Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Pediatr Nephrol ; 39(8): 2459-2465, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38589699

RESUMEN

BACKGROUND: Hemolytic uremic syndrome (HUS) is an important cause of acute kidney injury in children. HUS is known as an acute disease followed by complete recovery, but patients may present with kidney abnormalities after long periods of time. This study evaluates the long-term outcome of Shiga toxin-producing Escherichia coli-associated HUS (STEC-HUS) in pediatric patients, 10 years after the acute phase of disease to identify risk factors for long-term sequelae. METHODS: Over a 6-year period, 619 patients under 18 years of age with HUS (490 STEC-positive, 79%) were registered in Austria and Germany. Long-term follow-up data of 138 STEC-HUS-patients were available after 10 years for analysis. RESULTS: A total of 66% (n = 91, 95% CI 0.57-0.73) of patients fully recovered showing no sequelae after 10 years. An additional 34% (n = 47, 95% CI 0.27-0.43) presented either with decreased glomerular filtration rate (24%), proteinuria (23%), hypertension (17%), or neurological symptoms (3%). Thirty had sequelae 1 year after STEC-HUS, and the rest presented abnormalities unprecedented at the 2-year (n = 2), 3-year (n = 3), 5-year (n = 3), or 10-year (n = 9) follow-up. A total of 17 patients (36.2%) without kidney abnormalities at the 1-year follow-up presented with either proteinuria, hypertension, or decreased eGFR in subsequent follow-up visits. Patients needing extracorporeal treatments during the acute phase were at higher risk of presenting symptoms after 10 years (p < 0.05). CONCLUSIONS: Patients with STEC-HUS should undergo regular follow-up, for a minimum of 10 years following their index presentation, due to the risk of long-term sequelae of their disease. An initial critical illness, marked by need of kidney replacement therapy or plasma treatment may help predict poor long-term outcome.


Asunto(s)
Infecciones por Escherichia coli , Síndrome Hemolítico-Urémico , Escherichia coli Shiga-Toxigénica , Humanos , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/terapia , Síndrome Hemolítico-Urémico/complicaciones , Síndrome Hemolítico-Urémico/epidemiología , Escherichia coli Shiga-Toxigénica/aislamiento & purificación , Masculino , Femenino , Niño , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/diagnóstico , Preescolar , Estudios de Seguimiento , Adolescente , Lactante , Alemania/epidemiología , Factores de Riesgo , Tasa de Filtración Glomerular , Austria/epidemiología , Factores de Tiempo , Proteinuria/etiología , Proteinuria/microbiología , Proteinuria/diagnóstico
2.
Microb Cell Fact ; 21(1): 1, 2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-34983506

RESUMEN

BACKGROUND: With the steady increase of antibiotic resistance, several strategies have been proposed in the scientific community to overcome the crisis. One of many successful strategies is the re-evaluation of known compounds, which have been early discarded out of the pipeline, with state-of-the-art know-how. Xanthoepocin, a polyketide widespread among the genus Penicillium with an interesting bioactivity spectrum against gram-positive bacteria, is such a discarded antibiotic. The purpose of this work was to (i) isolate larger quantities of this metabolite and chemically re-evaluate it with modern technology, (ii) to explore which factors lead to xanthoepocin biosynthesis in P. ochrochloron, and (iii) to test if it is beside its known activity against methicillin-resistant Staphylococcus aureus (MRSA), also active against linezolid and vancomycin-resistant Enterococcus faecium (LVRE)-a very problematic resistant bacterium which is currently on the rise. RESULTS: In this work, we developed several new protocols to isolate, extract, and quantify xanthoepocin out of bioreactor batch and petri dish-grown mycelium of P. ochrochloron. The (photo)chemical re-evaluation with state-of-the-art techniques revealed that xanthoepocin is a photolabile molecule, which produces singlet oxygen under blue light irradiation. The intracellular xanthoepocin content, which was highest under ammonium-limited conditions, varied considerably with the applied irradiation conditions in petri dish and bioreactor batch cultures. Using light-protecting measures, we achieved MIC values against gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA), which were up to 5 times lower than previously published. In addition, xanthoepocin was highly active against a clinical isolate of linezolid and vancomycin-resistant Enterococcus faecium (LVRE). CONCLUSIONS: This interdisciplinary work underlines that the re-evaluation of known compounds with state-of-the-art techniques is an important strategy in the combat against multiresistant bacteria and that light is a crucial factor on many levels that needs to receive more attention. With appropriate light protecting measures in the susceptibility tests, xanthoepocin proved to be a powerful antibiotic against MRSA and LVRE. Exploring the light response of other polyketides may be pivotal for re-introducing previously discarded metabolites into the antibiotic pipeline and to identify photosensitizers which might be used for (antimicrobial) photodynamic therapies.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple , Compuestos Epoxi/farmacología , Bacterias Grampositivas/efectos de los fármacos , Luz , Penicillium/química , Pironas/farmacología , Dispersión Dinámica de Luz , Pruebas de Sensibilidad Microbiana , Fotólisis
3.
Eur J Clin Microbiol Infect Dis ; 40(7): 1441-1449, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33547522

RESUMEN

Bloodstream infections (BSIs) require an accurate and fast identification of causative pathogens. Molecular diagnostics, in particular polymerase chain reaction (PCR)-based approaches for BSI diagnostics directly from whole blood, suffer from limitations such as inhibition leading to invalid results. In this retrospective study, we analyzed 23 parameters for their potential interference with LightCycler SeptiFast PCR tests (n = 2167) routinely performed at our institution. The overall inhibition rate was 9.1%. Test date, type of ward, procalcitonin levels, high leukocyte counts, and absolute neutrophil count were significantly associated with inhibition. For a subset (n = 448), cut-off values for leukocyte counts of < 5700 cells/µL and ≥ 26,900 cells/µL were significantly associated with a low (5%) and high (67%) inhibition risk. For patients with a moderate to high leukocyte count (5700-26,900 cells/µL), the additional administration of hydrocortisone significantly increased the inhibition risk. Furthermore, freezing of blood samples prior to DNA extraction and SF testing appeared to neutralize inhibitory factors. It remains to be investigated whether other molecular diagnostic tests are susceptible to similar inhibiting parameters.


Asunto(s)
Hidrocortisona/administración & dosificación , Técnicas de Diagnóstico Molecular/métodos , Reacción en Cadena de la Polimerasa/métodos , Sepsis/microbiología , Adolescente , Adulto , Anciano , Cultivo de Sangre/métodos , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa Multiplex , Estudios Retrospectivos , Sensibilidad y Especificidad , Adulto Joven
4.
Cell Microbiol ; 21(5): e13000, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30578712

RESUMEN

Hemolytic uremic syndrome (eHUS) is a severe complication of human infections with Shiga toxins (Stxs)-producing Escherichia coli. A key step in the pathogenesis of eHUS is the interaction of Stxs with blood components before the targeting of renal endothelial cells. Here, we show that a single proteolytic cleavage in the Stx2a A-subunit, resulting into two fragments (A1 and A2) linked by a disulfide bridge (cleaved Stx2a), dictates different binding abilities. Uncleaved Stx2a was confirmed to bind to human neutrophils and to trigger leukocyte/platelet aggregate formation, whereas cleaved Stx2a was ineffective. Conversely, binding of complement factor H was confirmed for cleaved Stx2a and not for uncleaved Stx2a. It is worth noting that uncleaved and cleaved Stx2a showed no differences in cytotoxicity for Vero cells or Raji cells, structural conformation, and contaminating endotoxin. These results have been obtained by comparing two Stx2a batches, purified in different laboratories by using different protocols, termed Stx2a(cl; cleaved toxin, Innsbruck) and Stx2a(uncl; uncleaved toxin, Bologna). Stx2a(uncl) behaved as Stx2a(cl) after mild trypsin treatment. In this light, previous controversial results obtained with purified Stx2a has to be critically re-evaluated; furthermore, characterisation of the structure of circulating Stx2a is mandatory to understand eHUS-pathogenesis and to develop therapeutic approaches.


Asunto(s)
Escherichia coli/química , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Chlorocebus aethiops , Dicroismo Circular , Factor H de Complemento/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fluorescencia , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Unión Proteica , Conformación Proteica , Toxina Shiga II/genética , Trihexosilceramidas/metabolismo , Tripsina , Células Vero
5.
Immunol Cell Biol ; 97(6): 538-551, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30695101

RESUMEN

Mucosal-associated invariant T (MAIT) cells are an abundant human T-cell subset with antimicrobial properties. They can respond to bacteria presented via antigen-presenting cells (APCs) such as macrophages, which present bacterially derived ligands from the riboflavin synthesis pathway on MR1. Moreover, MAIT cells are also highly responsive to cytokines which enhance and even substitute for T-cell receptor-mediated signaling. The mechanisms leading to an efficient presentation of bacteria to MAIT cells by APCs have not been fully elucidated. Here, we showed that the monocytic cell line THP-1 and B cells activated MAIT cells differentially in response to Escherichia coli. THP-1 cells were generally more potent in inducing IFNγ and IFNγ/TNF production by MAIT cells. Furthermore, THP-1, but not B, cells produced TNF upon bacterial stimulation, which in turn supported IFNγ production by MAIT cells. Finally, we addressed the role of antibody-dependent opsonization of bacteria in the activation of MAIT cells using in vitro models. We found that opsonization had a substantial impact on downstream MAIT cell activation by monocytes. This was associated with enhanced activation of monocytes and increased TNF release. Importantly, this TNF acted in concert with other cytokines to drive MAIT cell activation. These data indicate both a significant interaction between adaptive and innate immunity in the response to bacteria, and an important role for TNF in MAIT cell triggering.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/fisiología , Monocitos/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Inmunidad Adaptativa , Anticuerpos Antibacterianos/metabolismo , Presentación de Antígeno , Humanos , Inmunidad Innata , Interferón gamma/metabolismo , Activación de Linfocitos , Proteínas Opsoninas/metabolismo , Fagocitosis , Transducción de Señal , Células THP-1 , Factor de Necrosis Tumoral alfa/metabolismo
6.
BMC Microbiol ; 19(1): 180, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31387527

RESUMEN

BACKGROUND: Early pathogen detection and identification are crucial for an effective and targeted antibiotic therapy in patients suffering from blood stream infection. Molecular diagnostic methods can accelerate pathogen identification as compared to blood culture, but frequently suffer from the inhibition of polymerase chain reation (PCR) by sample matrix components, such as host DNA, anticoagulants, or plasma proteins. To overcome this limitation, molecular diagnostic methods commonly rely on pathogen enrichment by selective lysis of blood cells and pelleting of intact pathogens prior to analysis. RESULTS: Here, we investigated the impact of antibiotic treatment on the recovery of pathogen DNA using an established pathogen enrichment protocol. Based on the hypothesis that induction of bacterial cell wall disintegration following antibiotic administration leads to incomplete pelleting of pathogen DNA, S. aureus was grown in human whole blood with or without addition of cell wall active (vancomycin, piperacillin) or non cell wall active (ciprofloxacin, clindamycin) antibiotics at clinically relevant concentrations. Pathogen detection remained unaffected by non cell wall active antibiotics or even increased in the presence of cell wall active antibiotics, indicating improved accessibility of pathogen DNA. Likewise, mechanical lysis of S. aureus prior to pathogen enrichment resulted in increased recovery of pathogen DNA. Quantification of pathogen and human DNA after selective lysis of blood cells and pathogen enrichment confirmed partial depletion of human DNA, leading to a net enrichment of pathogen DNA over human DNA. CONCLUSION: Concurrent antibiotic administration does not reduce the recovery of pathogen DNA during pathogen enrichment by selective lysis and centrifugation. Leads to a 10-fold human DNA depletion as compared to pathogen DNA. Moreover, we confirm that the recovery of pathogen DNA after pathogen enrichment is not negatively influenced by concurrent antibiotic administration.


Asunto(s)
ADN Bacteriano/aislamiento & purificación , Técnicas de Diagnóstico Molecular/métodos , Antibacterianos/efectos adversos , ADN Bacteriano/sangre , ADN Bacteriano/genética , Infecciones Estafilocócicas/diagnóstico , Staphylococcus aureus/genética , Staphylococcus aureus/aislamiento & purificación
7.
J Med Virol ; 91(4): 570-581, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30351487

RESUMEN

BACKGROUND: This study aimed to characterize the epidemiology of pathogenic respiratory agents in patients aged 0 to 12 months and hospitalized for acute respiratory infections in Tunisia between 2013 and 2014. METHODS: A total of 20 pathogens, including viruses, Mycoplasma pneumoniae, and Streptococcus pneumoniae, were detected using molecular sensitive assays, and their associations with the patient's demographic data and season were analyzed. RESULTS: Viral infectious agents were found in 449 (87.2%) of 515 specimens. Dual and multiple infectious agents were detected in 31.4% and 18.6% of the samples, respectively. Viral infection was predominant in the pediatric environment (90.8%, P < 0.001), male patients (88.0%), and spring (93.8%). Rhinovirus was the most detected virus (51.8%) followed by respiratory syncytial virus A/B (34.4%), coronavirus group (18.5%), adenovirus (17.9%), and parainfluenza viruses 1-4 (10.9%). Respiratory Syncytial virus A/B was significantly associated with gender (38.0% male cases vs 28.3% female cases, P = 0.02). Infections by Adenovirus, Bocavirus, and Metapneumovirus A/B increased with increasing age of patients (predominated cases aged 6-12 months, P < 0.001). S. pneumoniae was detected in 30.9% of th tested samples. In 18.2% of the negative viral infections, only S. pneumoniae was identified. CONCLUSION: A predominance of the rhinovirus infection was observed in this study. Coronavirus subtypes were described for the first time in Tunisia. The observed different pathogenic profiles across age groups could be helpful to avoid the misclassification of patients presenting with ARIs at the triage level when no standardized protocol is available. This study will provide clues for physicians informing decisions regarding preventive strategies and medication in Tunisia.


Asunto(s)
Bacterias/aislamiento & purificación , Infecciones Bacterianas/epidemiología , Infecciones Bacterianas/microbiología , Infecciones del Sistema Respiratorio/epidemiología , Virosis/epidemiología , Virosis/virología , Virus/aislamiento & purificación , Bacterias/clasificación , Demografía , Femenino , Hospitalización , Humanos , Lactante , Recién Nacido , Masculino , Técnicas de Diagnóstico Molecular , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Túnez/epidemiología , Virus/clasificación
8.
Int J Med Microbiol ; 308(7): 969-976, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30064820

RESUMEN

Escherichia coli-induced hemolytic uremic syndrome (eHUS) is a life-threatening complication of infection with Shiga toxin (Stx), in particular Stx2a-producing Escherichia coli. Enhanced coagulation activation with formation of microthrombi seems to be a key event in development of eHUS. Platelet activation has been postulated as a possible, but controversially debated mechanism. The present study investigated the effect of Stx2a on plasmatic coagulation and platelets. Binding studies were initially performed with ELISA and co-immunoprecipitation and supported by quartz crystal microbalance with dissipation monitoring (QCM-D). Antithrombin (AT) activity was measured using the automated BCS XP® system. ROTEM® was used for functional coagulation testing. Platelet binding and activation was studied with FACS and light-transmission aggregometry. We found binding of Stx2a to AT, an important inhibitor of blood coagulation, but only a mild albeit significant reduction of AT activity against FXa in the presence of Stx2a. QCM-D analysis also showed binding of Stx2a to heparin and an impaired binding of AT to Stx2a-bound heparin. ROTEM® using Stx2a-treated platelet-poor plasma revealed a significant, but only moderate shortening of clotting time. Neither binding nor activation of platelets by Stx2a could be demonstrated. In summary, data of this study suggest that Stx2a binds to AT, but does not induce major effects on plasmatic coagulation. In addition, no interaction with platelets occurred. The well-known non-beneficial administration of heparin in eHUS patients could be explained by the interaction of Stx2a with heparin.


Asunto(s)
Antitrombinas/metabolismo , Coagulación Sanguínea/fisiología , Heparina/metabolismo , Agregación Plaquetaria/inmunología , Toxina Shiga II/metabolismo , Plaquetas/inmunología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Unión Proteica/fisiología , Escherichia coli Shiga-Toxigénica/patogenicidad
9.
Infection ; 46(5): 701-704, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29971692

RESUMEN

PURPOSE: To determine the burden of antifungal resistance in fungi over the last 10 years. METHODS: Performance of a semi-nationwide surveillance on antifungal resistance. RESULTS: We observed a low frequency of azole resistance in Aspergillus fumigatus, a moderate increase of echinocandin resistance in yeasts, and a stable amphotericin B activity in yeasts and molds. Posaconazole resistance in Aspergillus terreus occurred in a few isolates. CONCLUSION: The burden of resistance in fungi seems to be low in Tyrol, Austria.


Asunto(s)
Antifúngicos/farmacología , Azoles/farmacología , Farmacorresistencia Fúngica , Hongos/efectos de los fármacos , Micosis/epidemiología , Micosis/microbiología , Austria/epidemiología , Humanos , Pruebas de Sensibilidad Microbiana , Prevalencia , Vigilancia en Salud Pública
10.
Med Genet ; 30(4): 400-409, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30930551

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a disorder characterized by thrombocytopenia and microangiopathic hemolytic anemia due to endothelial injury. aHUS is felt to be caused by defective complement regulation due to underlying genetic mutations in complement regulators or activators, most often of the alternative pathway. Mutations causing aHUS can be subdivided into two groups, loss of function mutations (affecting factor H, factor H-related proteins, membrane co-factor protein, and factor I), and gain of function mutations (affecting factor B and C3). As more information becomes available on the relationship between specific mutations and clinical outcome, complete genetic workup of aHUS patients becomes more and more important. In this review, we will discuss the genetic background of aHUS, the role of complement for aHUS pathogenesis, and the different groups of specific mutations known to be involved in the pathogenesis of aHUS.

11.
Infection ; 45(6): 867-875, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28956284

RESUMEN

BACKGROUND: Invasive fungal diseases (IFD) are an important cause of morbidity and mortality in immunocompromised patients, and early diagnosis and management are a challenge. We evaluated the clinical utility of computed tomography (CT)-guided percutaneous lung biopsies in diagnosing IFD. METHODS: Between 2003 and 2014, we analyzed 2671 CT-guided lung biopsies, from which 157 were IFD associated; we aimed to determine microbiological-based diagnostic accuracy of calcofluor white staining (CFWS), culture, Aspergillus antigen detection (GM), broad-range fungal PCR, and Aspergillus PCR per sample. RESULTS: 127 (81%) specimens were microscopically positive for any fungal elements, 30 (19%) negative. Aspergillus and non-Aspergillus like hyphae were obtained in 85 (67%) and 42 (33%) specimens, respectively. CFWS positivity was defined as proof of infection. Sensitivity, specificity, and positive (PPV) and negative predictive (NPV) values for CT scan were 100, 44, 80, and 100%, for Aspergillus PCR 89, 58, 88, and 58%, for broad-range fungal PCR 90, 83, 95, and 90%, and for GM 94, 83, 95, and 90%. The most common CT features were patchy opacifications with central necrosis (78%) or cavern defects (50%), less common were air bronchograms (39%) or ground glass halos (39%), and all other features were rare. The overall pneumothorax rate subsequent to biopsy was 19%, but in only 2% of all cases the placement of a chest tube was indicated. One case of fatal air embolism occurred. CONCLUSIONS: CT-guided lung biopsies have high diagnostic accuracy in terms of microscopic examination, and complication rates are low. Molecular-based and antigen tests applied on fungal hyphae-positive specimens showed comparable results.


Asunto(s)
Aspergilosis/diagnóstico , Huésped Inmunocomprometido , Enfermedades Pulmonares Fúngicas/diagnóstico , Pulmón/patología , Tomografía Computarizada por Rayos X/métodos , Anciano , Antígenos Fúngicos/sangre , Aspergilosis/diagnóstico por imagen , Aspergilosis/microbiología , Aspergillus/aislamiento & purificación , Austria , Bencenosulfonatos/química , Biopsia/métodos , Femenino , Humanos , Pulmón/diagnóstico por imagen , Enfermedades Pulmonares Fúngicas/diagnóstico por imagen , Enfermedades Pulmonares Fúngicas/microbiología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa/métodos , Manejo de Especímenes/métodos
13.
Eur J Haematol ; 94(3): 258-64, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25082655

RESUMEN

Mould-active antifungal prophylaxis is increasingly used in patients at risk for invasive fungal disease. Between June 2011 and June 2012, one hundred patients with various haematological malignancies at risk for invasive fungal disease received primary antifungal prophylaxis with intravenous micafungin at a daily dosage of 50 mg during neutropenia. The median number of days on micafungin prophylaxis was 14 (range, 6-48 d). The incidence of proven and probable breakthrough invasive fungal diseases (bIFDs) was 6% and 3%, respectively. There were two bloodstream infections caused by yeasts or yeast-like fungi (Candida krusei, Trichosporon asahii) in two patients during the neutropenic phase after allogeneic haematopoietic stem cell transplantation. Four proven bIFDs caused by non-Aspergillus moulds and three cases of probable pulmonary bIFDs were documented during the neutropenic phase after induction/consolidation chemotherapy for acute leukaemia. Colonisation with Candida spp. was documented in 51% of the patients with none of the isolates being in vitro micafungin resistant. Compared to a historical control, receiving primary prophylaxis with posaconazole micafungin is at least as effective in preventing IFD. In both cohorts, bIFDs were exclusively caused by emerging pathogens with a highly preserved in vitro sensitivity to amphotericin B.


Asunto(s)
Antifúngicos/uso terapéutico , Candidiasis/prevención & control , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Tricosporonosis/prevención & control , Adulto , Anciano , Anfotericina B/uso terapéutico , Candida/aislamiento & purificación , Candidiasis/complicaciones , Candidiasis/microbiología , Candidiasis/patología , Esquema de Medicación , Equinocandinas/uso terapéutico , Femenino , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/microbiología , Neoplasias Hematológicas/patología , Humanos , Inyecciones Intravenosas , Lipopéptidos/uso terapéutico , Masculino , Micafungina , Persona de Mediana Edad , Estudios Retrospectivos , Trasplante Homólogo , Triazoles/uso terapéutico , Trichosporon/aislamiento & purificación , Tricosporonosis/complicaciones , Tricosporonosis/microbiología , Tricosporonosis/patología
14.
Infect Immun ; 82(5): 1872-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24566618

RESUMEN

Hemolytic uremic syndrome (HUS) is mainly induced by Shiga toxin 2 (Stx2)-producing Escherichia coli. Proteinuria can occur in the early phase of the disease, and its persistence determines the renal prognosis. Stx2 may injure podocytes and induce proteinuria. Human serum amyloid P component (SAP), a member of the pentraxin family, has been shown to protect against Stx2-induced lethality in mice in vivo, presumably by specific binding to the toxin. We therefore tested the hypothesis that SAP can protect against Stx2-induced injury of human podocytes. To elucidate the mechanisms underlying podocyte injury in HUS-associated proteinuria, we assessed Stx2-induced activation of mitogen-activated protein kinases (MAPKs) and apoptosis in immortalized human podocytes and evaluated the impact of SAP on Stx2-induced damage. Human podocytes express Stx2-binding globotriaosylceramide 3. Stx2 applied to cultured podocytes was internalized and then activated p38α MAPK and c-Jun N-terminal kinase (JNK), important signaling steps in cell differentiation and apoptosis. Stx2 also activated caspase 3, resulting in an increased level of apoptosis. Coincubation of podocytes with SAP and Stx2 mitigated the effects of Stx2 and induced upregulation of antiapoptotic Bcl2. These data suggest that podocytes are a target of Stx2 and that SAP protects podocytes against Stx2-induced injury. SAP may therefore be a useful therapeutic option.


Asunto(s)
Apoptosis/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Podocitos/efectos de los fármacos , Componente Amiloide P Sérico/farmacología , Toxina Shiga II/toxicidad , Animales , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Fosforilación , Podocitos/metabolismo , Podocitos/fisiología , Transducción de Señal , Regulación hacia Arriba
15.
Semin Thromb Hemost ; 40(4): 503-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24799304

RESUMEN

Hemolytic uremic syndrome (HUS), the most common cause of acute renal failure in childhood, is mainly caused by infection with Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC). Besides its cytotoxic activity, Stx has been shown to interact with the complement system. Complement breakdown products have been found in serum of HUS patients suggesting complement activation and in vitro studies have demonstrated that Stx2 directly activates complement leading to formation of terminal complement complex. Furthermore, Stx2 has been found to bind to factor H (FH) resulting in a reduced cofactor activity on the cell surface. Binding of Stx2 has also been shown for other members of the FH family, namely FH-like protein 1 and FH-related protein 1. Both proteins also compete with FH for Stx binding, so that in the presence of FHR-1 less FH is bound to Stx and therefore more is available for endothelial cell protection. In addition, Stx2 has been demonstrated to downregulate the membrane-bound regulator CD59 on the surface of glomerular endothelial and tubulus epithelial cells on protein and at the mRNA level. In conclusion, Stx modulates complement regulator proteins leading to an impaired control and thus to enhanced complement activation. Its implication in the pathogenesis of EHEC-induced HUS in vivo and whether complement blockage might be a therapeutic option still has to be elucidated.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Escherichia coli Enterohemorrágica , Síndrome Hemolítico-Urémico/genética , Síndrome Hemolítico-Urémico/inmunología , Toxina Shiga II/química , Toxina Shiga/química , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD59/metabolismo , Membrana Celular/metabolismo , Activación de Complemento , Factor H de Complemento/fisiología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Glomérulos Renales/inmunología , Glomérulos Renales/metabolismo , Mutación
16.
Semin Thromb Hemost ; 40(4): 508-16, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24802085

RESUMEN

Treatment of enterohemorrhagic Escherichia coli-induced hemolytic uremic syndrome (eHUS) still mostly relies on supportive intensive care regimens. Antibiotic treatment, as administered to eHUS patients during the 2011 O104:H4 outbreak, may reduce the shedding period, but this may apply only to this particular strain. In any case, there is no evidence for a beneficial use in the diarrheal phase and earlier warnings that antibiotic therapy at this stage may actually increase the likelihood of HUS remain unrefuted. Plasma exchange, a frequently chosen therapy in acute atypical HUS, was not beneficial for the outbreak patients and a prospective study of 274 pediatric eHUS patients even indicates a poorer long-term outcome. As eHUS is a disease where complement plays a pathophysiological role and individual beneficial treatments had been published, eculizumab was broadly administered during the outbreak, in particular to severely ill patients. The equally good outcome of treated versus untreated patients obviously does not allow a clear-cut statement, but rather points toward an advantageous use, at least for the severe cases. Although the role of complement should not be overestimated, the use of a complement blocker--not necessarily being a therapeutic option for uncomplicated eHUS--in severe disease may actually make the difference between favorable or detrimental outcome.


Asunto(s)
Antibacterianos/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Escherichia coli Enterohemorrágica , Síndrome Hemolítico-Urémico/terapia , Adsorción , Proteínas del Sistema Complemento , Brotes de Enfermedades , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/terapia , Alemania , Síndrome Hemolítico-Urémico/epidemiología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Mutación , Intercambio Plasmático , Resultado del Tratamiento
17.
Front Immunol ; 15: 1330095, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38333209

RESUMEN

Introduction: The complement system is part of innate immunity and is comprised of an intricate network of proteins that are vital for host defense and host homeostasis. A distinct mechanism by which complement defends against invading pathogens is through the membrane attack complex (MAC), a lytic structure that forms on target surfaces. The MAC is made up of several complement components, and one indispensable component of the MAC is C7. The role of C7 in MAC assembly is well documented, however, inherent characteristics of C7 are yet to be investigated. Methods: To shed light on the molecular characteristics of C7, we examined the properties of serum-purified C7 acquired using polyclonal and novel monoclonal antibodies. The properties of serum­purified C7 were investigated through a series of proteolytic analyses, encompassing Western blot and mass spectrometry. The nature of C7 protein-protein interactions were further examined by a novel enzyme-linked immunosorbent assay (ELISA), as well as size­exclusion chromatography. Results: Protein analyses showcased an association between C7 and clusterin, an inhibitory complement regulator. The distinct association between C7 and clusterin was also demonstrated in serum-purified clusterin. Further assessment revealed that a complex between C7 and clusterin (C7-CLU) was detected. The C7-CLU complex was also identified in healthy serum and plasma donors, highlighting the presence of the complex in circulation. Discussion: Clusterin is known to dissociate the MAC structure by binding to polymerized C9, nevertheless, here we show clusterin binding to the native form of a terminal complement protein in vivo. The presented data reveal that C7 exhibits characteristics beyond that of MAC assembly, instigating further investigation of the effector role that the C7-CLU complex plays in the complement cascade.


Asunto(s)
Clusterina , Complemento C7 , Complemento C7/metabolismo , Proteínas del Sistema Complemento/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Activación de Complemento
18.
Cell Rep ; 43(1): 113611, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38159276

RESUMEN

Complement is a fundamental innate immune response component. Its alterations are associated with severe systemic diseases. To illuminate the complement's genetic underpinnings, we conduct genome-wide association studies of the functional activity of the classical (CP), lectin (LP), and alternative (AP) complement pathways in the Cooperative Health Research in South Tyrol study (n = 4,990). We identify seven loci, encompassing 13 independent, pathway-specific variants located in or near complement genes (CFHR4, C7, C2, MBL2) and non-complement genes (PDE3A, TNXB, ABO), explaining up to 74% of complement pathways' genetic heritability and implicating long-range haplotypes associated with LP at MBL2. Two-sample Mendelian randomization analyses, supported by transcriptome- and proteome-wide colocalization, confirm known causal pathways, establish within-complement feedback loops, and implicate causality of ABO on LP and of CFHR2 and C7 on AP. LP causally influences collectin-11 and KAAG1 levels and the risk of mouth ulcers. These results build a comprehensive resource to investigate the role of complement in human health.


Asunto(s)
Estudio de Asociación del Genoma Completo , Lectina de Unión a Manosa , Humanos , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Lectinas/metabolismo , Haplotipos/genética , Lectina de Unión a Manosa/genética
19.
Infect Immun ; 81(8): 2678-85, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23690395

RESUMEN

Infections with enterohemorrhagic Escherichia coli (EHEC) are a primary cause of hemolytic-uremic syndrome (HUS). Recently, Shiga toxin 2 (Stx2), the major virulence factor of EHEC, was reported to interact with complement, implying that the latter is involved in the pathogenesis of EHEC-induced HUS. The aim of the present study was to investigate the effect of Stx2 on the expression of membrane-bound complement regulators CD46, CD55, and CD59 on proximal tubular epithelial (HK-2) and glomerular endothelial (GEnC) cells derived from human kidney cells that are involved in HUS. Incubation with Stx2 did not influence the amount of CD46 or CD55 on the surface of HK-2 and GEnC cells, as determined by fluorescence-activated cell sorter analysis. In contrast, CD59 was significantly reduced by half on GEnC cells, but the reduction on HK-2 cells was less pronounced. With increasing amounts of Stx2, reduction of CD59 also reached significance in HK-2 cells. Enzyme-linked immunosorbent assay analyses showed that CD59 was not present in the supernatant of Stx2-treated cells, implying that CD59 reduction was not caused by cleavage from the cell surface. In fact, reverse transcription-quantitative PCR analyses showed downregulation of CD59 mRNA as the likely reason for CD59 cell surface reduction. In addition, a significant increase in terminal complement complex deposition on HK-2 cells was observed after treatment with Stx2, as a possible consequence of CD59 downregulation. In summary, Stx2 downregulates CD59 mRNA and protein levels on tubular epithelial and glomerular endothelial cells, and this downregulation likely contributes to complement activation and kidney destruction in EHEC-associated HUS.


Asunto(s)
Antígenos CD59/metabolismo , Células Endoteliales/metabolismo , Glomérulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Toxina Shiga II/metabolismo , Línea Celular , Escherichia coli Enterohemorrágica , Ensayo de Inmunoadsorción Enzimática , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/metabolismo , Citometría de Flujo , Síndrome Hemolítico-Urémico/metabolismo , Síndrome Hemolítico-Urémico/microbiología , Humanos , Microscopía Confocal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Clin Infect Dis ; 56(10): 1373-81, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23378282

RESUMEN

BACKGROUND: Enterohemorrhagic Escherichia coli (EHEC) O26 causes diarrhea and hemolytic uremic syndrome (HUS). Strains harboring the stx1a gene prevail, but strains with stx2a as the sole Shiga toxin-encoding gene are now emerging. The traits and virulence of the latter set of strains are unknown. We correlated stx genotypes of 272 EHEC O26 strains isolated in 7 European countries between 1996 and 2012 with disease phenotypes. We determined phylogeny, clonal structure, and plasmid gene profiles of the isolates and portray geographic and temporal distribution of the different subgroups. METHODS: The stx genotypes and plasmid genes were identified using polymerase chain reaction, phylogeny was assigned using multilocus sequence typing, and clonal relatedness was established using pulsed-field gel electrophoresis. RESULTS: Of the 272 EHEC O26 isolates, 107 (39.3%), 139 (51.1%), and 26 (9.6%) possessed stx1a, stx2a, or both genes, respectively. Strains harboring stx2a only were significantly associated with HUS (odds ratio, 14.2; 95% confidence interval, 7.9-25.6; P < .001) compared to other stx genotypes. The stx2a-harboring strains consist of 2 phylogenetically distinct groups defined by sequence type (ST) 21 and ST29. The ST29 strains are highly conserved and correspond by plasmid genes to the new virulent clone of EHEC O26 that emerged in Germany in the 1990s. This new clone occurred in 6 of the 7 countries and represented approximately 50% of all stx2a-harboring EHEC O26 strains isolated between 1996 and 2012. CONCLUSIONS: A new highly virulent clone of EHEC O26 has emerged in Europe. Its reservoirs and sources warrant identification.


Asunto(s)
Escherichia coli Enterohemorrágica/aislamiento & purificación , Infecciones por Escherichia coli/microbiología , Adolescente , Adulto , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Distribución de Chi-Cuadrado , Niño , Preescolar , ADN Bacteriano/genética , Electroforesis en Gel de Campo Pulsado , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/epidemiología , Europa (Continente)/epidemiología , Femenino , Humanos , Lactante , Masculino , Pruebas de Sensibilidad Microbiana , Fenotipo , Filogenia , Plásmidos/genética , Reacción en Cadena de la Polimerasa , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA