Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Kidney Dis ; 72(1): 84-92, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29429752

RESUMO

BACKGROUND: Cases reports and small series of patients with C3 glomerulopathy have reported variable efficacy of eculizumab. STUDY DESIGN: Case series of C3 glomerulopathy. SETTING & PARTICIPANTS: Pediatric and adult patients with C3 glomerulopathy treated with eculizumab between 2010 and 2016 were identified through the C3 glomerulopathy French registry database, and a questionnaire was sent to participating French pediatric and adult nephrology centers, as well as one pediatric referral center in Québec, Canada. OUTCOMES: Global or partial clinical renal response. MEASUREMENTS: Evolution of serum creatinine and proteinuria values. RESULTS: 26 patients (13 children/adolescents) were included. 22 (85%) patients had received steroids, plasma exchange, or immunosuppressive therapy before eculizumab, and 3 of them had rapid progression of their kidney disease despite treatment. At the initiation of eculizumab therapy, 11 (42%) patients had chronic kidney disease, 7 (27%) had rapidly progressive disease, and 3 (12%) required dialysis. After eculizumab treatment (median duration, 14 months), 6 (23%) patients had a global clinical response; 6 (23%), a partial clinical response; and 14 (54%), no response. Compared with those who had a partial clinical or no response, patients who had a global clinical response had lower estimated glomerular filtration rates, a more rapidly progressive course, and more extracapillary proliferation on kidney biopsy. Age, extent of renal fibrosis, frequency of nephrotic syndrome, low serum C3 and C3 nephritic factor and elevated soluble C5b-9 concentrations, or complement gene variants did not differ between responders and nonresponders. LIMITATIONS: Retrospective design without a control group, relatively small number of cases, inclusion of pediatric and adult cases. CONCLUSIONS: Eculizumab appears to be a potential treatment for patients with crescentic rapidly progressive C3 glomerulopathy. Its benefit in patients with non-rapidly progressing forms seems to be limited.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Complemento C3/metabolismo , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Adolescente , Adulto , Idoso , Criança , Feminino , Seguimentos , Glomerulonefrite Membranoproliferativa/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
2.
Lancet ; 390(10095): 681-696, 2017 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-28242109

RESUMO

Haemolytic uraemic syndrome is a form of thrombotic microangiopathy affecting predominantly the kidney and characterised by a triad of thrombocytopenia, mechanical haemolytic anaemia, and acute kidney injury. The term encompasses several disorders: shiga toxin-induced and pneumococcus-induced haemolytic uraemic syndrome, haemolytic uraemic syndrome associated with complement dysregulation or mutation of diacylglycerol kinase ɛ, haemolytic uraemic syndrome related to cobalamin C defect, and haemolytic uraemic syndrome secondary to a heterogeneous group of causes (infections, drugs, cancer, and systemic diseases). In the past two decades, experimental, genetic, and clinical studies have helped to decipher the pathophysiology of these various forms of haemolytic uraemic syndrome and undoubtedly improved diagnostic approaches. Moreover, a specific mechanism-based treatment has been made available for patients affected by atypical haemolytic uraemic syndrome due to complement dysregulation. Such treatment is, however, still absent for several other disease types, including shiga toxin-induced haemolytic uraemic syndrome.


Assuntos
Síndrome Hemolítico-Urêmica/terapia , Antibacterianos/uso terapêutico , Produtos Biológicos/uso terapêutico , Criança , Feminino , Síndrome Hemolítico-Urêmica/diagnóstico , Síndrome Hemolítico-Urêmica/etiologia , Humanos , Transplante de Rim , Gravidez , Complicações na Gravidez/diagnóstico , Complicações na Gravidez/etiologia , Complicações na Gravidez/terapia , Recidiva , Resultado do Tratamento
3.
Hum Mutat ; 36(8): 743-52, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25907713

RESUMO

Dent disease is a rare X-linked tubulopathy characterized by low molecular weight proteinuria, hypercalciuria, nephrocalcinosis and/or nephrolithiasis, progressive renal failure, and variable manifestations of other proximal tubule dysfunctions. It often progresses over a few decades to chronic renal insufficiency, and therefore molecular characterization is important to allow appropriate genetic counseling. Two genetic subtypes have been described to date: Dent disease 1 is caused by mutations of the CLCN5 gene, coding for the chloride/proton exchanger ClC-5; and Dent disease 2 by mutations of the OCRL gene, coding for the inositol polyphosphate 5-phosphatase OCRL-1. Herein, we review previously reported mutations (n = 192) and their associated phenotype in 377 male patients with Dent disease 1 and describe phenotype and novel (n = 42) and recurrent mutations (n = 24) in a large cohort of 117 Dent disease 1 patients belonging to 90 families. The novel missense and in-frame mutations described were mapped onto a three-dimensional homology model of the ClC-5 protein. This analysis suggests that these mutations affect the dimerization process, helix stability, or transport. The phenotype of our cohort patients supports and extends the phenotype that has been reported in smaller studies.


Assuntos
Canais de Cloreto/genética , Doença de Dent/genética , Mutação , Animais , Canais de Cloreto/química , Canais de Cloreto/metabolismo , Estudos de Coortes , Doença de Dent/metabolismo , Estudos de Associação Genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Linhagem
4.
Mol Genet Metab ; 111(3): 314-320, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24440466

RESUMO

OBJECTIVE: To establish the safety and efficacy of a new gel formulation of cysteamine hydrochloride (CH) eye drops, for the treatment of corneal complications of nephropathic cystinosis. DESIGN: Open label dose response clinical trial. PARTICIPANTS: Eight patients with infantile nephropathic cystinosis including 4 children, 3 adolescents, and 1 adult (mean age at inclusion, 12.1 ± 4.6 years) treated with CH 0.1% eye drops. INTERVENTION: Patients were treated, in both eyes, with the control CH 0.1% eye drop formulation on average 4 times daily for one month and then switched to Cystadrops® at the same dose frequency. Based on clinical ocular findings, the dose regimen was adapted at D30 and D90 in order to decrease the frequency of instillation. After D90, this dose frequency was maintained, except in cases of crystal density worsening. Patients had a follow-up visit every 6 months during 48 months. MAIN OUTCOME MEASURES: Safety assessment consisted of adverse event and serious adverse event monitoring and recording at each visit. For the efficacy study, the primary endpoint was the corneal cystine crystal density measured with an in vivo confocal microscopy (IVCM) score. RESULTS: All patients completed the study. During the 4-year study period, neither serious adverse events nor significant adverse events related to the study drug were reported. After switching to Cystadrops®, the IVCM total score decreased from baseline to D90 by a mean of 28.6 ± 17.5% (p<0.001). From D90 to M48, the IVCM total score remained stable and significantly decreased as compared to that at D1 despite a reduced dose regimen from D90. At M48, the mean IVCM total score was 8.13 ± 4.15, decreased by a mean 29.9 ± 26.29% from D1 (p = 0.001), with a reduced number of instillations compared to that at D1. The IVCM total score and photophobia were significantly correlated (p = 0.04). CONCLUSION: This study provides evidence that Cystadrops® gel is superior to the CH 0.1% formulation in terms of efficacy and has a good safety profile over a long follow-up period.


Assuntos
Cisteamina/administração & dosagem , Cistina/metabolismo , Cistinose/tratamento farmacológico , Administração Tópica , Adolescente , Adulto , Criança , Córnea/patologia , Cristalização , Cistina/química , Cistinose/metabolismo , Cistinose/patologia , Feminino , Géis/administração & dosagem , Humanos , Masculino , Soluções Oftálmicas/administração & dosagem , Adulto Jovem
5.
Am J Kidney Dis ; 63(1): 40-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24021908

RESUMO

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is a devastating form of renal thrombotic microangiopathy. Despite plasma exchange, the standard treatment of aHUS for decades, the renal prognosis for patients with aHUS has remained poor. We assessed the off-trial use of eculizumab in adult patients with aHUS affecting the native kidneys. STUDY DESIGN: A retrospective study was conducted. aHUS was defined as the presence of 3 or more of the following: acute kidney injury (serum creatinine >1.4 mg/dL [120 µmol/L]), mechanical hemolytic anemia, thrombocytopenia, and the presence of thrombotic microangiopathy features in a kidney biopsy specimen. Patients who had received 4 or more weekly 900-mg infusions of eculizumab were included. SETTING & PARTICIPANTS: 19 patients were identified through a query sent to all French nephrology centers. OUTCOMES & MEASUREMENTS: Evolution of kidney function, hemolysis, and thrombocytopenia after the initiation of eculizumab therapy. RESULTS: All patients had acute kidney injury (serum creatinine range, 2.2-17.0 mg/dL) and 12 required hemodialysis. Thirteen patients carried a mutation in 1 complement gene and 1 had anti-factor H antibodies. For first-line therapy, 16 patients underwent plasma exchange and 3 patients received eculizumab. Median time between aHUS onset and eculizumab therapy initiation was 6 (range, 1-60) days and median time to platelet count normalization after eculizumab therapy initiation was 6 (range, 2-42) days. At the 3-month follow-up, 4 patients still required dialysis, 8 had non-dialysis-dependent chronic kidney disease, and 7 had normalized kidney function. At last follow-up (range, 4-22 months), 3 patients remained dialysis dependent, 7 had non-dialysis-dependent chronic kidney disease (estimated glomerular filtration rate, 17-55 mL/min/1.73 m(2)), and 9 had normal kidney function. Risks of reaching end-stage renal disease within 3 months and 1 year of aHUS onset were reduced by half in eculizumab-treated patients compared with recent historical controls. LIMITATIONS: Retrospective study and use of historical controls. CONCLUSIONS: Our data indicate that eculizumab improves kidney disease outcome in patients with aHUS.


Assuntos
Injúria Renal Aguda , Anticorpos Monoclonais Humanizados , Síndrome Hemolítico-Urêmica , Falência Renal Crônica , Rim , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Adulto , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Síndrome Hemolítico-Urêmica Atípica , Biópsia/métodos , Biópsia/estatística & dados numéricos , Creatinina/sangue , Monitoramento de Medicamentos/métodos , Feminino , França , Síndrome Hemolítico-Urêmica/complicações , Síndrome Hemolítico-Urêmica/diagnóstico , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/fisiopatologia , Humanos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/efeitos adversos , Rim/patologia , Rim/fisiopatologia , Falência Renal Crônica/epidemiologia , Falência Renal Crônica/etiologia , Testes de Função Renal/métodos , Masculino , Contagem de Plaquetas/métodos , Diálise Renal/estatística & dados numéricos , Estudos Retrospectivos , Medição de Risco , Resultado do Tratamento
6.
Presse Med ; 41(3 Pt 2): e115-35, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22284541

RESUMO

2011 has been a special year for hemolytic uremic syndrome (HUS): on the one hand, the dramatic epidemic of Shiga toxin producing E. coli -associated HUS in Germany brought the disease to the attention of the general population, on the other hand it has been the year when eculizumab, the first complement blocker available for clinical practice, was demonstrated as the potential new standard of care for atypical HUS. Here we review the therapeutic options presently available for the various forms of hemolytic uremic syndrome and show how recent knowledge has changed the therapeutic approach and prognosis of atypical HUS.


Assuntos
Síndrome Hemolítico-Urêmica/terapia , Escherichia coli Shiga Toxigênica , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/terapia , Algoritmos , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/terapia , Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica , Volume Sanguíneo , Criança , Diuréticos/uso terapêutico , Fibrinolíticos/uso terapêutico , Alemanha/epidemiologia , Síndrome Hemolítico-Urêmica/diagnóstico , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/etiologia , Homocistinúria/complicações , Homocistinúria/diagnóstico , Homocistinúria/terapia , Humanos , Transplante de Rim , Plasma/imunologia , Troca Plasmática/métodos , Plasmaferese , Infecções Pneumocócicas/complicações , Prognóstico , Diálise Renal , Toxinas Shiga/imunologia , Toxinas Shiga/toxicidade , Streptococcus pneumoniae , Deficiência de Vitamina B 12/congênito , Adulto Jovem
7.
Pediatr Nephrol ; 26(3): 419-24, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21188423

RESUMO

The development of membranoproliferative glomerulonephritis (MPGN) is associated with uncontrolled activation of the complement alternative pathway. This dysregulation is related either to C3 nephritic factor (C3NeF), an auto-antibody directed against the alternative C3 convertase, or to homozygous loss-of-function mutation of the complement regulatory protein factor H. Heterozygous mutations in the genes coding for factor H, or for the other alternative pathway inhibitory proteins factor I and membrane cofactor protein, have recently been identified in a small number of patients with MPGN with exclusive C3 deposits. We report three hypocomplementemic children with dense deposit disease (n=1) or immune-complex-mediated MPGN type I (n=2), associated with both C3NeF activity and heterozygous mutation of factor H or factor I. These observations highlight the possible combination of genetic and acquired defect in complement control in various subtypes of MPGN, a finding that may influence the treatment strategy in some patients.


Assuntos
Fator Nefrítico do Complemento 3/análise , Fator I do Complemento/genética , Glomerulonefrite Membranoproliferativa/genética , Glomerulonefrite Membranoproliferativa/imunologia , Rim/imunologia , Adolescente , Biópsia , Criança , Pré-Escolar , Fator H do Complemento/genética , Predisposição Genética para Doença , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Glomerulonefrite Membranoproliferativa/patologia , Heterozigoto , Humanos , Imunossupressores/uso terapêutico , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Mutação , Fenótipo , Fatores de Tempo , Resultado do Tratamento
8.
J Am Soc Nephrol ; 21(12): 2180-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21051740

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a rare form of thrombotic microangiopathy that associates, in 70% of cases, with genetic or acquired disorders leading to dysregulation of the alternative pathway of complement. Autoantibody directed against Factor H causes at least 6% to 10% of aHUS cases, but only a few clinical reports are available. Here, we describe the clinical, biologic, genetic features, treatment, and outcome of 45 patients who presented with aHUS associated with anti-FH autoantibody. We found that this form of aHUS primarily affects children between 9 and 13 years old but it also affects adults. It presents with a high frequency of gastrointestinal symptoms and with extrarenal complications and has a relapsing course. Activation of the alternative pathway of complement at the onset of disease portends a poor prognosis. Early specific treatment may lead to favorable outcomes. These data should improve the recognition and diagnosis of this form of aHUS and help identify patients at high risk of a poor outcome.


Assuntos
Autoanticorpos/imunologia , Fator H do Complemento/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/terapia , Adulto , Fatores Etários , Criança , Pré-Escolar , Estudos de Coortes , Ativação do Complemento/imunologia , Feminino , Seguimentos , Síndrome Hemolítico-Urêmica/diagnóstico , Síndrome Hemolítico-Urêmica/epidemiologia , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Lactente , Transplante de Rim , Masculino , Pessoa de Meia-Idade , Troca Plasmática/métodos , Medição de Risco , Índice de Gravidade de Doença , Fatores Sexuais , Transplante de Células-Tronco/métodos , Fatores de Tempo , Resultado do Tratamento
9.
Nephrol Dial Transplant ; 25(10): 3430-3, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20587423

RESUMO

BACKGROUND: We report autism in 3 out of 53 children with cystic or hyperechogenic kidneys and heterozygous 17q12 region deletion encompassing hepatocyte nuclear factor-1beta (HNF1B). RESULTS: They presented mental retardation, social interaction impairments, verbal and non-verbal communication deficits and stereotyped behaviours. Deletion size and location of breakpoints were similar to those reported in patients with renal disease/diabetes only. CONCLUSION: Reciprocal genomic rearrangements of the 17q12 region, reported in patients with mental retardation and epilepsy, could also be involved in autism. Nephrologists should be aware of the possibility of autism in patients with 17q12 deletion including HNF1B locus.


Assuntos
Transtorno Autístico/genética , Deleção Cromossômica , Cromossomos Humanos Par 17 , Fator 1-beta Nuclear de Hepatócito/genética , Doenças Renais Císticas/genética , Criança , Pré-Escolar , Proteínas de Homeodomínio/genética , Humanos , Doenças Renais Císticas/diagnóstico por imagem , Proteínas com Homeodomínio LIM , Masculino , Fatores de Transcrição , Ultrassonografia
10.
J Med Genet ; 47(7): 445-52, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20591883

RESUMO

BACKGROUND: Mutations in the PLCE1 gene encoding phospholipase C epsilon 1 (PLCepsilon1) have been recently described in patients with early onset nephrotic syndrome (NS) and diffuse mesangial sclerosis (DMS). In addition, two cases of PLCE1 mutations associated with focal segmental glomerulosclerosis (FSGS) and later NS onset have been reported. METHOD: In order to better assess the spectrum of phenotypes associated with PLCE1 mutations, mutational analysis was performed in a worldwide cohort of 139 patients (95 familial cases belonging to 68 families and 44 sporadic cases) with steroid resistant NS presenting at a median age of 23.0 months (range 0-373). RESULTS: Homozygous or compound heterozygous mutations were identified in 33% (8/24) of DMS cases. PLCE1 mutations were found in 8% (6/78) of FSGS cases without NPHS2 mutations. Nine were novel mutations. No clear genotype-phenotype correlation was observed, with either truncating or missense mutations detected in both DMS and FSGS, and leading to a similar renal evolution. Surprisingly, three unaffected and unrelated individuals were also found to carry the homozygous mutations identified in their respective families. CONCLUSION: PLCE1 is a major gene of DMS and is mutated in a non-negligible proportion of FSGS cases without NPHS2 mutations. Although additional variants in 19 candidate genes (16 other PLC genes, BRAF,IQGAP1 and NPHS1) were not identified, it is speculated that other modifier genes or environmental factors may play a role in the renal phenotype variability observed in individuals bearing PLCE1 mutations. This observation needs to be considered in the genetic counselling offered to patients.


Assuntos
Análise Mutacional de DNA , Síndrome Nefrótica/genética , Fosfoinositídeo Fosfolipase C/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Predisposição Genética para Doença/genética , Glomerulosclerose Segmentar e Focal/genética , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Mutação , Síndrome Nefrótica/tratamento farmacológico , Fenótipo , Estatísticas não Paramétricas , Esteroides/uso terapêutico
11.
J Am Soc Nephrol ; 21(7): 1209-17, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20507940

RESUMO

Mutations in NPHS1, which encodes nephrin, are the main causes of congenital nephrotic syndrome (CNS) in Finnish patients, whereas mutations in NPHS2, which encodes podocin, are typically responsible for childhood-onset steroid-resistant nephrotic syndrome in European populations. Genotype-phenotype correlations are not well understood in non-Finnish patients. We evaluated the clinical presentation, kidney histology, and disease progression in non-Finnish CNS cases by mutational screening in 107 families (117 cases) by sequencing the entire coding regions of NPHS1, NPHS2, PLCE1, WT1, LAMB2, PDSS2, COQ2, and NEPH1. We found that CNS describes a heterogeneous group of disorders in non-Finnish populations. We identified nephrin and podocin mutations in most families and only rarely found mutations in genes implicated in other hereditary forms of NS. In approximately 20% of cases, we could not identify the underlying genetic cause. Consistent with the major role of nephrin at the slit diaphragm, NPHS1 mutations associated with an earlier onset of disease and worse renal outcomes than NPHS2 mutations. Milder cases resulting from mutant NPHS1 had either two mutations in the cytoplasmic tail or two missense mutations in the extracellular domain, including at least one that preserved structure and function. In addition, we extend the spectrum of known NPHS1 mutations by describing long NPHS1 deletions. In summary, these data demonstrate that CNS is not a distinct clinical entity in non-Finnish populations but rather a clinically and genetically heterogeneous group of disorders.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Mutação/genética , Síndrome Nefrótica/congênito , Síndrome Nefrótica/genética , Fenótipo , África do Norte/epidemiologia , Biópsia , Pré-Escolar , Estudos de Coortes , Progressão da Doença , Europa (Continente)/epidemiologia , Feminino , Testes Genéticos , Genótipo , Humanos , Lactente , Recém-Nascido , Estimativa de Kaplan-Meier , Rim/patologia , Falência Renal Crônica/patologia , Masculino , Síndrome Nefrótica/epidemiologia , Estudos Retrospectivos , Turquia/epidemiologia
12.
Eur J Pediatr ; 169(7): 801-11, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20013129

RESUMO

Schimke immunoosseous dysplasia (SIOD) is an autosomal recessive multisystem disorder characterized by prominent spondyloepiphyseal dysplasia, T cell deficiency, and focal segmental glomerulosclerosis. Biallelic mutations in swi/snf-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a-like 1 (SMARCAL1) are the only identified cause of SIOD, but approximately half of patients referred for molecular studies do not have detectable mutations in SMARCAL1. We hypothesized that skeletal features distinguish between those with or without SMARCAL1 mutations. Therefore, we analyzed the skeletal radiographs of 22 patients with and 11 without detectable SMARCAL1 mutations. We found that patients with SMARCAL1 mutations have a spondyloepiphyseal dysplasia (SED) essentially limited to the spine, pelvis, capital femoral epiphyses, and possibly the sella turcica, whereas the hands and other long bones are basically normal. Additionally, we found that several of the adolescent and young adult patients developed osteoporosis and coxarthrosis. Of the 11 patients without detectable SMARCAL1 mutations, seven had a SED indistinguishable from patients with SMARCAL1 mutations. We conclude therefore that SED is a feature of patients with SMARCAL1 mutations and that skeletal features do not distinguish who of those with SED have SMARCAL1 mutations.


Assuntos
Osso e Ossos/diagnóstico por imagem , DNA Helicases/genética , Mutação , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/genética , Adolescente , Adulto , Criança , Pré-Escolar , Diagnóstico Diferencial , Heterogeneidade Genética , Glomerulosclerose Segmentar e Focal/genética , Humanos , Linfopenia/genética , Fenótipo , Radiografia , Síndrome
13.
Ophthalmology ; 116(5): 870-6, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19410944

RESUMO

OBJECTIVE: To analyze the corneas of patients with nephropathic cystinosis using in vivo confocal microscopy (IVCM) and anterior segment optical coherence tomography (AS-OCT). DESIGN: Prospective case series. PARTICIPANTS: Sixteen eyes of 8 patients with nephropathic cystinosis aged 8 to 21 years. METHODS: The ophthalmologic evaluation included best-corrected visual acuity, evaluation of photophobia (0-4), slit-lamp biomicroscopy analysis, intraocular pressure measurement, evaluation of crystal density using a slit-lamp-based scoring of the cornea, as well as AS-OCT and IVCM analysis. MAIN OUTCOME MEASURES: The depth of crystal deposition (DCD) in the central cornea and the central cornea thickness (CCT) were assessed using AS-OCT and IVCM. The IVCM images were evaluated for crystal density in each corneal layer and an IVCM score was calculated for each eye. RESULTS: All eyes had corneal crystal deposits, with a mean slit-lamp photography score of 2.90+/-0.13 (range, 2.75-3.00). Using AS-OCT, corneal crystals were observed in all eyes. These crystals appeared as hyperreflective punctuate deposits, predominantly observed within the anterior stroma. Measured with AS-OCT, the mean depth of DCD in the central cornea was 291.40+/-81.42 microm (range, 200-531 microm); the mean CCT was 543.47+/-29.62 microm. Using IVCM, the crystals appeared as spindle, needle-shaped, and fusiform hyperreflective bodies measuring from 1 to 175 microm in length and 1 to 30 microm in thickness. Except for the endothelium, randomly oriented crystals were observed in all corneal layers, with the greatest density observed within the anterior stroma. Measured with IVCM, the mean DCD was 426.07+/-88.19 microm (range, 284-531 microm); the mean CCT was 531.87+/-34.77 microm. There was no significant difference between the CCT measurements obtained with IVCM and with AS-OCT (mean difference, 11.31 microm; P = 0.07). Nevertheless, the DCD was significantly higher with IVCM than with AS-OCT (mean difference, 126.25 microm; P<0.0001). CONCLUSIONS: In patients with nephropathic cystinosis, IVCM could precisely quantify the density of crystals within the central cornea. Anterior segment OCT and IVCM should be used in further studies evaluating crystal deposition in patients with nephropathic cystinosis. FINANCIAL DISCLOSURE(S): The authors have no proprietary or commercial interest in any materials discussed in this article.


Assuntos
Córnea/patologia , Doenças da Córnea/diagnóstico , Cistinose/diagnóstico , Microscopia Confocal/métodos , Síndrome Nefrótica/diagnóstico , Tomografia de Coerência Óptica/métodos , Adolescente , Adulto , Criança , Doenças da Córnea/metabolismo , Cristalização , Cistina/metabolismo , Cistinose/metabolismo , Técnicas de Diagnóstico Oftalmológico , Feminino , Humanos , Pressão Intraocular , Masculino , Síndrome Nefrótica/metabolismo , Estudos Prospectivos , Acuidade Visual , Adulto Jovem
14.
Pediatr Nephrol ; 24(2): 395-402, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18985393

RESUMO

Data on the use of enteric-coated mycophenolic acid (EC-MPS) in pediatric transplantation cases are scarce. We undertook a 12-month, multicenter, open-label pilot study in which 16 de novo renal transplant patients aged 5-16 years received EC-MPS with cyclosporine A microemulsion (CsA-ME), steroids, and anti-interleukin-2 receptor antibody induction. The mean dose of EC-MPS was 916 +/- 93 mg/m(2) per day during weeks 1-2, 810 +/- 193 mg/m(2) per day during months 3-6, and 827 +/- 153 mg/m(2) per day during months 6-12. The mean CsA C(2) level exceeded target range up to month 6 post-transplant. Efficacy failure (biopsy-proven acute rejection, graft loss, death or loss to follow-up) occurred in two patients: one patient with primary non-function underwent nephrectomy, and one patient experienced biopsy-proven acute rejection (Grade 1B, day 344) following EC-MPS dose reduction. There were no deaths. Creatinine clearance (Schwartz) was 103 +/- 30 mL/min per 1.73 m(2) at month 6 and 100 +/- 16 mL/min per 1.73 m(2) at month 12. The majority of adverse events were mild or moderate (101/126, 80.2%). In this pilot study, EC-MPS 450 mg/m(2) administered twice daily with CsA, steroids, and interleukin-2 antibody induction resulted in a low rate of rejection with good renal function in a pediatric population. However, a larger, controlled trial is required to confirm these results.


Assuntos
Rejeição de Enxerto/tratamento farmacológico , Imunossupressores/administração & dosagem , Transplante de Rim , Ácido Micofenólico/administração & dosagem , Adolescente , Criança , Pré-Escolar , Creatinina/metabolismo , Ciclosporina/administração & dosagem , Ciclosporina/farmacocinética , Quimioterapia Combinada , Feminino , Rejeição de Enxerto/imunologia , Humanos , Imunossupressores/farmacocinética , Masculino , Ácido Micofenólico/farmacocinética , Projetos Piloto , Receptores de Interleucina-2/imunologia , Esteroides/administração & dosagem , Comprimidos com Revestimento Entérico , Resultado do Tratamento
15.
Pediatr Transplant ; 12(6): 619-29, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18482212

RESUMO

About 60% of non-Stx-associated aHUS are due to the defect of protection of endothelial cells from complement activation, secondary to mutations in the genes of CFH, MCP, IF, BF, or C3. In addition, 10% of patients have anti-CFH antibodies. While the risk of post-transplant recurrence is less than 1% in Stx-HUS patients, it is approximately 80% in CFH or IF-mutated patients, 20% in MCP-mutated patients, and 30% in patients with no mutation. Patients with anti-CFH antibodies probably also are at risk of recurrence. While MCP-mutated patients can reasonably go to transplantation, recent reports suggest that plasmatherapy started before surgery and maintained life-long may prevent recurrence in CFH-mutated patients. Four successful liver-kidney transplantation utilizing plasmatherapy in CFH-mutated children have been reported recently. In summary, the risk of post-transplant recurrence can now be approached according to genotype. Therefore, aHUS patients should undergo complement determination, screening for anti-CFH antibodies, and genotyping before transplantation. Kidney or kidney + liver transplantation with concomitant plasmatherapy need to be evaluated by prospective trials in patients with hereditary complement abnormalities.


Assuntos
Síndrome Hemolítico-Urêmica/diagnóstico , Transplante de Rim/efeitos adversos , Adulto , Pré-Escolar , Complemento C3/biossíntese , Convertases de Complemento C3-C5/metabolismo , Fator B do Complemento/biossíntese , Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/patologia , Humanos , Lactente , Transplante de Fígado/métodos , Proteína Cofatora de Membrana/biossíntese , Recidiva , Toxina Shiga/metabolismo
16.
Pediatr Transplant ; 12(4): 456-63, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18466433

RESUMO

There are few prospective clinical trials of mTOR inhibitors (or proliferation signal inhibitors) combined with CNI inhibitors in de novo pediatric renal transplantation. Results reported here are from a multicenter, open-label study in de novo pediatric renal transplant patients (

Assuntos
Corticosteroides/administração & dosagem , Ciclosporina/administração & dosagem , Imunossupressores/administração & dosagem , Transplante de Rim/métodos , Sirolimo/análogos & derivados , Adolescente , Biópsia , Criança , Quimioterapia Combinada , Everolimo , Feminino , Rejeição de Enxerto , Sobrevivência de Enxerto , Humanos , Masculino , Sirolimo/administração & dosagem , Resultado do Tratamento
18.
J Immunol ; 180(4): 2669-78, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250479

RESUMO

Inhibitory signaling is an emerging function of ITAM-bearing immunoreceptors in the maintenance of homeostasis. Monovalent targeting of the IgA Fc receptor (FcalphaRI or CD89) by anti-FcalphaRI Fab triggers potent inhibitory ITAM (ITAM(i)) signaling through the associated FcRgamma chain (FcalphaRI-FcRgamma ITAM(i)) that prevents IgG phagocytosis and IgE-mediated asthma. It is not known whether FcalphaRI-FcRgamma ITAM(i) signaling controls receptors that do not function through an ITAM and whether this inhibition requires Src homology protein 1 phosphatase. We show in this study that FcalphaRI-Fcgamma ITAM(i) signals depend on Src homology protein 1 phosphatase to target multiple non-ITAM-bearing receptors such as chemotactic receptors, cytokine receptors, and TLRs. We found that anti-FcalphaRI Fab treatment in vivo reduced kidney inflammation in models of immune-mediated glomerulonephritis and nonimmune obstructive nephropathy by a mechanism that involved decreased inflammatory cell infiltration and fibrosis development. This treatment also prevented ex vivo LPS activation of monocytes from patients with lupus nephritis or vasculitis, as well as receptor activation through serum IgA complexes from IgA nephropathy patients. These findings point to a crucial role of FcalphaRI-FcRgamma ITAM(i) signaling in the control of multiple heterologous or autologous inflammatory responses. They also identify anti-FcalphaRI Fab as a new potential therapeutic tool for preventing progression of renal inflammatory diseases.


Assuntos
Inibição de Migração Celular/imunologia , Movimento Celular/imunologia , Glomerulonefrite por IGA/patologia , Glomerulonefrite por IGA/prevenção & controle , Receptores Imunológicos/fisiologia , Transdução de Sinais/imunologia , Motivos de Aminoácidos/imunologia , Animais , Antígenos CD/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Glomerulonefrite por IGA/imunologia , Glomerulonefrite por IGA/metabolismo , Humanos , Rim/imunologia , Rim/patologia , Leucócitos/imunologia , Leucócitos/metabolismo , Leucócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Ratos , Receptores Fc/fisiologia , Receptores de IgG/fisiologia
19.
Circ Cardiovasc Genet ; 1(2): 133-40, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20016754

RESUMO

BACKGROUND: Generalized arterial calcification of infancy has been reported to be frequently lethal, and the efficiency of any therapy, including bisphosphonates, is unknown. A phosphate-poor diet markedly increases survival of NPP1 null mice, a model of generalized arterial calcification of infancy. METHODS AND RESULTS: We performed a multicenter genetic study and retrospective observational analysis of 55 subjects affected by generalized arterial calcification of infancy to identify prognostic factors. Nineteen (34%) patients survived the critical period of infancy. In all 8 surviving patients tested, hypophosphatemia due to reduced renal tubular phosphate reabsorption developed during childhood. Eleven of 17 (65%) patients treated with bisphosphonates survived. Of 26 patients who survived their first day of life and were not treated with bisphosphonates only 8 (31%) patients survived beyond infancy. Forty different homozygous or compound heterozygous mutations, including 16 novel mutations in ENPP1, were found in 41 (75%) of the 55 patients. Twenty-nine (71%) of these 41 patients died in infancy (median, 30 days). Seven of the 14 (50%) patients without ENPP1 mutations died in infancy (median, 9 days). When present on both alleles, the mutation p.P305T was associated with death in infancy in all 5 cases; otherwise, no clear genotype-phenotype correlation was seen. CONCLUSION: ENPP1 coding region mutations are associated with generalized arterial calcification of infancy in approximately 75% of subjects. Except for the p.P305T mutation, which was universally lethal when present on both alleles, the identified ENPP1 mutations per se have no discernable effect on survival. However, survival seems to be associated with hypophosphatemia linked with hyperphosphaturia and also with bisphosphonate treatment.


Assuntos
Calcinose/genética , Difosfonatos/uso terapêutico , Hipofosfatemia Familiar/genética , Hipofosfatemia/genética , Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética , Alelos , Angiografia , Artérias/diagnóstico por imagem , Calcinose/tratamento farmacológico , Calcinose/mortalidade , Estudos de Coortes , Feminino , Heterozigoto , Homozigoto , Humanos , Hipofosfatemia/complicações , Hipofosfatemia Familiar/complicações , Lactente , Recém-Nascido , Masculino , Mutação , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/metabolismo , Análise de Sobrevida , Ultrassonografia
20.
Nephrol Dial Transplant ; 23(4): 1291-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18065803

RESUMO

BACKGROUND AND OBJECTIVES: Diffuse mesangial sclerosis (DMS) is a histologically distinct variant of nephrotic syndrome (NS) that is characterized by early onset and by progression to end-stage kidney disease (ESKD). Besides syndromic DMS, isolated (non-syndromic) DMS (IDMS) has been described. The etiology and pathogenesis of DMS is not understood. We recently identified by positional cloning recessive mutations in the gene PLCE1/NPHS3 as a novel cause of IDMS. We demonstrated a role of PLCE1 in glomerulogenesis. Mutations in two other genes WT1 and LAMB2 may also cause IDMS. We therefore determine in this study the relative frequency of mutations in PLCE1, WT1 or LAMB2 as the cause of IDMS in a worldwide cohort. METHODS: We identified 40 children from 35 families with IDMS from a worldwide cohort of 1368 children with NS. All the subjects were analyzed for mutations in all exons of PLCE1 by multiplex capillary heteroduplex analysis and direct sequencing, by direct sequencing of exons 8 and 9 of WT1, and all the exons of LAMB2. RESULTS: The median (range) age at onset of NS was 11 (1-72) months. We detected truncating mutations in PLCE1 in 10/35 (28.6%) families and WT1 mutations in 3/35 (8.5%) families. We found no mutations in LAMB2. CONCLUSIONS: PLCE1 mutation is the most common cause of IDMS in this cohort. We previously reported that one child with truncating mutation in PLCE1 responded to cyclosporine therapy. If this observation is confirmed in a larger study, mutations in PLCE1 may serve as a biomarker for selecting patients with IDMS who may benefit from treatment.


Assuntos
Mesângio Glomerular/patologia , Mutação , Nefroesclerose/genética , Fosfoinositídeo Fosfolipase C/genética , Biópsia , Pré-Escolar , DNA , Análise Mutacional de DNA , Éxons , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Lactente , Laminina/genética , Laminina/metabolismo , Masculino , Nefroesclerose/metabolismo , Nefroesclerose/patologia , Fosfoinositídeo Fosfolipase C/metabolismo , Reação em Cadeia da Polimerase , Prognóstico , Índice de Gravidade de Doença , Proteínas WT1/genética , Proteínas WT1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA