Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
BMC Nephrol ; 21(1): 260, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32646497

RESUMO

BACKGROUND: The seroprevalence of human Parvovirus B19 (PVB19) is 70-85% in adults worldwide. PVB19 is the etiologic agent of the fifth disease, is a cause of aplastic anemia, and can be associated with kidney injury. We aimed to describe the cases of 4 patients with kidney injury related to PVB19 primary infection, and to evaluate the seroprevalence of PVB19 and the incidence of PVB19 primary infection in patients undergoing a native kidney biopsy. METHODS: Cases of PVB19 infection with kidney injury were reviewed from the archives of the department of Nephrology. A systematic screening of anti-PVB19 IgG and IgM antibodies and viral DNA was performed in sera from 100 consecutive patients with a kidney biopsy in 2017-2018. RESULTS: The 4 patients with PVB19 infection-associated kidney disease displayed: one lupus-like glomerulonephritis (GN) without lupus auto-antibodies, one minimal change disease with tubular necrosis, one secondary hemolytic and uremic syndrome and one membrano-proliferative GN. In the 100 patients biopsied, 67 had elevated anti-PVB19 IgG, among whom 8 had elevated IgM, without circulating viral DNA, without any particular renal pathological pattern. One additional patient showed a seroconversion at the time of kidney biopsy, which revealed a class V lupus nephritis. CONCLUSION: PVB19 primary infection can be associated with different kidney diseases. The seroprevalence of PVB19 among patients with a kidney biopsy is similar to the overall population, and primary infection is rarely documented (1%) after systematic screening. Whether PV19 is nephrotoxic, or triggers renal endothelial injury and immune activation, remains to be elucidated.


Assuntos
Injúria Renal Aguda/virologia , Anticorpos Antivirais/imunologia , DNA Viral/sangue , Eritema Infeccioso/imunologia , Parvovirus B19 Humano/imunologia , Injúria Renal Aguda/sangue , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Eritema Infeccioso/sangue , Eritema Infeccioso/complicações , Feminino , Glomerulonefrite/sangue , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Glomerulonefrite/virologia , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/imunologia , Glomerulonefrite Membranoproliferativa/patologia , Glomerulonefrite Membranoproliferativa/virologia , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/patologia , Síndrome Hemolítico-Urêmica/virologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Incidência , Rim , Necrose Tubular Aguda/sangue , Necrose Tubular Aguda/imunologia , Necrose Tubular Aguda/patologia , Necrose Tubular Aguda/virologia , Masculino , Pessoa de Meia-Idade , Nefrose Lipoide/sangue , Nefrose Lipoide/imunologia , Nefrose Lipoide/patologia , Nefrose Lipoide/virologia , Parvovirus B19 Humano/genética , Estudos Soroepidemiológicos , Viremia/sangue , Adulto Jovem
2.
BMC Nephrol ; 21(1): 96, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32164588

RESUMO

BACKGROUND: Thrombotic microangiopathy (TMA) syndromes are characterized by the association of hemolytic anemia, thrombocytopenia and organ injury due to arteriolar and capillary thrombosis. CASE PRESENTATION: We report the first case of adult onset cobalamin C (Cbl C) disease associated with anti-factor H antibody-associated hemolytic uremic syndrome (HUS). A 19-year-old woman was admitted to the nephrology department owing to acute kidney failure, proteinuria, and hemolytic anemia with schizocytes. TMA was diagnosed and plasma exchanges were started in emergency. Exhaustive analyses showed 1) circulating anti factor H antibody and 2) hyperhomocysteinemia, hypomethioninemia and high levels of methylmalonic aciduria pointing towards Clb C disease. Cbl C disease has been confirmed by methylmalonic aciduria and homocystinuria type C protein gene sequencing revealing two heterozygous pathogenic variants. The kidney biopsy showed 1) intraglomerular and intravascular thrombi 2) noticeable thickening of the capillary wall with a duplication aspect of the glomerular basement membrane and a glomerular capillary wall IgM associated with Cbl C disease related TMA. We initiated treatment including hydroxycobalamin, folinic acid, betaine and levocarnitine and Eculizumab. Rituximab infusions were performed allowing a high decrease in anti-factor H antibody rate. Six month after the disease onset, Eculizumab was weaning and vitaminotherapy continued. Outcome was favorable with a dramatic improvement in kidney function. CONCLUSION: TMA with renal involvement can have a complex combination of risk factors including anti-FH autoantibody in the presence of cblC deficiency.


Assuntos
Autoanticorpos/sangue , Fator H do Complemento/imunologia , Síndrome Hemolítico-Urêmica/complicações , Síndrome Hemolítico-Urêmica/imunologia , Deficiência de Vitamina B 12/complicações , Adulto , Biópsia , Feminino , Síndrome Hemolítico-Urêmica/patologia , Síndrome Hemolítico-Urêmica/terapia , Humanos
3.
Pediatr Nephrol ; 34(8): 1465-1482, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30989342

RESUMO

BACKGROUND: Hemolytic uremic syndrome (HUS) is a leading cause of acute kidney injury in children. Although international guidelines emphasize comprehensive evaluation and treatment with eculizumab, access to diagnostic and therapeutic facilities is limited in most developing countries. The burden of Shiga toxin-associated HUS in India is unclear; school-going children show high prevalence of anti-factor H (FH) antibodies. The aim of the consensus meeting was to formulate guidelines for the diagnosis and management of HUS in children, specific to the needs of the country. METHODS: Four workgroups performed literature review and graded research studies addressing (i) investigations, biopsy, genetics, and differential diagnosis; (ii) Shiga toxin, pneumococcal, and infection-associated HUS; (iii) atypical HUS; and (iv) complement blockade. Consensus statements developed by the workgroups were discussed during a consensus meeting in March 2017. RESULTS: An algorithm for classification and evaluation was developed. The management of Shiga toxin-associated HUS is supportive; prompt plasma exchanges (PEX) is the chief therapy in patients with atypical HUS. Experts recommend that patients with anti-FH-associated HUS be managed with a combination of PEX and immunosuppressive medications. Indications for eculizumab include incomplete remission with plasma therapy, life-threatening features, complications of PEX or vascular access, inherited defects in complement regulation, and recurrence of HUS in allografts. Priorities for capacity building in regional and national laboratories are highlighted. CONCLUSIONS: Limited diagnostic capabilities and lack of access to eculizumab prevent the implementation of international guidelines for HUS in most developing countries. We propose practice guidelines for India, which will perhaps be applicable to other developing countries.


Assuntos
Conferências de Consenso como Assunto , Síndrome Hemolítico-Urêmica/diagnóstico , Nefrologia/normas , Guias de Prática Clínica como Assunto , Escherichia coli Shiga Toxigênica/imunologia , Consenso , Países em Desenvolvimento , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Índia , Nefrologia/métodos , Troca Plasmática , Escherichia coli Shiga Toxigênica/isolamento & purificação
4.
Medicina (B.Aires) ; 78(2): 107-112, abr. 2018. ilus
Artigo em Espanhol | LILACS | ID: biblio-954958

RESUMO

El síndrome urémico hemolítico (SUH) típico es una enfermedad huérfana causada por cepas de Escherichia coli productoras de toxina Shiga (Stx) y caracterizada por daño renal agudo, anemia hemolítica microangiopática y plaquetopenia. Es endémico en Argentina, el país con mayor incidencia de SUH en el mundo. Debido al rol fundamental de la Stx en su patogenia, se puede considerar que, como otras toxemias conocidas, el SUH podría ser tratado con anticuerpos. Este trabajo describe el desarrollo de un nuevo tratamiento capaz de neutralizar el efecto tóxico de distintas variantes de la Stx. El tratamiento consiste en fragmentos F(ab')2 provenientes de un antisuero equino cuya eficacia y potencia contra Stx1 y Stx2 se comprobó en diferentes modelos preclínicos. El producto mostró ser seguro en animales, presentó la farmacocinética descripta para compuestos similares y se pudo establecer una posible ventana terapéutica para su adecuada administración. En conjunto, los resultados preclínicos obtenidos validan la realización de un estudio clínico de primer uso en humanos. En dicho estudio, que se realizará en el Hospital Italiano de Buenos Aires, se analizará la seguridad y la farmacocinética del producto en voluntarios adultos sanos. Estos resultados sentarán las bases para la realización del estudio clínico fase II en pacientes pediátricos con infección por cepas de E. coli productoras de Stx.


The typical hemolytic uremic syndrome (HUS) is an orphan disease caused by Shiga toxin(Stx) -producing Escherichia coli strains and characterized by acute kidney damage, microangiopathic hemolytic anemia and low platelet count. It is endemic in Argentina, the country with the highest incidence of HUS in the world. Stx is essential for its development and therefore, HUS is considered a toxemic non-bacteremic disorder, which could be treated with antibodies. Herein we describe the development of a new treatment capable of neutralizing the toxic effect of Stx and its variants. The treatment consists of F(ab')2 fragments from an equine antiserum whose efficacy and potency against Stx1 and Stx2 were proved in different preclinical models. The product was shown to be safe in animals. Furthermore, the anti-Stx F(ab')2 pharmacokinetic was shown to be similar to that of analogous compounds and a therapeutic window for its administration was determined. Altogether, these preclinical results warrant testing in humans. The phase I clinical trial will be performed at the Hospital Italiano in Buenos Aires to evaluate the safety and pharmacokinetics of the product in healthy adult volunteers. Based on the results of this study, a phase II clinical trial will be planned in pediatric patients diagnosed with infection by Stx-producing E. coli strains.


Assuntos
Humanos , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Drogas em Investigação , Toxina Shiga I/antagonistas & inibidores , Toxina Shiga II/antagonistas & inibidores , Infecções por Escherichia coli/tratamento farmacológico , Síndrome Hemolítico-Urêmica/prevenção & controle , Argentina , Ensaios Clínicos Fase II como Assunto , Toxina Shiga I/imunologia , Toxina Shiga II/imunologia , Escherichia coli/isolamento & purificação , Escherichia coli/imunologia , Infecções por Escherichia coli/complicações , Síndrome Hemolítico-Urêmica/imunologia , Anticorpos/imunologia
5.
Pediatr Nephrol ; 33(11): 2057-2071, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29372302

RESUMO

Hemolytic uremic syndrome (HUS) is defined as a triad of noninmune microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. The most frequent presentation is secondary to Shiga toxin (Stx)-producing Escherichia coli (STEC) infections, which is termed postdiarrheal, epidemiologic or Stx-HUS, considering that Stx is the necessary etiological factor. After ingestion, STEC colonize the intestine and produce Stx, which translocates across the intestinal epithelium. Once Stx enters the bloodstream, it interacts with renal endothelial and epithelial cells, and leukocytes. This review summarizes the current evidence about the involvement of inflammatory components as central pathogenic factors that could determine outcome of STEC infections. Intestinal inflammation may favor epithelial leakage and subsequent passage of Stx to the systemic circulation. Vascular damage triggered by Stx promotes not only release of thrombin and increased fibrin concentration but also production of cytokines and chemokines by endothelial cells. Recent evidence from animal models and patients strongly indicate that several immune cells types may participate in HUS physiopathology: neutrophils, through release of proteases and reactive oxygen species (ROS); monocytes/macrophages through secretion of cytokines and chemokines. In addition, high levels of Bb factor and soluble C5b-9 (sC5b-9) in plasma as well as complement factors adhered to platelet-leukocyte complexes, microparticles and microvesicles, suggest activation of the alternative pathway of complement. Thus, acute immune response secondary to STEC infection, the Stx stimulatory effect on different immune cells, and inflammatory stimulus secondary to endothelial damage all together converge to define a strong inflammatory status that worsens Stx toxicity and disease.


Assuntos
Infecções por Escherichia coli/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Microvasos/patologia , Escherichia coli Shiga Toxigênica/imunologia , Animais , Via Alternativa do Complemento/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/patologia , Humanos , Mucosa Intestinal/microbiologia , Rim/irrigação sanguínea , Rim/imunologia , Rim/patologia , Microvasos/citologia , Microvasos/imunologia , Escherichia coli Shiga Toxigênica/isolamento & purificação
6.
Biochem J ; 473(21): 3791-3804, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27623774

RESUMO

Escherichia coli O157:H7 is a subtype of Shiga toxin-producing E. coli that is associated with haemorrhagic colitis and haemolytic uremic syndrome (HUS). Studies of populations in endemic areas have reported that the presence of specific antibodies against the O157 lipopolysaccharide (LPS) is associated with a lower incidence of diarrhoea and HUS. Phage display and IgG anti-O157 LPS antibodies were used in the present study to select peptide mimotopes of O157 LPS expressed in protein III of the M13 phage. Synthetic peptides (SP) were designed using the derived amino acid sequences obtained from DNA nucleotides of 63 selected phagotopes. The LxP/YP/SxL motif was identified in five of the phagotope amino acid sequences. Antibody responses against the phagotopes and their corresponding SPs were evaluated. SP12, one of the designed SP, induced the production of antibodies against the homologous peptide (1:800) and O157 LPS (1:200). The specificity of anti-SP12 antiserum was confirmed by analyzing its response to SP3, an SP with a different amino acid sequence than that of SP12, as well as against an E. coli LPS different from O157. Competitive studies with SP12 and O157 LPS showed a significant decrease in anti-SP12 and anti-LPS O157 antiserum responses against SP12 and O157 LPS, respectively. Eighteen (82%) of the 22 human serum samples with positive reactivity against E coli O157 LPS reacted with SP12 SP (cut-off >0.4). These results support the idea that SP12 is an immunogenic mimotope of O157 LPS.


Assuntos
Epitopos/química , Epitopos/imunologia , Escherichia coli O157/imunologia , Peptídeos/química , Peptídeos/imunologia , Animais , Anticorpos/imunologia , Escherichia coli O157/metabolismo , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/metabolismo , Humanos , Imunoglobulina G/imunologia , Lipopolissacarídeos/imunologia , Coelhos , Soro/imunologia
7.
Kidney Int ; 90(4): 774-82, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27378476

RESUMO

Hemolytic uremic syndrome caused by Shiga toxin-producing Escherichia coli (STEC HUS) is a worldwide endemic problem, and its pathophysiology is not fully elucidated. Here we tested whether the mannose-binding lectin (MBL2), an initiating factor of lectin complement pathway activation, plays a crucial role in STEC HUS. Using novel human MBL2-expressing mice (MBL2 KI) that lack murine Mbls (MBL2(+/+)Mbl1(-/-)Mbl2(-/-)), a novel STEC HUS model consisted of an intraperitoneal injection with Shiga toxin-2 (Stx-2) with or without anti-MBL2 antibody (3F8, intraperitoneal). Stx-2 induced weight loss, anemia, and thrombocytopenia and increased serum creatinine, free serum hemoglobin, and cystatin C levels, but a significantly decreased glomerular filtration rate compared with control/sham mice. Immunohistochemical staining revealed renal C3d deposition and fibrin deposition in glomeruli in Stx-2-injected mice. Treatment with 3F8 completely inhibited serum MBL2 levels and significantly attenuated Stx-2 induced-renal injury, free serum hemoglobin levels, renal C3d, and fibrin deposition and preserved the glomerular filtration rate. Thus, MBL2 inhibition significantly protected against complement activation and renal injury induced by Stx-2. This novel mouse model can be used to study the role of complement, particularly lectin pathway-mediated complement activation, in Stx-2-induced renal injury.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C3d/metabolismo , Infecções por Escherichia coli/tratamento farmacológico , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Imunoglobulina G/uso terapêutico , Lectina de Ligação a Manose/imunologia , Toxina Shiga II/toxicidade , Animais , Anticorpos Monoclonais Murinos , Ativação do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Técnicas de Introdução de Genes , Taxa de Filtração Glomerular , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Imuno-Histoquímica , Rim/imunologia , Masculino , Lectina de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Toxina Shiga II/imunologia , Escherichia coli Shiga Toxigênica/metabolismo
8.
J Immunol ; 197(4): 1276-86, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27421478

RESUMO

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.


Assuntos
Complemento C6/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Infecções por Escherichia coli/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Animais , Pré-Escolar , Complemento C6/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Modelos Animais de Doenças , Escherichia coli Êntero-Hemorrágica , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
9.
J Innate Immun ; 8(4): 400-11, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27230920

RESUMO

Hemolytic uremic syndrome (HUS), a vascular disease characterized by hemolytic anemia, thrombocytopenia, and acute renal failure, is caused by enterohemorrhagic Shiga toxin (Stx)-producing bacteria, which mainly affect children. Besides Stx, the inflammatory response mediated by neutrophils (PMN) is essential to HUS evolution. PMN can release neutrophil extracellular traps (NET) composed of DNA, histones, and other proteins. Since NET are involved in infectious and inflammatory diseases, the aim of this work was to investigate the contribution of NET to HUS. Plasma from HUS patients contained increased levels of circulating free-DNA and nucleosomes in comparison to plasma from healthy children. Neutrophils from HUS patients exhibited a greater capacity to undergo spontaneous NETosis. NET activated human glomerular endothelial cells, stimulating secretion of the proinflammatory cytokines IL-6 and IL-8. Stx induced PMN activation as judged by its ability to trigger reactive oxygen species production, increase CD11b and CD66b expression, and induce NETosis in PMN from healthy donors. During HUS, NET can contribute to the inflammatory response and thrombosis in the microvasculature and thus to renal failure. Intervention strategies to inhibit inflammatory mechanisms mediated by PMN, such as NETosis, could have a potential therapeutic impact towards amelioration of the severity of HUS.


Assuntos
Infecções Bacterianas/imunologia , Células Endoteliais/imunologia , Armadilhas Extracelulares/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Rim/patologia , Neutrófilos/imunologia , Toxina Shiga/imunologia , Injúria Renal Aguda , Anemia Hemolítica , Apoptose , Células Cultivadas , Criança , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Ativação de Neutrófilo , Neutrófilos/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Trombocitopenia
10.
Blood ; 126(18): 2085-90, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26396094

RESUMO

Hemolytic-uremic syndrome (HUS) is a thrombotic microangiopathy that is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and renal failure. Excess complement activation underlies atypical HUS and is evident in Shiga toxin-induced HUS (STEC-HUS). This Spotlight focuses on new knowledge of the role of Escherichia coli-derived toxins and polyphosphate in modulating complement and coagulation, and how they affect disease progression and response to treatment. Such new insights may impact on current and future choices of therapies for STEC-HUS.


Assuntos
Coagulação Sanguínea , Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Infecções por Escherichia coli/complicações , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/microbiologia , Escherichia coli Shiga Toxigênica/imunologia , Proteínas do Sistema Complemento/genética , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/genética , Humanos , Mutação , Polifosfatos/imunologia
11.
Hematol Oncol Clin North Am ; 29(3): 509-24, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26043389

RESUMO

The molecular linkage between ultralarge (UL) von Willebrand factor (VWF) multimers and the alternative complement pathway (AP) has recently been described. Endothelial cell (EC)-secreted and anchored ULVWF multimers (in long stringlike structures) function as both hyperadhesive sites that initiate platelet adhesion and aggregation and activating surfaces for the AP. In vitro, the active form of C3, C3b binds to the EC-anchored ULVWF multimeric strings and initiates the assembly on the strings of C3 convertase (C3bBb) and C5 convertase (C3bBbC3b). In vivo, activation of the AP via this mechanism proceeds all the way to generation of terminal complement complexes (C5b-9).


Assuntos
Via Alternativa do Complemento/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Ativação Plaquetária/imunologia , Agregação Plaquetária/imunologia , Púrpura Trombocitopênica Trombótica/imunologia , Fator de von Willebrand/imunologia , Complemento C3/imunologia , Complemento C3/metabolismo , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Síndrome Hemolítico-Urêmica/metabolismo , Humanos , Modelos Imunológicos , Púrpura Trombocitopênica Trombótica/metabolismo , Fator de von Willebrand/metabolismo
12.
Hematol Oncol Clin North Am ; 29(3): 525-39, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26043390

RESUMO

Shiga toxin associated hemolytic uremic syndrome (Stx HUS), a thrombotic microangiopathy, is the most common cause of pediatric acute kidney injury but has no direct treatment. A better understanding of disease pathogenesis may help identify new therapeutic targets. For this reason, the role of complement is being actively studied while eculizumab, the C5 monoclonal antibody, is being used to treat Stx HUS but with conflicting results. A randomized controlled trial would help properly evaluate its use in Stx HUS while more research is required to fully evaluate the role complement plays in the disease pathogenesis.


Assuntos
Injúria Renal Aguda/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Toxina Shiga/imunologia , Escherichia coli Shiga Toxigênica/imunologia , Microangiopatias Trombóticas/imunologia , Injúria Renal Aguda/etiologia , Criança , Via Alternativa do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Síndrome Hemolítico-Urêmica/complicações , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Modelos Imunológicos , Toxina Shiga/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Escherichia coli Shiga Toxigênica/fisiologia , Microangiopatias Trombóticas/complicações
13.
Transfusion ; 55(6 Pt 2): 1563-71, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25556575

RESUMO

BACKGROUND: Pneumococcal hemolytic uremic syndrome (P-HUS) is a rare but severe complication of invasive pneumococcal disease (IPD) in young children. Consensual biologic diagnosis criteria are currently lacking. STUDY DESIGN AND METHODS: A prospective study was conducted on 10 children with culture-confirmed IPD. Five presented with full-blown P-HUS, three had an incomplete form with hemolytic anemia and mild or no uremia (P-HA), and two had neither HUS nor HA. Thomsen-Friedenreich (T), Th, and Tk cryptantigens and sialic acid expression were determined on red blood cells (RBCs) with peanut (PNA), Glycine soja (SBA), Bandeiraea simplicifolia II, and Maackia amurensis lectins. Plasma concentrations of the major endogenous T-antigen-binding protein, galectin-3 (Gal-3), were analyzed. RESULTS: We found that RBCs strongly reacted with PNA and SBA lectins in all P-HUS and P-HA patients. Three P-HUS and three P-HA patients showed also concomitant Tk activation. Direct antiglobulin test (DAT) was positive in three P-HUS (one with anti-C3d and two with anti-IgG) and two P-HA patients (one with anti-C3d and one with anti-IgG). RBCs derived from the two uncomplicated IPD patients reacted with PNA but not with SBA lectin. Gal-3 plasma concentrations were increased in all P-HUS patients. CONCLUSIONS: The results indicate high levels of neuraminidase activity and desialylation in both P-HUS and P-HA patients. T-antigen activation is more sensitive than DAT for P-HUS diagnosis. Combining PNA and SBA lectins is needed to improve the specificity of T-antigen activation. High concentrations of Gal-3 in P-HUS patients suggest that Gal-3 may contribute to the pathogenesis of P-HUS.


Assuntos
Anemia Hemolítica/microbiologia , Antígenos Glicosídicos Associados a Tumores/metabolismo , Eritrócitos/metabolismo , Galectina 3/sangue , Síndrome Hemolítico-Urêmica/microbiologia , Infecções Pneumocócicas/complicações , Streptococcus pneumoniae/fisiologia , Anemia Hemolítica/sangue , Anemia Hemolítica/imunologia , Antígenos Glicosídicos Associados a Tumores/imunologia , Teste de Coombs , Eritrócitos/imunologia , Feminino , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Lactente , Masculino , Neuraminidase/metabolismo , Infecções Pneumocócicas/sangue , Infecções Pneumocócicas/imunologia , Estudos Retrospectivos
14.
Immunology ; 145(2): 258-67, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25580516

RESUMO

Enterohaemorrhagic Escherichia coli (EHEC) O157:H7 infection in humans can cause acute haemorrhagic colitis and severe haemolytic uraemic syndrome. The role of enterohaemolysin (Ehx) in the pathogenesis of O157:H7-mediated disease in humans remains undefined. Recent studies have revealed the importance of the inflammatory response in O157:H7 pathogenesis in humans. We previously reported that Ehx markedly induced interleukin-1ß (IL-1ß) production in human macrophages. Here, we investigated the disparity in Ehx-induced IL-1ß production between human and mouse macrophages and explored the underlying mechanism regarding the activation of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasomes. In contrast to the effects on human differentiated THP-1 cells and peripheral blood mononuclear cells, Ehx exerted no effect on IL-1ß production in mouse macrophages and splenocytes because of a disparity in pro-IL-1ß cleavage into mature IL-1ß upon caspase-1 activation. Additionally, Ehx significantly contributed to O157:H7-induced ATP release from THP-1 cells, which was not detected in mouse macrophages. Confocal microscopy demonstrated that Ehx was a key inducer of cathepsin B release in THP-1 cells but not in mouse IC-21 cells upon O157:H7 challenge. Inhibitor experiments indicated that O157:H7-induced IL-1ß production was largely dependent upon caspase-1 activation and partially dependent upon ATP signalling and cathepsin B release, which were both involved in NLRP3 activation. Moreover, inhibition of K(+) efflux drastically diminished O157:H7-induced IL-1ß production and cytotoxicity. The findings in this study may shed light on whether and how the Ehx contributes to the development of haemolytic uraemic syndrome in human O157:H7 infection.


Assuntos
Proteínas de Transporte/imunologia , Escherichia coli O157 , Proteínas de Escherichia coli/toxicidade , Proteínas Hemolisinas/toxicidade , Síndrome Hemolítico-Urêmica/imunologia , Interleucina-1beta/imunologia , Macrófagos/imunologia , Animais , Caspase 1/imunologia , Catepsina B/imunologia , Linhagem Celular Tumoral , Síndrome Hemolítico-Urêmica/patologia , Humanos , Inflamassomos/imunologia , Macrófagos/patologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Especificidade da Espécie
15.
Infect Immun ; 83(1): 286-91, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368111

RESUMO

Hemolytic-uremic syndrome (HUS), caused by Shiga toxin (Stx)-producing Escherichia coli (STEC), remains untreatable. Production of human monoclonal antibodies against Stx, which are highly effective in preventing Stx sequelae in animal models, is languishing due to cost and logistics. We reported previously that the production and evaluation of a camelid heavy-chain-only VH domain (VHH)-based neutralizing agent (VNA) targeting Stx1 and Stx2 (VNA-Stx) protected mice from Stx1 and Stx2 intoxication. Here we report that a single intramuscular (i.m.) injection of a nonreplicating adenovirus (Ad) vector carrying a secretory transgene of VNA-Stx (Ad/VNA-Stx) protected mice challenged with Stx2 and protected gnotobiotic piglets infected with STEC from fatal systemic intoxication. One i.m. dose of Ad/VNA-Stx prevented fatal central nervous system (CNS) symptoms in 9 of 10 animals when it was given to piglets 24 h after bacterial challenge and in 5 of 9 animals when it was given 48 h after bacterial challenge, just prior to the onset of CNS symptoms. All 6 placebo animals died or were euthanized with severe CNS symptoms. Ad/VNA-Stx treatment had no impact on diarrhea. In conclusion, Ad/VNA-Stx treatment is effective in protecting piglets from fatal Stx2-mediated CNS complications following STEC challenge. With a low production cost and further development, this could presumably be an effective treatment for patients with HUS and/or individuals at high risk of developing HUS due to exposure to STEC.


Assuntos
Adenovírus Humanos/genética , Anticorpos Neutralizantes/uso terapêutico , Infecções por Escherichia coli/tratamento farmacológico , Escherichia coli O157/imunologia , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Toxina Shiga I/antagonistas & inibidores , Toxina Shiga II/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/genética , Modelos Animais de Doenças , Portadores de Fármacos/administração & dosagem , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Escherichia coli O157/genética , Feminino , Vetores Genéticos , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/microbiologia , Injeções Intramusculares , Camundongos , Toxina Shiga I/imunologia , Toxina Shiga II/imunologia , Análise de Sobrevida , Suínos , Fatores de Tempo
16.
Nephron Clin Pract ; 128(3-4): 238-42, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25412932

RESUMO

The complement pathway is a central part of the innate immune system and also modulates adaptive immunity. It is implicated in the pathogenesis of glomerular disease by a number of clinical findings. These include the presence of complement components in renal biopsy samples, decreases in circulating levels indicating consumption, the presence of autoantibodies to complement proteins and the association of genetic mutations with disease either in individuals or within families. Further support and mechanistic insights comes from animal models. This review provides an overview of the role of complement in glomerular diseases and discusses the data from patients and animal models with reference to specific diseases. These include atypical haemolytic uraemic syndrome, C3 glomerulopathy, anti-neutrophil cytoplasmic antibody vasculitis, lupus nephritis and membranous nephropathy. The implications for therapy are also discussed.


Assuntos
Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Nefropatias/imunologia , Animais , Complexo Antígeno-Anticorpo/imunologia , Autoanticorpos/imunologia , Ativação do Complemento/genética , Proteínas do Sistema Complemento/genética , Glomerulonefrite Membranosa/tratamento farmacológico , Glomerulonefrite Membranosa/genética , Glomerulonefrite Membranosa/imunologia , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Nefropatias/tratamento farmacológico , Nefropatias/genética , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia
17.
Br J Haematol ; 166(5): 729-38, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24931452

RESUMO

Frequencies of human leucocyte antigens (HLA) were determined in 287 classic hairy cell leukaemia (HCL) patients. With respect to both population (n = 287) and allele (2n = 574) frequency respectively, the most common HLA class I and II antigens expressed were HLA-A*02 (49·1% and 28·6%), HLA-B*07 (21·3% and 11·1%), HLA-C*07 (46·7 and 28·2%), HLA-DQB1*03 (62·7% and 37·3%), HLA-DRB1*11 (30·0% and 16·0%) and HLA-DRB4*01 (45·3% and 29·6%). In comparing 6-14 databases of control Caucasians to 267 Caucasian HCL patients, only HLA-DRB1*11 was consistently over-represented in HCL, 31·1% of patients vs. 17-19·9% of controls (P = 0·0055 to <0·0001) and 16·5% of alleles vs. 6·5-12·3% of control alleles (P = 0·022 to <0·0001). HLA-DRB1*11 is a known risk factor for acquired thrombotic microangiopathy. Anti-CD22 recombinant immunotoxin BL22 in HCL was associated with a 12% incidence of completely reversible grade 3-4 haemolytic uraemic syndrome (HUS), mainly during the second or third retreatment cycle. Of 49 HCL patients receiving ≥2 cycles of BL22, 7 (14%) had HUS and HLA-DRB1*11 was expressed in 71% of 7 with HUS compared with only 21% of 42 without (P = 0·015). These data suggest that DBR1*11 may be a marker for increased susceptibility to HCL and, among HCL patients, could be a risk factor for BL22-induced HUS.


Assuntos
Cadeias HLA-DRB1/biossíntese , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/metabolismo , Leucemia de Células Pilosas/genética , Leucemia de Células Pilosas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Feminino , Frequência do Gene , Predisposição Genética para Doença , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/terapia , Humanos , Leucemia de Células Pilosas/imunologia , Leucemia de Células Pilosas/terapia , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Microangiopatias Trombóticas , Doadores de Tecidos , Adulto Jovem
18.
Thromb Res ; 133 Suppl 2: S117-21, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24862131

RESUMO

The complement system is important part of our innate immune system and interacts directly with the hemostatic system. Disorders of complement activation or dysregulation resulting in excess complement generation, such as Paroxysmal Nocturnal Hemoglobinuria (PNH), atypical Hemolytic uremic Syndrome (aHUS) and antiphospholipid syndrome (APLS) have been associated with significant thrombophilia. Terminal Complement (C5b-9) deposition on endothelial and tumor cell membranes has also been reported in a variety of cancer. Recent developments in complement inhibition have given us new insights into the mechanism of thrombosis in these disorders.


Assuntos
Proteínas do Sistema Complemento/imunologia , Hemoglobinúria Paroxística/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Hemostasia/imunologia , Neoplasias/imunologia , Trombose/imunologia , Hemoglobinúria Paroxística/sangue , Síndrome Hemolítico-Urêmica/sangue , Humanos , Neoplasias/sangue , Trombose/sangue
19.
Zhonghua Er Ke Za Zhi ; 52(3): 223-6, 2014 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-24824395

RESUMO

OBJECTIVE: To investigate the clinical characteristics, renal pathology, treatment and prognosis of children with atypical hemolytic uremic syndrome associated with H factor antibody. METHOD: Four children less than 18 yr of age admitted from Nov. 2010 to May 2011 in Peking University First Hospital were included. They all met the criteria for atypical hemolytic uremic syndrome and with positive serum anti factor H antibody. They aged from 5 to 11 yr. Data on clinical manifestations, renal pathology, treatment and prognosis were analyzed. RESULT: All of the 4 cases had gastrointestinal symptoms such as vomiting, abdominal pain, or abdominal distension. None of them had diarrhea. Two children had hypertension. One child had episodes of convulsion. One child had history of atypical hemolytic uremic syndrome. All of them had low serum complement C3. Three of them had low serum factor H (38.0, 88.4, 209.4 mg/L). All of them had serum antibody to factor H (1: 7 068, 1: 1 110, 1: 174, and 1: 869). Three of them received renal biopsy, all of them showed thrombotic microangiopathy. All of them were treated with steroid combined with mycophenolate mofetil. Two children received plasma exchange. They were followed up for 8 to 29 months. The renal function became normal and proteinuria relieved in all of them. The serum factor H concentration increased to 405.8, 155.8 and 438.4 mg/L, respectively. The titer of anti factor H antibody decreased to 1: 119, 1: 170, 1: 123, and 1: 674, respectively. CONCLUSION: Gastrointestinal symptom is common in children with atypical hemolytic uremic syndrome associated with H factor antibody. Hypocomplementemia was observed in all of them. Steroid combined with mycophenolate mofetil seemed to be effective for them. The monitoring of serum factor H and antibody to factor H may help diagnosis and treatment.


Assuntos
Autoanticorpos/imunologia , Fator H do Complemento/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Rim/patologia , Síndrome Hemolítico-Urêmica Atípica , Autoanticorpos/sangue , Criança , Pré-Escolar , Creatinina/sangue , Feminino , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Síndrome Hemolítico-Urêmica/patologia , Humanos , Rim/fisiopatologia , Testes de Função Renal , Masculino , Ácido Micofenólico/administração & dosagem , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapêutico , Troca Plasmática , Prednisolona/administração & dosagem , Prednisolona/uso terapêutico , Prognóstico , Estudos Retrospectivos
20.
J. bras. nefrol ; 36(2): 208-220, Apr-Jun/2014. tab, graf
Artigo em Português | LILACS | ID: lil-714671

RESUMO

O grupo de doenças que podem manifestar-se com MAT podem apresentar superposição clínica, dificultando o diagnóstico diferencial. Entre estas ressaltamos a PTT e SHU, sendo que esta última pode ocorrer pela ação de toxinas, doenças sistêmicas, hiperativação da via alterantiva descontrolada por alterações nas proteínas reguladoras desta via (SHUa) e por fim, idiopática. Deve-se proceder a uma série de exames para diferenciá-las. Ressaltando que SHUa é um diagnóstico de exclusão de outras causas de MAT. O tratamento da SHUa com infusão de plasma ou plasmaferese, resulta na maior parte dos casos com boa resposta, especialmente hematológica a curto-prazo, porém é uma doença grave e devastadora e, pode levar a óbito e doença renal crônica terminal. Tratamento com plasma apresenta grande recorrência da doença a longo-prazo e evolução renal desfavorável. Eculizumab, um anticorpo monoclonal anti-C5, tem surgido como uma esperança no prognóstico a curto e a longo-prazo nestes pacientes.


There is a group of diseases that may manifest with thrombotic microangiopathy and present clinical overlap. Among these we emphasize the thrombotic thrombocytopenic purpura and Hemolytic Uremic Syndrome, and the latter can occur by the action of toxins, systemic diseases, overactivation of the alternative complement system pathway, which can occur due to changes in regulatory proteins (atypical HUS) and finally, idiopathic. You must carry out a series of tests to differentiate them. aHUS is a diagnosis of exclusion of other causes of MAT. The treatment of aHUS with plasma therapy, results in most cases with good shortterm response, especially hematological; however, it is a progressive and devastating disease and can lead to death and terminal chronic renal disease. Treatment with plasma displays great recurrence of long-term disease and renal insufficiency. Eculizumab, a monoclonal antibody anti-C5, has been associated with hematological remission, benefits on renal function and no need of plasma therapy.


Assuntos
Humanos , Criança , Síndrome Hemolítico-Urêmica/diagnóstico , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/terapia , Púrpura Trombocitopênica Trombótica/diagnóstico , Ativação do Complemento , Diagnóstico Diferencial
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA