3/genética , Diagnóstico Diferencial , Femenino , Glomerulonefritis Membranoproliferativa/etiología , Humanos , Masculino , Proteína Cofactora de Membrana/genética , Mutación/genética , Linaje , Reacción en Cadena de la Polimerasa , Pronóstico , Microangiopatías Trombóticas/etiología
20.
Front Immunol ; 12: 690821, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177949

RESUMEN

Complement factor B (FB) mutant variants are associated with excessive complement activation in kidney diseases such as atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy and membranoproliferative glomerulonephritis (MPGN). Patients with aHUS are currently treated with eculizumab while there is no specific treatment for other complement-mediated renal diseases. In this study the phenotype of three FB missense variants, detected in patients with aHUS (D371G and E601K) and MPGN (I242L), was investigated. Patient sera with the D371G and I242L mutations induced hemolysis of sheep erythrocytes. Mutagenesis was performed to study the effect of factor D (FD) inhibition on C3 convertase-induced FB cleavage, complement-mediated hemolysis, and the release of soluble C5b-9 from glomerular endothelial cells. The FD inhibitor danicopan abrogated C3 convertase-associated FB cleavage to the Bb fragment in patient serum, and of the FB constructs, D371G, E601K, I242L, the gain-of-function mutation D279G, and the wild-type construct, in FB-depleted serum. Furthermore, the FD-inhibitor blocked hemolysis induced by the D371G and D279G gain-of-function mutants. In FB-depleted serum the D371G and D279G mutants induced release of C5b-9 from glomerular endothelial cells that was reduced by the FD-inhibitor. These results suggest that FD inhibition can effectively block complement overactivation induced by FB gain-of-function mutations.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/inmunología , Activación de Complemento , Factor B del Complemento/genética , Factor D del Complemento/antagonistas & inhibidores , Glomerulonefritis Membranoproliferativa/inmunología , Animales , Síndrome Hemolítico Urémico Atípico/genética , Niño , Convertasas de Complemento C3-C5/inmunología , Complemento C3b/inmunología , Factor B del Complemento/inmunología , Factor D del Complemento/inmunología , Células Endoteliales/inmunología , Eritrocitos , Femenino , Glomerulonefritis Membranoproliferativa/genética , Hemólisis , Humanos , Lactante , Glomérulos Renales/citología , Masculino , Persona de Mediana Edad , Mutación , Fenotipo , Conejos , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA