Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Br J Haematol ; 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38934404

RESUMO

Glomerular hyperfiltration and albuminuria are frequent kidney abnormalities in children with sickle cell anaemia (SCA). However, little is known about their persistence in African SCA children. This prospective study included 600 steady-state SCA children aged 2-18 years from the Democratic Republic of Congo. Participants were genotyped for apolipoprotein L1 (APOL1) risk variants (RVs) and haem oxygenase-1 (HMOX1) GT-dinucleotide repeats. Kidney abnormalities were defined as albuminuria, hyperfiltration or decreased estimated creatinine-based glomerular filtration rate (eGFRcr). At baseline, 247/600 (41.2%) participants presented with kidney abnormalities: 82/592 (13.8%) with albuminuria, 184/587 (31.3%) with hyperfiltration and 15/587 (2.6%) with decreased eGFRcr. After a median follow-up of 5 months, repeated testing was performed in 180/247 (72.9%) available participants. Persistent hyperfiltration and persistent albuminuria (PA) were present in 29.2% (38/130) and 39.7% (23/58) respectively. eGFR normalized in all participants with a baseline decreased eGFRcr. Haemoglobinuria (p = 0.017) and male gender (p = 0.047) were significantly associated with PA and persistent hyperfiltration respectively. APOL1 RVs (G1G1/G2G2/G1G2) were borderline associated with PA (p = 0.075), while HMOX1 long repeat was not associated with any persistent kidney abnormality. This study reveals that a single screening can overestimate the rate of kidney abnormalities in children with SCA and could lead to overtreatment.

2.
Pediatr Nephrol ; 38(8): 2509-2521, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36472655

RESUMO

HIV infection remains one of the leading causes of morbidity and mortality worldwide, especially in children living in resource-limited settings. Although the World Health Organization (WHO) recently recommended antiretroviral therapy (ART) initiation upon diagnosis regardless of the number of CD4, ART access remains limited, especially in children living in sub-Saharan Africa (SSA). HIV-infected children who do not receive appropriate ART are at increased risk of developing HIV-associated nephropathy (HIVAN). Although due to genetic susceptibility, SSA is recognized to be the epicenter of HIVAN, limited information is available regarding the burden of HIVAN in children living in Africa. The present review discusses the information available to date on the prevalence, pathogenesis, risk factors, diagnosis, and management of HIVAN in children, focusing on related challenges in a resource-limited setting.


Assuntos
Nefropatia Associada a AIDS , Infecções por HIV , Humanos , Criança , Nefropatia Associada a AIDS/diagnóstico , Nefropatia Associada a AIDS/epidemiologia , Nefropatia Associada a AIDS/etiologia , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/epidemiologia , Região de Recursos Limitados , Fatores de Risco , África Subsaariana/epidemiologia
3.
Br J Haematol ; 196(1): 204-214, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34545573

RESUMO

Clinical and genetic factors have been reported as influencing the development of sickle cell nephropathy (SCN). However, such data remain limited in the paediatric population. In this cross-sectional study, we enrolled 361 sickle cell disease children from the Democratic Republic of Congo. Participants were genotyped for the beta (ß)-globin gene, apolipoprotein L1 (APOL1) risk variants, and haem oxygenase-1 (HMOX1) GT-dinucleotide repeats. As markers of kidney damage, albuminuria, hyperfiltration and decreased estimated glomerular filtration with creatinine (eGFRcr) were measured. An association of independent clinical and genetic factors with these markers of kidney damage were assessed via regression analysis. Genetic sequencing confirmed sickle cell anaemia in 326 participants. Albuminuria, hyperfiltration and decreased eGFRcr were present in 65 (20%), 52 (16%) and 18 (5·5%) patients, respectively. Regression analysis revealed frequent blood transfusions, indirect bilirubin and male gender as clinical predictors of SCN. APOL1 high-risk genotype (G1/G1, G2/G2 and G1/G2) was significantly associated with albuminuria (P = 0·04) and hyperfiltration (P = 0·001). HMOX1 GT-dinucleotide long repeats were significantly associated with lower eGFRcr. The study revealed a high burden of kidney damage among Congolese children and provided evidence of the possible role of APOL1 and HMOX1 in making children more susceptible to kidney complications.


Assuntos
Anemia Falciforme/complicações , Anemia Falciforme/epidemiologia , População Negra , Suscetibilidade a Doenças , Nefropatias/epidemiologia , Nefropatias/etiologia , Adolescente , Anemia Falciforme/diagnóstico , Anemia Falciforme/genética , Apolipoproteína L1/genética , Criança , Pré-Escolar , Estudos Transversais , Índices de Eritrócitos , Feminino , Predisposição Genética para Doença , Variação Genética , Taxa de Filtração Glomerular , Heme Oxigenase-1/genética , Humanos , Nefropatias/diagnóstico , Testes de Função Renal , Masculino , Mutação , Globinas beta/metabolismo
4.
Pediatr Nephrol ; 37(6): 1231-1243, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34050806

RESUMO

The life expectancy of individuals with sickle cell disease has increased over the years, majorly due to an overall improvement in diagnosis and medical care. Nevertheless, this improved longevity has resulted in an increased prevalence of chronic complications such as sickle cell nephropathy (SCN), which poses a challenge to the medical care of the patient, shortening the lifespan of patients by 20-30 years. Clinical presentation of SCN is age-dependent, with kidney dysfunction slowly beginning to develop from childhood, progressing to chronic kidney disease and kidney failure during the third and fourth decades of life. This review explores the epidemiology, pathology, pathophysiology, clinical presentation, and management of SCN by focusing on the pediatric population. It also discusses the factors that can modify SCN susceptibility.


Assuntos
Anemia Falciforme , Insuficiência Renal Crônica , Doenças Vasculares , Anemia Falciforme/complicações , Anemia Falciforme/epidemiologia , Anemia Falciforme/terapia , Criança , Humanos , Prevalência , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/etiologia
5.
Pediatr Nephrol ; 36(4): 777-788, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32253519

RESUMO

Individuals of African origin have an increased risk of developing various progressive chronic kidney diseases (CKD). This risk has been attributed to genetic variants (G1, G2) in apolipoprotein-L1 (APOL1) gene. In the pediatric population, especially in children affected by sickle cell disease (SCD), by human immunodeficiency virus (HIV), or with various glomerular diseases, APOL1 risk variants have been associated with the development of hypertension, albuminuria, and more rapid decline of kidney function. The present review focuses on existing APOL1-related epidemiological data in children with CKD. It also includes data from studies addressing racial disparities in CKD, the APOL1-related innate immunity, and the relationship between APOL1 and CKD and pathogenic pathways mediating APOL1-related kidney injury.


Assuntos
Apolipoproteína L1 , Insuficiência Renal Crônica , Albuminúria , Apolipoproteína L1/genética , Criança , Predisposição Genética para Doença , Humanos , Rim , Insuficiência Renal Crônica/epidemiologia , Insuficiência Renal Crônica/genética
6.
Kidney Int ; 89(5): 1037-1048, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27083281

RESUMO

The involvement of the glomerulus in the pathogenesis of cystinosis, caused by loss-of-function mutations in cystinosin (CTNS, 17p13), is a matter of controversy. Although patients with cystinosis demonstrate glomerular lesions and high-molecular-weight proteinuria starting from an early age, a mouse model of cystinosis develops only signs of proximal tubular dysfunction. Here we studied podocyte damage in patients with cystinosis by analyzing urinary podocyte excretion and by in vitro studies of podocytes deficient in cystinosin. Urine from patients with cystinosis presented a significantly higher amount of podocytes compared with controls. In culture, cystinotic podocytes accumulated cystine compatible with cystinosin deficiency. The expression of podocyte specific genes CD2AP, podocalyxin, and synaptopodin and of the WT1 protein was evident in all cell lines. Conditionally immortalized podocyte lines of 2 patients with different CTNS mutations had altered cytoskeleton, impaired cell adhesion sites, and increased individual cell motility. Moreover, these cells showed enhanced phosphorylation of both Akt1 and Akt2 (isoforms of protein kinase B). Inhibition of Akt by a specific inhibitor (Akti inhibitor 1/2) resulted in normalization of the hypermotile phenotype. Thus, our study extends the list of genetic disorders causing podocyte damage and provides the evidence of altered cell signaling cascades resulting in impaired cell adhesion and enhanced cell motility in cystinosis.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Movimento Celular , Cistinose/metabolismo , Podócitos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adolescente , Sistemas de Transporte de Aminoácidos Neutros/deficiência , Sistemas de Transporte de Aminoácidos Neutros/genética , Estudos de Casos e Controles , Adesão Celular , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Criança , Pré-Escolar , Cistinose/genética , Cistinose/patologia , Cistinose/urina , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Regulação para Baixo , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Mutação , Fenótipo , Fosforilação , Podócitos/efeitos dos fármacos , Podócitos/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Transdução de Sinais , Urina/citologia , Proteínas WT1/genética , Proteínas WT1/metabolismo , Adulto Jovem
7.
Kidney Int ; 89(6): 1192-203, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27181776

RESUMO

Nephropathic cystinosis is an autosomal recessive metabolic, lifelong disease characterized by lysosomal cystine accumulation throughout the body that commonly presents in infancy with a renal Fanconi syndrome and, if untreated, leads to end-stage kidney disease (ESKD) in the later childhood years. The molecular basis is due to mutations in CTNS, the gene encoding for the lysosomal cystine-proton cotransporter, cystinosin. During adolescence and adulthood, extrarenal manifestations of cystinosis develop and require multidisciplinary care. Despite substantial improvement in prognosis due to cystine-depleting therapy with cysteamine, no cure of the disease is currently available. Kidney Disease: Improving Global Outcomes (KDIGO) convened a Controversies Conference on cystinosis to review the state-of-the-art knowledge and to address areas of controversies in pathophysiology, diagnostics, monitoring, and treatment in different age groups. More importantly, promising areas of investigation that may lead to optimal outcomes for patients afflicted with this lifelong, systemic disease were discussed with a research agenda proposed for the future.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Cisteamina/uso terapêutico , Eliminadores de Cistina/uso terapêutico , Cistina/metabolismo , Cistinose/etiologia , Doenças Raras/etiologia , Adolescente , Adulto , Fatores Etários , Criança , Congressos como Assunto , Cisteamina/efeitos adversos , Eliminadores de Cistina/efeitos adversos , Cistinose/complicações , Cistinose/diagnóstico , Cistinose/terapia , Síndrome de Fanconi/complicações , Síndrome de Fanconi/tratamento farmacológico , Testes Genéticos , Terapia Genética , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas , Humanos , Terapia de Imunossupressão/efeitos adversos , Lactente , Falência Renal Crônica/etiologia , Transplante de Rim/efeitos adversos , Lisossomos/metabolismo , Mutação , Doenças Raras/complicações , Doenças Raras/diagnóstico , Doenças Raras/terapia , Diálise Renal
8.
J Inherit Metab Dis ; 39(3): 457-464, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26909499

RESUMO

Lysosomes play a central role in regulating autophagy via activation of mammalian target of rapamycin complex 1 (mTORC1). We examined mTORC1 signalling in the lysosomal storage disease nephropathic cystinosis (MIM 219800), in which accumulation of autophagy markers has been previously demonstrated. Cystinosis is caused by mutations in the lysosomal cystine transporter cystinosin and initially affects kidney proximal tubules causing renal Fanconi syndrome, followed by a gradual development of end-stage renal disease and extrarenal complications. Using proximal tubular kidney cells obtained from healthy donors and from cystinotic patients, we demonstrate that cystinosin deficiency is associated with a perturbed mTORC1 signalling, delayed reactivation of mTORC1 after starvation and abnormal lysosomal retention of mTOR during starvation. These effects could not be reversed by treatment with cystine-depleting drug cysteamine. Altered mTORC1 signalling can contribute to the development of proximal tubular dysfunction in cystinosis and points to new possibilities in therapeutic intervention through modulation of mTORC-dependent signalling cascades.


Assuntos
Cistinose/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Transdução de Sinais/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Autofagia/fisiologia , Células Cultivadas , Cisteamina/metabolismo , Cistinose/patologia , Síndrome de Fanconi/metabolismo , Síndrome de Fanconi/patologia , Humanos , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Lisossomos/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(17): 7014-9, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23572577

RESUMO

Receptor-mediated endocytosis, involving megalin and cubilin, mediates renal proximal-tubular reabsorption and is decreased in Dent disease because of mutations of the chloride/proton antiporter, chloride channel-5 (CLC-5), resulting in low-molecular-weight proteinuria, hypercalciuria, nephrolithiasis, and renal failure. To facilitate studies of receptor-mediated endocytosis and the role of CLC-5, we established conditionally immortalized proximal-tubular epithelial cell lines (ciPTECs) from three patients with CLC-5 mutations (30:insH, R637X, and del132-241) and a normal male. Confocal microscopy using the tight junction marker zona occludens-1 (ZO-1) and end-binding protein-1 (EB-1), which is specific for the plus end of microtubules demonstrated that the ciPTECs polarized. Receptor-mediated endocytic uptake of fluorescent albumin and transferrin in 30:insH and R637X ciPTECs was significantly decreased, compared with normal ciPTECs, and could be further reduced by competition with 10-fold excess of unlabeled albumin and transferrin, whereas in the del132-241 ciPTEC, receptor-mediated endocytic uptake was abolished. Investigation of endosomal acidification by live-cell imaging of pHluorin-VAMP2 (vesicle-associated membrane protein-2), a pH-sensitive-GFP construct, revealed that the endosomal pH in normal and 30:insH ciPTECs was similar, whereas in del132-241 and R637X ciPTECs, it was significantly more alkaline, indicating defective acidification in these ciPTECs. The addition of bafilomycin-A1, a V-ATPase inhibitor, raised the pH significantly in all ciPTECs, demonstrating that the differences in acidification were not due to alterations in the V-ATPase, but instead to abnormalities of CLC-5. Thus, our studies, which have established human Dent disease ciPTECs that will facilitate studies of mechanisms in renal reabsorption, demonstrate that Dent disease-causing CLC-5 mutations have differing effects on endosomal acidification and receptor-mediated endocytosis that may not be coupled.


Assuntos
Doença de Dent/fisiopatologia , Endocitose/fisiologia , Endossomos/química , Células Epiteliais/fisiologia , Túbulos Renais Proximais/citologia , Linhagem Celular , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Doença de Dent/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Microscopia Confocal , Mutação/genética , Proteína 2 Associada à Membrana da Vesícula/metabolismo
10.
Am J Physiol Renal Physiol ; 308(11): F1200-6, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25656364

RESUMO

The calcium-sensing receptor (CaSR) is a G protein-coupled receptor, which plays an essential role in regulating Ca(2+) homeostasis. Here we show that conditionally immortalized proximal tubular epithelial cell line (ciPTEC) obtained by immortalizing and subcloning cells exfoliated in the urine of a healthy subject expresses functional endogenous CaSR. Immunolocalization studies of polarized ciPTEC revealed the apical localization of the receptor. By Western blotting of ciPTEC lysates, both monomeric and dimeric forms of CaSR at 130 and ∼250 kDa, respectively, were detected. Functional studies indicated that both external calcium and the positive CaSR allosteric modulator, NPS-R568, induced a significant increase in cytosolic calcium, proving a high sensitivity of the endogenous receptor to its agonists. Calcium depletion from the endoplasmic reticulum using cyclopiazonic acid abolished the increase in cytosolic calcium elicited by NPS-R568, confirming calcium exit from intracellular stores. Activation of CaSR by NPS-R significantly reduced the increase in cAMP elicited by forskolin (FK), a direct activator of adenylate cyclase, further confirming the functional expression of the receptor in this cell line. CaSR expressed in ciPTEC was found to interact with Gq as a downstream effector, which in turn can cause release of calcium from intracellular stores via phospholipase C activation. We conclude that human proximal tubular ciPTEC express functional CaSR and respond to its activation with a release of calcium from intracellular stores. These cell lines represent a valuable tool for research into the disorder associated with gain or loss of function of the CaSR by producing cell lines from patients.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Células Epiteliais/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Compostos de Anilina/farmacologia , Cálcio/metabolismo , Linhagem Celular , Separação Celular/métodos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Epiteliais/efeitos dos fármacos , Humanos , Fenetilaminas , Propilaminas , Receptores de Detecção de Cálcio/análise
11.
Pediatr Nephrol ; 30(8): 1349-60, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25956701

RESUMO

BACKGROUND: Cystinosis is a rare, inherited autosomal recessive disease caused by the accumulation of free cystine in lysosomes. It is treated by the administration of cysteamine, which should be monitored by trough white blood cell (WBC) cystine measurements to ensure effective treatment. CASE-DIAGNOSIS/TREATMENT: The index case had an older brother who had previously been diagnosed with cystinosis, allowing early diagnosis of the index case at the age of 5 months. Cysteamine therapy was started at the age of 3 years; however, monitoring of WBC cystine levels did not occur on a regular basis during most of his life. Growth retardation improved after correction of electrolyte disturbances, the initiation of cysteamine therapy and treatment with recombinant human growth hormone. Renal replacement therapy was started at the age of 11 years, and renal transplantation was performed at the age of 12 years. Extra-renal cystine accumulation caused multiple endocrinopathies (including adrenal insufficiency, hypothyroidism and primary hypogonadism), neurological symptoms, pancytopenia owing to splenomegaly and portal hypertension due to nodular regenerative hyperplasia, aggravated by splenic vein thrombosis and partial portal vein thrombosis. The patient died of diffuse intra-abdominal bleeding caused by severe portal hypertension. CONCLUSION: Cysteamine treatment should be started as early as possible, and dosage should be monitored and adapted based on trough WBC cystine levels. RELEVANT INTERNATIONAL GUIDELINE: Emma F et al. (2014) Nephropathic cystinosis: an international consensus document. Nephrol Dial Transplant 29:iv87-iv94.


Assuntos
Cisteamina/administração & dosagem , Cistinose/tratamento farmacológico , Cistinose/fisiopatologia , Adolescente , Adulto , Criança , Pré-Escolar , Cistinose/complicações , Gerenciamento Clínico , Evolução Fatal , Humanos , Lactente , Recém-Nascido , Transplante de Rim , Masculino , Nefrologia , Pediatria , Linhagem , Insuficiência Renal/etiologia , Insuficiência Renal/cirurgia , Adulto Jovem
12.
Kidney Int Rep ; 9(2): 214-224, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38344731

RESUMO

Cystinosis is a rare, inherited, lysosomal storage disorder characterized by the progressive accumulation of intralysosomal cystine and subsequent organ and tissue damage. The kidneys are the first and most severely impacted organ. Although cystinosis was once considered a fatal pediatric disease, patients with cystinosis are living well into adulthood with advances in medical care, including kidney transplant and early and continuous use of cysteamine therapy. This increase in life expectancy has revealed an extrarenal phenotype of cystinosis that emerges in adolescence and adulthood, affecting nearly all body systems, including the endocrine and reproductive systems. As individuals with cystinosis are planning for the future, reproductive health and fertility have become areas of increased focus. This narrative review aims to summarize the current understanding of reproductive health and fertility in patients with cystinosis and discuss practical considerations for monitoring and managing these complications.

13.
Genet Med ; 15(1): 55-63, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22899094

RESUMO

PURPOSE: Aquaporin 7 (AQP7) belongs to the aquaglyceroporin family, which transports glycerol and water. AQP7-deficient mice develop obesity, insulin resistance, and hyperglyceroluria. However, AQP7's pathophysiologic role in humans is not yet known. METHODS: Three children with psychomotor retardation and hyperglyceroluria were screened for AQP7 mutations. The children were from unrelated families. Urine and plasma glycerol levels were measured using a three-step enzymatic approach. Platelet morphology and function were studied using electron microscopy, aggregations, and adenosine triphosphate (ATP) secretion tests. RESULTS: The index patients were homozygous for AQP7 G264V, which has previously been shown to inhibit transport of glycerol in Xenopus oocytes. We also detected a subclinical platelet secretion defect with reduced ATP secretion, and the absence of a secondary aggregation wave after epinephrine stimulation. Electron microscopy revealed round platelets with centrally located granules. Immunostaining showed AQP7 colocalization, with dense granules that seemed to be released after strong platelet activation. Healthy relatives of these patients, who were homozygous (not heterozygous) for G264V, also had hyperglyceroluria and platelet granule abnormalities. CONCLUSION: The discovery of an association between urine glycerol loss and a platelet secretion defect is a novel one, and our findings imply the involvement of AQPs in platelet secretion. Additional studies are needed to define whether AQP7 G264V is also a risk factor for mental disability.


Assuntos
Aquaporinas/genética , Transtornos Plaquetários/genética , Homozigoto , Mutação , Adolescente , Adulto , Substituição de Aminoácidos , Aquaporina 3/genética , Aquaporinas/metabolismo , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Criança , Pré-Escolar , Códon , Feminino , Glicerol/sangue , Glicerol/urina , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Linhagem , Transporte Proteico , Adulto Jovem
14.
J Pediatr ; 163(3): 754-60, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23651769

RESUMO

OBJECTIVES: To assess whether copper deficiency plays a role in the recently described cysteamine toxicity in patients with cystinosis, and to examine whether polymorphisms in copper transporters, lysyl oxidase, and/or type I procollagen genes could be responsible for the occurrence of cysteamine toxicity in a small subset of patients with cystinosis. STUDY DESIGN: Thirty-six patients with cystinosis were included: 22 with Fanconi syndrome (including 7 with cysteamine toxicity), 12 after renal transplantation, 1 receiving hemodialysis, and 1 with ocular cystinosis. Serum copper and ceruloplasmin levels and urinary copper/creatinine ratio were measured. Genes ATP7A and CTR1 (encoding copper transporters), LOX (encoding lysyl oxidase), and COL1A1 and COL1A2 (encoding type I procollagen) were analyzed in patients with (n = 6) and without (n = 5) toxicity. Fibroblast (pro)collagen synthesis was compared in patients with (n = 3) and those without (n = 2) cysteamine toxicity. RESULTS: All 22 patients with Fanconi syndrome had increased urinary copper excretion. Serum copper and ceruloplasmin levels were decreased in 9 patients, including all 7 patients with cysteamine toxicity. No specific sequence variations were associated with toxicity. All fibroblasts exhibited normal (pro)collagen synthesis. CONCLUSION: Patients with cystinosis with cysteamine toxicity demonstrate copper deficiency. This can cause decreased activity of lysyl oxidase, the enzyme that generates the aldehydes required for collagen cross-linking. Thus, copper supplementation might prevent cysteamine toxicity.


Assuntos
Cobre/deficiência , Cisteamina/efeitos adversos , Cistinose/complicações , Substâncias Protetoras/efeitos adversos , Fármacos Renais/efeitos adversos , Adenosina Trifosfatases/genética , Adolescente , Adulto , Biomarcadores/metabolismo , Proteínas de Transporte de Cátions/genética , Ceruloplasmina/metabolismo , Criança , Pré-Escolar , Colágeno/metabolismo , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Cobre/metabolismo , Transportador de Cobre 1 , ATPases Transportadoras de Cobre , Cisteamina/uso terapêutico , Cistinose/tratamento farmacológico , Cistinose/genética , Cistinose/metabolismo , Síndrome de Fanconi/complicações , Síndrome de Fanconi/tratamento farmacológico , Síndrome de Fanconi/genética , Síndrome de Fanconi/metabolismo , Feminino , Marcadores Genéticos , Humanos , Masculino , Polimorfismo Genético , Substâncias Protetoras/uso terapêutico , Proteína-Lisina 6-Oxidase/genética , Fármacos Renais/uso terapêutico , Análise de Sequência de DNA , Adulto Jovem
15.
Biochim Biophys Acta ; 1812(6): 643-51, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21371554

RESUMO

Recent evidence implies that impaired metabolism of glutathione has a role in the pathogenesis of nephropathic cystinosis. This recessive inherited disorder is characterized by lysosomal cystine accumulation and results in renal Fanconi syndrome progressing to end stage renal disease in the majority of patients. The most common treatment involves intracellular cystine depletion by cysteamine, delaying the development of end stage renal disease by a yet elusive mechanism. However, cystine depletion does not arrest the disease nor cures Fanconi syndrome in patients, indicating involvement of other yet unknown pathologic pathways. Using a newly developed proximal tubular epithelial cell model from cystinotic patients, we investigate the effect of cystine accumulation and cysteamine on both glutathione and ATP metabolism. In addition to the expected increase in cystine and defective sodium-dependent phosphate reabsorption, we observed less negative glutathione redox status and decreased intracellular ATP levels. No differences between control and cystinosis cell lines were observed with respect to protein turnover, albumin uptake, cytosolic and mitochondrial ATP production, total glutathione levels, protein oxidation and lipid peroxidation. Cysteamine treatment increased total glutathione in both control and cystinotic cells and normalized cystine levels and glutathione redox status in cystinotic cells. However, cysteamine did not improve decreased sodium-dependent phosphate uptake. Our data implicate that cysteamine increases total glutathione and restores glutathione redox status in cystinosis, which is a positive side-effect of this agent next to cystine depletion. This beneficial effect points to a potential role of cysteamine as anti-oxidant for other renal disorders associated with enhanced oxidative stress.


Assuntos
Cisteamina/farmacologia , Cistinose/metabolismo , Glutationa/metabolismo , Túbulos Renais Proximais/metabolismo , Trifosfato de Adenosina/metabolismo , Adolescente , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Criança , Pré-Escolar , Células Epiteliais/metabolismo , Feminino , Humanos , Lactente , Masculino , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
16.
Pediatr Nephrol ; 27(11): 2123-2127, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22664570

RESUMO

BACKGROUND: Cystinosis is an autosomal recessive disorder characterized by intralysosomal cystine accumulation. Growth retardation is more pronounced in cystinosis than in other chronic kidney diseases and is mostly not corrected by cysteamine. METHODS: Growth was evaluated in nine cystinosis patients, all treated with cysteamine, both after cysteamine and recombinant human growth hormone (rhGH) therapy initiation. Growth hormone (GH) secretion was studied by nocturnal GH measurements in four of nine patients and by glucagon test in four of nine patients. RESULTS: RhGH was administered to seven of nine patients. At rhGH initiation, height was below -2 SDS in five of seven patients, final height was above -2 SDS in six of seven. In two patients not treated with rhGH, final height remained below -4 SDS despite cysteamine treatment being started at the age of 6.1 and 8.1 years, respectively. Nocturnal GH secretion was normal in all patients. Glucagon tests revealed GH deficiency in one patient; two of four patients had abnormal GH peak timing. CONCLUSIONS: We present the first reported case of GH deficiency in cystinosis. Although no overt GH deficiency was detected in other patients, abnormal GH peak timing can indicate a subclinical GH secretion problem. RhGH significantly improved growth in cystinosis patients and should be initiated early in life.


Assuntos
Ritmo Circadiano , Cistinose/sangue , Transtornos do Crescimento/sangue , Hormônio do Crescimento Humano/sangue , Biomarcadores/sangue , Estatura/efeitos dos fármacos , Estudos de Casos e Controles , Criança , Pré-Escolar , Cisteamina/uso terapêutico , Cistinose/complicações , Cistinose/tratamento farmacológico , Feminino , Glucagon , Transtornos do Crescimento/tratamento farmacológico , Transtornos do Crescimento/etiologia , Terapia de Reposição Hormonal , Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/uso terapêutico , Humanos , Lactente , Masculino , Fatores de Tempo , Resultado do Tratamento
17.
J Pediatr ; 159(6): 1004-11, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21784456

RESUMO

OBJECTIVE: To report new adverse effects of cysteamine. STUDY DESIGN: Detailed clinical information was obtained from the patients' physicians. RESULTS: New adverse events were reported in 8 of 550 patients with cystinosis treated with cysteamine in Europe during the last 5 years. Detailed clinical information was not available for 2 of these patients, 1 of whom died from cerebral ischemia. The 6 evaluable patients developed vascular elbow lesions (6/6), neurologic symptoms (1/6), bone and muscle pain (2/6), and/or skin striae (2/6). Analysis of biopsy specimens from the elbow lesions demonstrated angioendotheliomatosis with irregular collagen fibers. In 3 of the 6 patients, the daily cysteamine dose exceeded the recommended maximum of 1.95 g/m(2)/day. Dose reduction led to improvement of signs and symptoms in all 6 patients, suggesting a causal relationship with cysteamine administration. CONCLUSION: Cysteamine administration can be complicated by the development of skin, vascular, neurologic, muscular, and bone lesions. These lesions improve after cysteamine dose reduction. Doses >1.95 g/m(2)/day should be prescribed with great caution, but underdosing is not advocated.


Assuntos
Cisteamina/toxicidade , Cistinose/tratamento farmacológico , Toxidermias/etiologia , Pré-Escolar , Humanos , Lactente , Masculino
18.
J Inherit Metab Dis ; 34(3): 811-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21305353

RESUMO

BACKGROUND: Cystinosis is characterized by intralysosomal cystine accumulation, causing end stage renal disease around 10 years of age if not treated with cysteamine. Cystine accumulation in blood vessels might increase atheroma formation or arterial stiffness and therefore increase the risk for cardiovascular disease (CVD). This study aimed to investigate the risk for CVD by non-invasive measures of atherosclerosis (NIMA) and to evaluate the effect of cysteamine treatment. PATIENTS AND METHODS: Thirteen Dutch adult cystinosis patients were included. White blood cell (WBC) cystine levels, glomerular filtration rate (GFR) and concommitant medications were obtained from medical records. NIMA included carotid intima-media thickness (cIMT, n = 13), pulse wave velocity (PWV, n = 8) and pulse wave analysis (PWA, n = 6). RESULTS: GFR ranged between 4-95 mL/min/1.73 m². All but one patient were treated with cysteamine, mean WBC cystine values ranged between 0.34-1.64 nmol cystine/mg protein, 8 patients had mean WBC cystine levels <1 nmol cystine/mg protein. When compared to healthy subjects, cIMT and PWV levels were above normal values in 1 patient for each measure. PWA measurements showed high augmentation index in three patients who did not receive lipid-lowering medication. When corrected for renal function, cIMT and PWV levels were within the normal range. CONCLUSION: Young adult cystinosis patients treated with cysteamine have no additional risk for CVD when compared to patients with chronic kidney disease of other causes.


Assuntos
Aterosclerose/complicações , Aterosclerose/diagnóstico , Cistinose/complicações , Técnicas de Diagnóstico Cardiovascular , Adulto , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Pressão Sanguínea , Cistinose/diagnóstico , Cistinose/patologia , Cistinose/fisiopatologia , Feminino , Taxa de Filtração Glomerular , Frequência Cardíaca , Humanos , Masculino , Tamanho do Órgão , Túnica Íntima/patologia , Adulto Jovem
19.
Pediatr Nephrol ; 26(2): 205-15, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20734088

RESUMO

Cystinosis is the major cause of inherited Fanconi syndrome, and should be suspected in young children with failure to thrive and signs of renal proximal tubular damage. The diagnosis can be missed in infants, because not all signs of renal Fanconi syndrome are present during the first months of life. In older patients cystinosis can mimic idiopathic nephrotic syndrome due to focal and segmental glomerulosclerosis. Measuring elevated white blood cell cystine content is the corner stone for the diagnosis. The diagnosis is confirmed by molecular analysis of the cystinosin gene. Corneal cystine crystals are invariably present in all patients with cystinosis after the age of 1 year. Treatment with the cystine depleting drug cysteamine should be initiated as soon as possible and continued lifelong to prolong renal function survival and protect extra-renal organs. This educational feature provides practical tools for the diagnosis and treatment of cystinosis.


Assuntos
Cisteamina/uso terapêutico , Cistinose/diagnóstico , Cistinose/tratamento farmacológico , Diagnóstico Diferencial , Humanos , Rim/patologia
20.
Trends Mol Med ; 27(7): 673-686, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33975805

RESUMO

Nephropathic cystinosis is a severe, monogenic systemic disorder that presents early in life and leads to progressive organ damage, particularly affecting the kidneys. It is caused by mutations in the CTNS gene, which encodes the lysosomal transporter cystinosin, resulting in intralysosomal accumulation of cystine. Recent studies demonstrated that the loss of cystinosin is associated with disrupted autophagy dynamics, accumulation of distorted mitochondria, and increased oxidative stress, leading to abnormal proliferation and dysfunction of kidney cells. We discuss these molecular mechanisms driving nephropathic cystinosis. Further, we consider how unravelling molecular mechanisms supports the identification and development of new strategies for cystinosis by the use of small molecules, biologicals, and genetic rescue of the disease in vitro and in vivo.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinose/terapia , Terapia Genética/métodos , Mutação , Bibliotecas de Moléculas Pequenas/uso terapêutico , Cistinose/genética , Cistinose/patologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA