Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Clin Exp Nephrol ; 27(12): 1010-1020, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37634218

RESUMO

BACKGROUND: Thrombotic microangiopathy (TMA) after kidney transplantation (KTx), particularly early onset de novo (dn) TMA, requires immediate interventions to prevent irreversible organ damage. This multicenter study was performed to investigate the allogeneic clinical factors and complement genetic background of dnTMA after KTx. METHODS: Perioperative dnTMA after KTx within 1 week after KTx were diagnosed based on pathological or/and hematological criteria at each center, and their immunological backgrounds were researched. Twelve aHUS-related gene variants were examined in dnTMA cases. RESULTS: Seventeen recipients (15 donors) were enrolled, and all dnTMA cases were onset within 72-h of KTx, and 16 of 17 cases were ABO incompatible. The implementation rate of pre-transplant plasmaphereses therapies were low, including cases with high titers of anti-A/anti-B antibodies. Examination of aHUS-related gene variants revealed some deletions and variants with minor allele frequency (MAF) in Japan or East Asian genome databases in genes encoding alternative pathways and complement regulatory factors. These variants was positive in 8 cases, 6 of which were positive in both recipient and donor, but only in one graft loss case. CONCLUSIONS: Although some immunological risks were found for dnTMA after KTx, only a few cases developed into TMA. The characteristic variations revealed in the present study may be novel candidates related to dnTMA in Japanese or Asian patients, but not pathogenic variants of aHUS. Future studies on genetic and antigenic factors are needed to identify factors contributing to dnTMA after KTx.


Assuntos
Transplante de Rim , Microangiopatias Trombóticas , Humanos , Transplante de Rim/efeitos adversos , Doadores Vivos , População do Leste Asiático , Estudos Retrospectivos , Microangiopatias Trombóticas/etiologia , Microangiopatias Trombóticas/genética , Proteínas do Sistema Complemento/genética
2.
Am J Med Genet A ; 185(7): 2175-2179, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33884742

RESUMO

Transient receptor potential channel C6 encoded by TRPC6 is involved in slit diaphragm formation in podocytes, and abnormalities of the TRPC6 protein cause various glomerular diseases. The first identified pathogenic variant of TRPC6 was found to cause steroid-resistant nephrotic syndrome that typically developed in adulthood and then slowly led to end-stage renal disease, along with a renal pathology of focal segmental glomerulosclerosis. Here, we report a patient with rapidly progressing infantile nephrotic syndrome and a heterozygous missense TRPC6 variant. The patient, a 2-year-old Japanese boy, developed steroid-resistant nephrotic syndrome at age 11 months. His renal function deteriorated rapidly, and peritoneal dialysis was introduced at age 1 year and 6 months. His renal pathology, obtained at age 1 year and 1 month, was consistent with diffuse mesangial sclerosis (DMS). Clinical exome analysis and custom panel analysis for hereditary renal diseases revealed a reported heterozygous missense variant in TRPC6 (NM_004621.5:c.523C > T:p.Arg175Trp). This is the first report of a patient with a TRPC6-related renal disorder associated with DMS.


Assuntos
Nefropatias/genética , Síndrome Nefrótica/genética , Esclerose/genética , Canal de Cátion TRPC6/genética , Pré-Escolar , Exoma/genética , Predisposição Genética para Doença , Glomerulosclerose Segmentar e Focal/complicações , Glomerulosclerose Segmentar e Focal/diagnóstico por imagem , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Heterozigoto , Humanos , Lactente , Rim/diagnóstico por imagem , Rim/patologia , Nefropatias/complicações , Nefropatias/diagnóstico por imagem , Nefropatias/patologia , Masculino , Mutação de Sentido Incorreto/genética , Síndrome Nefrótica/complicações , Síndrome Nefrótica/diagnóstico por imagem , Síndrome Nefrótica/patologia , Podócitos/metabolismo , Podócitos/patologia , Esclerose/complicações , Esclerose/diagnóstico por imagem , Esclerose/patologia
3.
Pediatr Blood Cancer ; 67(12): e28733, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33001557

RESUMO

BACKGROUND: Accurate evaluation of kidney function before and after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is important for both informed decision making and detection of chronic kidney disease. However, to the best of our knowledge, no report has evaluated the glomerular filtration rate (GFR) in pediatric patients who underwent HSCT using the gold standard GFR measurement, as well as inulin-based GFR (iGFR). METHODS: We assessed iGFR before and after allo-HSCT to evaluate the impact of allo-HSCT on GFR in a prospective cohort study of 17 pediatric patients. We also assessed the accuracy and bias of the values of estimated GFR (eGFR) calculated using serum creatinine (Cr), cystatin C (CysC), beta-2 microglobulin (ß2 MG), 24-h creatinine clearance (24hCcr), and the full chronic kidney disease in children (CKiD) index that combines Cr, CysC, and blood urea nitrogen-based equations with iGFR as a reference to identify the most reliable equation for GFR. RESULTS: There was no significant difference between the values before and after allo-HSCT. CKiD CysC-, 24hCcr-, and full CKiD-based values showed good within 30% (P30) accuracy (80.6%, 79.3%, and 80.6%, respectively), but only 24hCcr and full CKiD had good mean bias (8.5% and 8.9%, respectively) and narrow 95% limits of agreement (-32.2 to 52.7 mL/min/1.73 m2 and -29.3 to 47.4 mL/min/1.73 m2 , respectively) compared with the corresponding iGFR. CONCLUSION: There was no significant impact of allo-HSCT on GFR in our cohort. The most reliable equations for pediatric patients with allo-HSCT were eGFR-24hCcr and eGFR-full CKiD.


Assuntos
Biomarcadores/análise , Taxa de Filtração Glomerular , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Inulina/análise , Rim/fisiopatologia , Adolescente , Criança , Pré-Escolar , Creatinina/sangue , Cistatina C/sangue , Feminino , Seguimentos , Neoplasias Hematológicas/patologia , Humanos , Testes de Função Renal , Masculino , Prognóstico , Estudos Prospectivos
4.
BMC Nephrol ; 21(1): 362, 2020 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-32838737

RESUMO

BACKGROUND: Mutations in the Wilms tumor 1 gene cause a spectrum of podocytopathy ranging from diffuse mesangial sclerosis to focal segmental glomerulosclerosis. In a considerable fraction of patients with Wilms tumor 1 mutations, the distinctive histology of immune-complex-type glomerulonephritis has been reported. However, the clinical relevance and etiologic mechanisms remain unknown. CASE PRESENTATION: A 5-year-old child presented with steroid-resistant nephrotic range proteinuria. Initial renal biopsy revealed predominant diffuse mesangial proliferation with a double-contour and coexisting milder changes of focal segmental glomerulosclerosis. Immunofluorescence and electron microscopy revealed a full-house-pattern deposition of immune complexes in the subendothelial and paramesangial areas. Serial biopsies at 6 and 8 years of age revealed that more remarkable changes of focal segmental glomerulosclerosis had developed on top of the initial proliferative glomerulonephritis. Identification of a de novo Wilms tumor 1 splice donor-site mutation in intron 9 (NM_024426.6:c.1447 + 4C > T) and 46,XY-gonadal dysgenesis led to the diagnosis of Frasier syndrome. CONCLUSIONS: Our findings, together with those of others, point to the importance of heterogeneity in clinicopathological phenotypes caused by Wilms tumor 1 mutations and suggest that immune-complex-mediated membranoproliferative glomerulopathy should be considered as a histological variant.


Assuntos
Complexo Antígeno-Anticorpo , Síndrome de Frasier/patologia , Glomerulonefrite Membranoproliferativa/patologia , Glomerulosclerose Segmentar e Focal/patologia , Rim/patologia , Criança , Pré-Escolar , Progressão da Doença , Síndrome de Frasier/genética , Humanos , Masculino , Proteínas WT1/genética
5.
Clin Exp Nephrol ; 23(1): 112-121, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30039480

RESUMO

BACKGROUND: In 2013, eculizumab was approved for treatment of the atypical hemolytic-uremic syndrome (aHUS) in Japan, which was defined as a thrombotic microangiopathy (TMA) excluding Shiga toxin-producing Escherichia coli-HUS and thrombotic thrombocytopenic purpura. Simultaneously, post-marketing surveillance was started to assess its safety and effectiveness. In 2016, Japanese clinical guide redefined terms to limit the use of "aHUS" to complement-mediated HUS only. Accordingly, TMA with other causes was defined as secondary TMA. Here we report the interim analysis of post-marketing surveillance of pediatric patients with aHUS and secondary TMA. METHODS: Pediatric patients treated with eculizumab from approval to 15 March 2017 were included in this observational real-world study. Clinical endpoints of effectiveness were TMA event-free status, complete TMA response, platelet count normalization, and improvement of estimated glomerular filtration rate (eGFR). Adverse reactions to eculizumab were also analyzed. RESULTS: In 27 pediatric patients with aHUS, median age at diagnosis was 4 years. Complement genes' variants were detected in 14 of 21 patients (66.7%). Median time from diagnosis to eculizumab initiation was 2.0 days. TMA event-free status, complete TMA response, platelet normalization, and improvement in eGFR were achieved in 85.2, 36.4, 78.3, and 75.0% of patients, respectively. Three patients with aHUS died. Twenty-four and 10 adverse reactions were reported in 31 aHUS patients and 17 secondary TMA patients, respectively; however, no eculizumab-related death or meningococcal infection was reported. CONCLUSIONS: This interim analysis confirmed that eculizumab is well-tolerated and effective for Japanese pediatric patients with aHUS in a real-world setting.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Inativadores do Complemento/efeitos adversos , Inativadores do Complemento/uso terapêutico , Adolescente , Síndrome Hemolítico-Urêmica Atípica/genética , Criança , Pré-Escolar , Complemento C5/antagonistas & inibidores , Proteínas do Sistema Complemento/genética , Feminino , Variação Genética/genética , Taxa de Filtração Glomerular , Humanos , Lactente , Recém-Nascido , Japão , Testes de Função Renal , Masculino , Segurança do Paciente , Contagem de Plaquetas , Vigilância de Produtos Comercializados , Resultado do Tratamento
6.
Clin Exp Nephrol ; 23(1): 65-75, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29959568

RESUMO

BACKGROUND: Eculizumab has been available for the treatment of atypical hemolytic-uremic syndrome (aHUS) in Japan since 2013. To assess safety and effectiveness of eculizumab in adult aHUS patients in the real-life setting, we performed interim analysis of a post-marketing surveillance mandated by Japanese regulations. METHODS: This study enrolled any patient who was diagnosed with TMA excluding Shiga toxin-producing Escherichia coli-HUS or thrombotic thrombocytopenic purpura based on Japanese clinical guide published in 2013 as inclusion criteria and treated with eculizumab. Although the term aHUS was redefined to denote only complement-mediated HUS in the guide revised in 2016, the patients with TMA caused by other causes (secondary TMA) were included. Patient outcomes and safety were evaluated at 6 months, 12 months, and annually thereafter. RESULTS: Thirty-three patients with aHUS and 27 patients with secondary TMA were enrolled. Median treatment duration of aHUS was 24weeks. Complement genes variants were detected in 11 of 18 patients with aHUS (61.1%). Among the 29 aHUS patients with available baseline data, platelet count (PLT), lactic dehydrogenase and serum creatinine (SCr) improved within 1-month after eculizumab initiation. TMA event-free status, complete TMA response, PLT normalization, and SCr decrease were achieved in 67.9% (19/28), 27.8% (5/18), 56.5% (13/23), and 57.1% (16/28) of patients, respectively. Thirty-three and 11 adverse reactions were observed in patients with aHUS (13/33 patients) and secondary TMA (6/27 patients), respectively. CONCLUSIONS: This interim analysis confirmed the acceptable safety profile and effectiveness of eculizumab for Japanese adult aHUS patients in real-world settings.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Inativadores do Complemento/efeitos adversos , Inativadores do Complemento/uso terapêutico , Adulto , Idoso , Síndrome Hemolítico-Urêmica Atípica/genética , Complemento C5/antagonistas & inibidores , Proteínas do Sistema Complemento/genética , Feminino , Variação Genética/genética , Humanos , Japão , Testes de Função Renal , Masculino , Pessoa de Meia-Idade , Segurança do Paciente , Vigilância de Produtos Comercializados , Resultado do Tratamento , Adulto Jovem
7.
BMC Infect Dis ; 18(1): 128, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29534676

RESUMO

BACKGROUND: Influenza virus transmission may be prevented by infection control measures, including vaccination, wearing a mask, gargling with water, and hand washing. It is unclear, however, whether these measures affect influenza epidemics in school settings. METHODS: A prospective epidemiological survey in all public elementary schools in Matsumoto City, Japan, during the 2014/2015 season evaluated the number of diagnosed patients in each school and calculated the reproduction number of schoolchildren. At the end of the prospective survey, a cross-sectional survey evaluated the implementation of infection control measures in these schools. Both results were combined and associations among infection control measures including vaccination, mask wearing, hand washing, water gargling, and epidemic level were evaluated. RESULTS: Of the 13,217 schoolchildren in 29 schools, 2548 were diagnosed with seasonal influenza. A significant negative association was observed between vaccination coverage and reproduction number at each school, but not between other infection control measures and the reproduction number. A regression curve with exponential function was most predictive. At 0% vaccination, the reproduction number was estimated to be 1.39. CONCLUSION: These findings provide evidence that high vaccination coverage was associated with reduced epidemic levels in schools and suggest the need for increased vaccination of schoolchildren.


Assuntos
Epidemias , Vacinas contra Influenza/imunologia , Influenza Humana/epidemiologia , Cobertura Vacinal , Criança , Cidades , Feminino , Humanos , Controle de Infecções , Influenza Humana/diagnóstico , Influenza Humana/prevenção & controle , Japão/epidemiologia , Modelos Lineares , Masculino , Estudos Prospectivos , Instituições Acadêmicas , Estações do Ano , Inquéritos e Questionários
9.
Biochim Biophys Acta Gen Subj ; 1861(2): 1-14, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27864148

RESUMO

BACKGROUND: Pentraxins (PTXs) are a superfamily of multifunctional conserved proteins involved in acute-phase responses. Recently, we have shown that collectin placenta 1 (CL-P1) and C-reactive protein (CRP) mediated complement activation and failed to form terminal complement complex (TCC) in normal serum conditions because of complement factor H inhibition. METHODS: We used CL-P1 expressing CHO/ldlA7 cells to study the interaction with PTXs. Soluble type CL-P1 was used in an ELISA assay for the binding, C3 and TCC deposition experiments. Furthermore, we used our previously established CL-P1 expressing HEK293 cells for the C3 fragment and TCC deposition assay. RESULTS: We demonstrated that CL-P1 also bound serum amyloid p component (SAP) and pentraxin 3 (PTX3) to activate the classical pathway and the alternative pathway using factor B. CRP and PTX3 further amplified complement deposition by properdin. We found that CRP and PTX3 recruit CFH, whereas SAP recruits C4 binding protein on CL-P1 expressing cell surfaces to prevent the formation of TCC in normal serum conditions. In addition, depletion of CFH, C4BP and complement factor I (CFI) failed to prevent TCC formation both in ELISA and cell experiments. Furthermore, soluble complement receptor 1, an inhibitor of all complement pathways prevents PTX induced TCC formation. CONCLUSION: Our current study hypothesizes that the interaction of pentraxins with CL-P1 is involved in complement activation. GENERAL SIGNIFICANCE: CL-P1 might generally inhibit PTX induced complement activation and host damage to protect self-tissues.


Assuntos
Proteína C-Reativa/metabolismo , Colectinas/metabolismo , Ativação do Complemento/fisiologia , Componente Amiloide P Sérico/metabolismo , Reação de Fase Aguda/metabolismo , Animais , Células CHO , Linhagem Celular , Fator H do Complemento/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/fisiologia , Cricetulus , Células HEK293 , Humanos , Ligação Proteica/fisiologia , Transdução de Sinais/fisiologia
10.
Clin Exp Nephrol ; 20(2): 265-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26156042

RESUMO

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is a severe life-threatening disease with frequent progression to end-stage renal disease (ESRD). Eculizumab, a humanized anti-C5 monoclonal antibody targeting the activated complement pathway, has recently been introduced as a novel therapy against aHUS. We, therefore, investigated the efficacy and safety of eculizumab in Japanese pediatric patients. METHODS: We retrospectively analyzed clinical course and laboratory data of the first ten children with aHUS treated with eculizumab nationwide. RESULTS: Seven patients were resistant to plasma therapy and three were dependent on it. Causative gene mutations were found in five patients. Two patients had anti-complement factor H autoantibody. Three patients had a family history of thrombotic microangiopathy (TMA). After initiation of eculizumab, all patients immediately achieved hematological remission and could successfully discontinue plasma therapy. The median periods to normalization of platelet count, lactate dehydrogenase levels and disappearance of schistocytes were 5.5, 17 and 12 days, respectively. Nine patients recovered their renal function and the median period to terminate renal replacement therapy (RRT) was 3 days. However, two patients progressed to ESRD and required chronic RRT at the last observation. No patients had a relapse of TMA under regular eculizumab therapy. No serious adverse events occurred during the follow-up period. CONCLUSIONS: Eculizumab is efficacious and well-tolerated therapy for children with aHUS. Although pathogenic mutations could not be detected in five patients, all patients showed immediate normalization of hematological abnormalities, strongly suggesting complement-related aHUS. This prompt hematological amelioration can become an indicator for therapeutic efficacy of eculizumab. However, appropriate indications and optimal duration of the treatment remain unclear.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Estudos Retrospectivos
11.
Clin Exp Nephrol ; 20(4): 536-543, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27422619

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a rare disease characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. In 2013, we developed diagnostic criteria to enable early diagnosis and timely initiation of appropriate treatment for aHUS. Recent clinical and molecular findings have resulted in several proposed classifications and definitions of thrombotic microangiopathy and aHUS. Based on recent advances in this field and the emerging international consensus to exclude secondary TMAs from the definition of aHUS, we have redefined aHUS and proposed diagnostic algorithms, differential diagnosis, and therapeutic strategies for aHUS.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/etiologia , Síndrome Hemolítico-Urêmica Atípica/terapia , Humanos , Japão
12.
Pediatr Int ; 58(7): 549-55, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27460397

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a rare disease characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. In 2013, we developed diagnostic criteria to enable early diagnosis and timely initiation of appropriate treatment for aHUS. Recent clinical and molecular findings have resulted in several proposed classifications and definitions of thrombotic microangiopathy and aHUS. Based on recent advances in this field and the emerging international consensus to exclude secondary TMAs from the definition of aHUS, we have redefined aHUS and proposed diagnostic algorithms, differential diagnosis, and therapeutic strategies for aHUS.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Diagnóstico Precoce , Guias de Prática Clínica como Assunto , Síndrome Hemolítico-Urêmica Atípica/epidemiologia , Humanos , Incidência , Japão/epidemiologia
13.
Am J Med Genet A ; 167A(3): 592-601, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25691411

RESUMO

6p duplication syndrome is a rare chromosomal disorder that frequently manifests renal complications, including proteinuria, hypoplastic kidney, and hydronephrosis. We report a girl with the syndrome, manifesting left hydronephrosis, proteinuria/hematuria, and focal segmental glomerular sclerosis (FSGS) resulting in chronic end-stage renal failure, successfully treated with renal transplantation. Microarray comparative genomic hybridization showed the derivative chromosome 6 to have a 6.4-Mb duplication at 6p25.3-p25.1 with 32 protein-coding genes and a 220-Kb deletion at 6p25.3 with two genes of no possible relation to the renal pathology. Review of the literature shows that variation of renal complications in the syndrome is compatible with congenital anomalies of the kidney and urinary tract (CAKUT). FSGS, observed in another patient with 6p duplication syndrome, could be a non-coincidental complication. FOXC1, located within the 6.4-Mb duplicated region at 6p25.3-p25.2, could be a candidate gene for CAKUT, but its single gene duplication effect would not be sufficient. FSGS would be a primary defect associated with duplicated gene(s) albeit no candidate could be proposed, or might occur in association with CAKUT.


Assuntos
Glomerulosclerose Segmentar e Focal/genética , Hidronefrose/genética , Proteinúria/genética , Trissomia , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Biópsia , Criança , Bandeamento Cromossômico , Cromossomos Humanos Par 6 , Hibridização Genômica Comparativa , Fácies , Feminino , Estudo de Associação Genômica Ampla , Glomerulosclerose Segmentar e Focal/diagnóstico , Humanos , Hidronefrose/diagnóstico , Rim/anormalidades , Rim/patologia , Proteinúria/diagnóstico , Síndrome , Ultrassonografia , Sistema Urinário/anormalidades
15.
Transpl Int ; 28(5): 565-74, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25644234

RESUMO

Pericardial effusion is a potentially fatal complication following hematopoietic stem cell transplantation (HSCT). Therefore, the identification of risk factors could improve the outcome. Prolonged QT dispersion (QTD) and corrected QTD (QTcD) are associated with serious arrhythmias and sudden death in many forms of heart disease. However, no study has evaluated the efficacy of QTD and QTcD to predict pericardial effusion post-HSCT. We studied 89 pediatric HSCT patients to identify preoperative risk factors for pericardial effusion with particular focus on QTD and QTcD. Pericardial effusion occurred in 15 patients (cumulative onset rate: 17.4%) within one year post-HSCT, of which 8 (9.2%) were symptomatic. Patients with pericardial effusion following allogeneic HSCT showed significantly lower overall survival; however, pericardial effusion was not the direct cause of death in any patient. Univariate and multivariate analyses revealed that transplantation-associated thrombotic microangiopathy (TA-TMA) was an independent risk factor for post-HSCT pericardial effusion. In addition, pretransplant QTcD was significantly prolonged in the pericardial effusion group. These results suggest that pediatric patients with abnormally prolonged QTcD before the preparative regimen for HSCT should be regularly followed-up by echocardiography to detect pericardial effusion, particularly when accompanied by complications including TA-TMA.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Derrame Pericárdico/patologia , Microangiopatias Trombóticas/patologia , Adolescente , Arritmias Cardíacas , Síndrome de Brugada , Doença do Sistema de Condução Cardíaco , Tamponamento Cardíaco , Criança , Pré-Escolar , Ecocardiografia , Feminino , Sistema de Condução Cardíaco/anormalidades , Células-Tronco Hematopoéticas/citologia , Humanos , Masculino , Derrame Pericárdico/complicações , Pericardiocentese , Curva ROC , Estudos Retrospectivos , Fatores de Risco , Microangiopatias Trombóticas/complicações , Resultado do Tratamento
16.
Clin Exp Nephrol ; 18(1): 4-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24343712

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is rare and comprises the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. Recently, abnormalities in the mechanisms underlying complement regulation have been focused upon as causes of aHUS. The prognosis for patients who present with aHUS is very poor, with the first aHUS attack being associated with a mortality rate of ~25 %, and with ~50 % of cases resulting in end-stage renal disease requiring dialysis. If treatment is delayed, there is a high risk of this syndrome progressing to renal failure. Therefore, we have developed diagnostic criteria for aHUS to enable its early diagnosis and to facilitate the timely initiation of appropriate treatment. We hope these diagnostic criteria will be disseminated to as many clinicians as possible and that they will be used widely.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Nefrologia/normas , Pediatria/normas , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/etiologia , Síndrome Hemolítico-Urêmica Atípica/imunologia , Síndrome Hemolítico-Urêmica Atípica/terapia , Biomarcadores/sangue , Ativação do Complemento , Proteínas do Sistema Complemento , Consenso , Diagnóstico Precoce , Humanos , Contagem de Plaquetas/normas , Valor Preditivo dos Testes , Prognóstico , Fatores de Risco
17.
Pediatr Int ; 56(3): 429-32, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24894932

RESUMO

Fulminant Wilson's disease (WD) is life-threatening. The revised WD prognostic index (RWPI) has been used to predict the severity of the disease, with a score ≥11 indicating fatal outcome without liver transplantation (LTx). We here report the case of a 10-year-old female patient with fulminant WD (RWPI, 16) who recovered fully after plasma exchange and continuous hemodiafiltration, followed by treatment with copper chelate agents. To the best of our knowledge, there have been five fulminant WD patients with RWPI ≥ 11 including the present patient, in whom LTx was not done. Based on the therapeutic modalities in these five cases, non-surgical treatment (blood purification and copper chelate agents) may be able to avoid LTx in fulminant WD even with very high RWPI, although preparation for LTx is necessary.


Assuntos
Degeneração Hepatolenticular/terapia , Criança , Feminino , Hemodiluição , Humanos , Troca Plasmática
18.
Pediatr Int ; 56(1): 1-5, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24548192

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is rare and comprises the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. Recently, abnormalities in the mechanisms underlying complement regulation have been focused upon as causes of aHUS. The prognosis for patients who present with aHUS is very poor, with the first aHUS attack being associated with a mortality rate of approximately 25%, and with approximately 50% of cases resulting in end-stage renal disease requiring dialysis. If treatment is delayed, there is a high risk of this syndrome progressing to renal failure. Therefore, we have developed diagnostic criteria for aHUS to enable its early diagnosis and to facilitate the timely initiation of appropriate treatment. We hope these diagnostic criteria will be disseminated to as many clinicians as possible and that they will be used widely.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Técnicas de Diagnóstico Urológico/normas , Guias como Assunto , Nefrologia , Sociedades Médicas , Criança , Humanos , Japão
19.
Front Immunol ; 15: 1360855, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38524137

RESUMO

Mutations in the complement factor H (CFH) gene are associated with complement dysregulation and the development of atypical hemolytic uremic syndrome (aHUS). Several fusion genes that result from genomic structural variation in the CFH and complement factor H-related (CFHR) gene regions have been identified in aHUS. However, one allele has both CFHR gene duplication and CFH::CFHR1 fusion gene have not been reported. An 8-month-old girl (proband) presented with aHUS and was treated with ravulizumab. Her paternal grandfather developed aHUS previously and her paternal great grandmother presented with anti-neutrophil cytoplasmic antibody-associated vasculitis and thrombotic microangiopathy (TMA). However, the proband's parents have no history of TMA. A genetic analysis revealed the presence of CFH::CFHR1 fusion gene and a CFHR3-1-4-2 gene duplication in the patient, her father, and her paternal grandfather. Although several fusion genes resulting from structural variations of the CFH-CFHR genes region have been identified, this is the first report of the combination of a CFH::CFHR1 fusion gene with CFHR gene duplication. Because the CFH-CFHR region is highly homologous, we hypothesized that CFHR gene duplication occurred. These findings indicate a novel pathogenic genomic structural variation associated with the development of aHUS.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Fator H do Complemento , Humanos , Feminino , Lactente , Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/genética , Duplicação Gênica , Proteínas do Sistema Complemento/genética , Mutação , Proteínas Sanguíneas/genética , Proteínas Inativadoras do Complemento C3b/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA