Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
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 ; 102(1): 14-16, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35738827

RESUMEN

Krüppel-like factors (KLFs) are transcription factors with important roles in tissue homeostasis. KLF4 possesses antithrombotic and anti-inflammatory properties. In this issue, Estrada et al. show that endothelial KLF4 prevents complement deposition in glomeruli and in its absence the cell-bound complement regulator CD55 was reduced. The study included endothelial-specific KLF4 knockdown mice that mimic thrombotic microangiopathy and thrombotic microangiopathy patient biopsies showing decreased KLF4 and CD55. The results suggest that KLF4 is involved in the regulation of glomerular complement deposition.


Asunto(s)
Activación de Complemento , Enfermedades Renales , Factor 4 Similar a Kruppel , Microangiopatías Trombóticas , Animales , Humanos , Enfermedades Renales/etiología , Glomérulos Renales , Factor 4 Similar a Kruppel/genética , Ratones , Factores de Transcripción
4.
Int J Mol Sci ; 23(23)2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36498939

RESUMEN

A recently developed inhibitor of retrograde transport, namely Retro-2.1, proved to be a potent and broad-spectrum lead in vitro against intracellular pathogens, such as toxins, parasites, intracellular bacteria and viruses. To circumvent its low aqueous solubility, a formulation in poly(ethylene glycol)-block-poly(D,L)lactide micelle nanoparticles was developed. This formulation enabled the study of the pharmacokinetic parameters of Retro-2.1 in mice following intravenous and intraperitoneal injections, revealing a short blood circulation time, with an elimination half-life of 5 and 6.7 h, respectively. To explain the poor pharmacokinetic parameters, the metabolic stability of Retro-2.1 was studied in vitro and in vivo, revealing fast cytochrome-P-450-mediated metabolism into a less potent hydroxylated analogue. Subcutaneous injection of Retro-2.1 formulated in a biocompatible and bioresorbable polymer-based thermosensitive hydrogel allowed for sustained release of the drug, with an elimination half-life of 19 h, and better control of its metabolism. This study provides a guideline on how to administer this promising lead in vivo in order to study its efficacy.


Asunto(s)
Hidrogeles , Nanopartículas , Ratones , Animales , Preparaciones de Acción Retardada , Polietilenglicoles , Polímeros , Temperatura
5.
Pediatr Nephrol ; 34(1): 11-30, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29181712

RESUMEN

Extracellular vesicles are cell-derived membrane particles ranging from 30 to 5,000 nm in size, including exosomes, microvesicles, and apoptotic bodies. They are released under physiological conditions, but also upon cellular activation, senescence, and apoptosis. They play an important role in intercellular communication. Their release may also maintain cellular integrity by ridding the cell of damaging substances. This review describes the biogenesis, uptake, and detection of extracellular vesicles in addition to the impact that they have on recipient cells, focusing on mechanisms important in the pathophysiology of kidney diseases, such as thrombosis, angiogenesis, tissue regeneration, immune modulation, and inflammation. In kidney diseases, extracellular vesicles may be utilized as biomarkers, as they are detected in both blood and urine. Furthermore, they may contribute to the pathophysiology of renal disease while also having beneficial effects associated with tissue repair. Because of their role in the promotion of thrombosis, inflammation, and immune-mediated disease, they could be the target of drug therapy, whereas their favorable effects could be utilized therapeutically in acute and chronic kidney injury.


Asunto(s)
Comunicación Celular/fisiología , Micropartículas Derivadas de Células/fisiología , Exosomas/fisiología , Enfermedades Renales/patología , Riñón/patología , Apoptosis/fisiología , Senescencia Celular/fisiología , Humanos
6.
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
7.
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
8.
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
9.
Kidney Int ; 91(1): 96-105, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27914700

RESUMEN

During vasculitis, activation of the kinin system induces inflammation, whereby the kinin B1-receptor is expressed and activated after ligand binding. Additionally, activated blood cells release microvesicles into the circulation. Here we determined whether leukocyte-derived microvesicles bear B1-kinin receptors during vasculitis, and if microvesicles transfer functional B1-receptors to recipient cells, thus promoting inflammation. By flow cytometry, plasma from patients with vasculitis were found to contain high levels of leukocyte-derived microvesicles bearing B1-receptors. Importantly, renal biopsies from two patients with vasculitis showed leukocyte-derived microvesicles bearing B1-receptors docking on glomerular endothelial cells providing in vivo relevance. Microvesicles derived from B1-receptor-transfected human embryonic kidney cells transferred B1-receptors to wild-type human embryonic kidney cells, lacking the receptor, and to glomerular endothelial cells. The transferred B1-receptors induced calcium influx after B1-receptor agonist stimulation: a response abrogated by a specific B1-receptor antagonist. Microvesicles derived from neutrophils also transferred B1-receptors to wild-type human embryonic kidney cells and induced calcium influx after stimulation. Thus, we found a novel mechanism by which microvesicles transfer functional receptors and promote kinin-associated inflammation.


Asunto(s)
Bradiquinina/metabolismo , Micropartículas Derivadas de Células/metabolismo , Inflamación/metabolismo , Riñón/metabolismo , Receptor de Bradiquinina B1/metabolismo , Vasculitis/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Calcio , Línea Celular , Niño , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Humanos , Riñón/citología , Cininas , Leucocitos/metabolismo , Masculino , Persona de Mediana Edad , Neutrófilos/metabolismo , Receptor de Bradiquinina B1/sangre , Receptor de Bradiquinina B1/genética , Transfección , Adulto Joven
10.
J Intern Med ; 281(2): 123-148, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27723152

RESUMEN

Haemolytic uraemic syndrome (HUS) is defined by the simultaneous occurrence of nonimmune haemolytic anaemia, thrombocytopenia and acute renal failure. This leads to the pathological lesion termed thrombotic microangiopathy, which mainly affects the kidney, as well as other organs. HUS is associated with endothelial cell injury and platelet activation, although the underlying cause may differ. Most cases of HUS are associated with gastrointestinal infection with Shiga toxin-producing enterohaemorrhagic Escherichia coli (EHEC) strains. Atypical HUS (aHUS) is associated with complement dysregulation due to mutations or autoantibodies. In this review, we will describe the causes of HUS. In addition, we will review the clinical, pathological, haematological and biochemical features, epidemiology and pathogenetic mechanisms as well as the biochemical, microbiological, immunological and genetic investigations leading to diagnosis. Understanding the underlying mechanisms of the different subtypes of HUS enables tailoring of appropriate treatment and management. To date, there is no specific treatment for EHEC-associated HUS but patients benefit from supportive care, whereas patients with aHUS are effectively treated with anti-C5 antibody to prevent recurrences, both before and after renal transplantation.


Asunto(s)
Síndrome Hemolítico-Urémico , Diagnóstico Diferencial , Síndrome Hemolítico-Urémico/diagnóstico , Síndrome Hemolítico-Urémico/epidemiología , Síndrome Hemolítico-Urémico/fisiopatología , Síndrome Hemolítico-Urémico/terapia , Humanos , Pronóstico
11.
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
12.
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
13.
Pediatr Nephrol ; 32(1): 1-6, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27738765

RESUMEN

Orphan drugs designed to treat rare diseases are often overpriced per patient. Novel treatments are sometimes even more expensive for patients with ultra-rare diseases, in part due to the limited number of patients. Pharmaceutical companies that develop a patented life-saving drug are in a position to charge a very high price, which, at best, may enable these companies to further develop drugs for use in rare disease. However, is there a limit to how much a life-saving drug should cost annually per patient? Government interventions and regulations may opt to withhold a life-saving drug solely due to its high price and cost-effectiveness. Processes related to drug pricing, reimbursement, and thereby availability, vary between countries, thus having implications on patient care. These processes are discussed, with specific focus on three drugs used in pediatric nephrology: agalsidase beta (for Fabry disease), eculizumab (for atypical hemolytic uremic syndrome), and cysteamine bitartrate (for cystinosis). Access to and costs of orphan drugs have most profound implications for patients, but also for their physicians, hospitals, insurance policies, and society at large, particularly from financial and ethical standpoints.


Asunto(s)
Enfermedades Renales/tratamiento farmacológico , Nefrología/ética , Producción de Medicamentos sin Interés Comercial/ética , Enfermedades Raras/tratamiento farmacológico , Niño , Análisis Costo-Beneficio , Costos de los Medicamentos , Industria Farmacéutica , Humanos , Enfermedades Renales/economía , Nefrología/economía , Producción de Medicamentos sin Interés Comercial/economía , Producción de Medicamentos sin Interés Comercial/legislación & jurisprudencia , Políticas
14.
Kidney Int ; 90(4): 726-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27633864

RESUMEN

Complement is activated during Shiga toxin-producing Escherichia coli-associated hemolytic uremic syndrome (STEC-HUS). There is evidence of complement activation via the alternative pathway in STEC-HUS patients as well as from in vivo and in vitro models. Ozaki et al. demonstrate activation of the mannose-binding lectin (MBL) pathway in Shiga toxin-treated mice expressing human MBL2, but lacking murine Mbls. Treatment with anti-human MBL2 antibody was protective, suggesting that MBL pathway activation also contributes to Shiga toxin-mediated renal injury.


Asunto(s)
Infecciones por Escherichia coli , Síndrome Hemolítico-Urémico , Lectina de Unión a Manosa , Escherichia coli Shiga-Toxigénica , Animales , Humanos , Riñón , Ratones , Toxina Shiga
15.
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
16.
Pediatr Nephrol ; 31(1): 15-39, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25859752

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) emerged during the last decade as a disease largely of complement dysregulation. This advance facilitated the development of novel, rational treatment options targeting terminal complement activation, e.g., using an anti-C5 antibody (eculizumab). We review treatment and patient management issues related to this therapeutic approach. We present consensus clinical practice recommendations generated by HUS International, an international expert group of clinicians and basic scientists with a focused interest in HUS. We aim to address the following questions of high relevance to daily clinical practice: Which complement investigations should be done and when? What is the importance of anti-factor H antibody detection? Who should be treated with eculizumab? Is plasma exchange therapy still needed? When should eculizumab therapy be initiated? How and when should complement blockade be monitored? Can the approved treatment schedule be modified? What approach should be taken to kidney and/or combined liver-kidney transplantation? How should we limit the risk of meningococcal infection under complement blockade therapy? A pressing question today regards the treatment duration. We discuss the need for prospective studies to establish evidence-based criteria for the continuation or cessation of anticomplement therapy in patients with and without identified complement mutations.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/terapia , Nefrología/normas , Adolescente , Factores de Edad , Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/epidemiología , Síndrome Hemolítico Urémico Atípico/inmunología , Niño , Preescolar , Terapia Combinada , Activación de Complemento/efectos de los fármacos , Consenso , Conducta Cooperativa , Monitoreo de Drogas , Humanos , Factores Inmunológicos/uso terapéutico , Lactante , Recién Nacido , Cooperación Internacional , Trasplante de Riñón , Trasplante de Hígado , Monitorización Inmunológica , Selección de Paciente , Intercambio Plasmático , Valor Predictivo de las Pruebas , Factores de Riesgo , Resultado del Tratamiento
17.
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
18.
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
19.
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
20.
Pediatr Nephrol ; 29(11): 2225-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24924752

RESUMEN

BACKGROUND: Immunoglobulin A (IgA) nephropathy is a chronic glomerulonephritis with excessive glomerular deposition of IgA1, C3 and C5b-9, which may lead to renal failure. CASE DIAGNOSIS/TREATMENT: We describe the clinical course of an adolescent with rapidly progressive disease leading to renal failure in spite of immunosuppressive treatment. Due to refractory disease the patient was treated with eculizumab (anti-C5) for 3 months in an attempt to rescue renal function. Treatment led to clinical improvement with stabilization of the glomerular filtration rate and reduced proteinuria. Discontinuation of treatment led to a rapid deterioration of renal function. This was followed by a single dose of eculizumab, which again reduced creatinine levels temporarily. CONCLUSIONS: Early initiation of eculizumab therapy in patients with progressive IgA nephropathy may have a beneficial effect by blocking complement-mediated renal inflammation.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Glomerulonefritis por IGA/tratamiento farmacológico , Glomerulonefritis por IGA/patología , Riñón/patología , Terapia Recuperativa/métodos , Adolescente , Biopsia , Creatinina/sangre , Tasa de Filtración Glomerular , Humanos , Glomérulos Renales/patología , Masculino , Insuficiencia Renal/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA